GRADUATE STUDENT SYMPOSIUM
Transcription
GRADUATE STUDENT SYMPOSIUM
GRADUATE STUDENT SYMPOSIUM The Graduate School of Biomedical Sciences Baylor College of Medicine Thursday, October 23, 2014 THE 26th ANNUAL GRADUATE STUDENT RESEARCH SYMPOSIUM The Graduate School of Biomedical Sciences Baylor College of Medicine Houston, Texas October 23, 2014 Program ....................................................................................... iii Awards Ceremony ...................................................................... iv Welcome ........................................................................................ v Graduate Student Council ......................................................... vi Message from Dr. Klotman ....................................................... vii Message from Dr. Johnson ....................................................... viii The Joseph L. Melnick Lecturer ............................................... ix Awards 2013-14 ............................................................................ x Acknowledgements ................................................................... xiii Cover Legend: “Embryonic stem cell (ESC)-derived neurons on ESC-derived glial cells” Mouse embryonic stem cells were genetically targetd to constitutively express a red fluorescent reporter, then differentiated into neurons (green, stained with the neuronal marker beta III-tubulin) and glial cells (red). Blue marks DAPI-positive nuclei.” Developmental Biology student Isabella Garcia from Dr. Benjamin Arenkiel’s lab. A publication of The Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030 ii Oral Presentations – Cullen Poster Presentations Rayzor Lounge 9:15 – 9:30 9:30 – 9:35 9:35 – 9:40 9:40 – 9:45 9:45 – 10:00 10:00 – 10:15 10:15 – 10:30 10:30 – 10:45 10:45 – 11:00 Breakfast (Coffee & Pastries provided) Introductions Welcome Announce Poster Finalist Sarah Hein Deborah L. Johnson, Ph.D. Gayle Slaughter, Ph.D. Carolyn Adamski Biochemistry & Molecular Biology (pg 8) Shrenik Mehta Pharmacology (pg 9) Corey Hecksel Molecular Virology & Microbiology (pg 10) Redwan Huq Molecular Physiology (pg11) Break (Coffee & Snacks provided) The Joseph L. Melnick Distinguished Guest Speaker 11:00 – 12:00 Mina Bissell, Ph.D. “Why don't we get more cancer? The crucial role of Extracellular Matrix and Microenvironment in metastasis and dormancy.” 12:00 – 1:15 1:15 – 1:30 1:30 – 1:45 1:45 – 2:00 2:00 – 2:15 2:15 – 2:30 2:30 – 2:45 2:45 – 3:00 3:00 – 3:15 3:15 – 3:30 3:30 – 3:45 3:45 – 4:00 4:00 – 5:00 Finalist Poster Session Molecular & Human Genetics (pg 12) Developmental Biology (pg 13) Molecular & Cellular Biology (pg 14) Clinical Scientist Traning Program (pg 15) Structural & Comp. Biology (pg 16) Jessica Sowa Andrew Folick Jonathan Shepherd Maria Monica Gramatges Corey Hryc Break (Coffee & Snacks provided) Integrative Molec & Biomed Sciences (pg 17) Translational Biology & Mol. Med. (pg18) Neuroscience (pg 19) Immunology (pg 20) Lauren Figard Tabassum Majid Daniel Zollinger Xiaoyi Yuan Break (Coffee & Snacks provided) Awards Ceremony & Reception iii Awards Ceremony & Reception Thursday, October 23, 2014 4:00 – 5:00 PM 8-Stranded Beta-Barrel Jelly Roll Awards Professor John J. Trentin Scholarship Awards Deborah K. Martin Achievement Award in Biomedical Sciences Marc Dresden Excellence in Graduate Education Award Milton Gregory Poster Awards Beckman Poster Awards Mavis P. Kelsey Student Speaker Awards Beckman Platform Award Abstract Book can be located at: https://www.bcm.edu/education/schools/graduate-school-of-biomedical-sciences/currentstudents/graduate-student-council/activities-and-opportunities/gsbs-symposium iv Welcome back to the Annual GSBS Graduate Student Symposium! Thank you for joining us to honor the graduate students performing exceptional research here at the Baylor College of Medicine. Every year, students from across the graduate school take a break from their busy scientific schedules to showcase their work. These poster and speaker presentations promote the free sharing of ideas and foster communication and collaboration between different lab groups. Furthermore, each department has competitively selected an outstanding student for a platform presentation. In addition to the exceptional student work, we are particularly excited to have Dr. Mina Bissell, a distinguished scientist and senior advisor at the Lawrence Berkeley National Laboratory, here to deliver the Joseph L. Melnick Distinguished Lecture. We owe a special thanks to the students and faculty who have made today possible. The members of the Graduate Student Council have spent many hours during the planning and execution of the symposium and other student events throughout the year, and for their efforts we are sincerely grateful. Special thanks are needed for Melissa Houghton and Dr. Gayle Slaughter for their critical roles in planning and organizing the symposium today. It would not be possible without their efforts every year. We also thank our poster and presentation judges who volunteered their time today to make this a successful symposium. In addition, we would like to extend our sincere gratitude to Dr. Deborah Johnson for her mentorship of the GSC and the student body this year. We would like to thank the faculty and staff members here at BCM who work tirelessly to ensure that our students receive the best educational experience possible. We would especially like to recognize Dr. Gad Shaulsky, Dr. Carolyn Smith, Dr. Rick Sifers, Emily Legler, Codysue Congdon and all of our faculty mentors and program administrators for their continued dedication. And of course, we extend our thanks to Dr. Mina Bissell for joining us here today to deliver the Distinguished Lecture. It has been our privilege and pleasure to serve you over the last year and we hope that you enjoy today’s Symposium. Sarah Hein, GSC President Timothy Dosey, GSC Vice President v GRADUATE STUDENT COUNCIL MEMBERS Sarah Hein, President Tim Dosey, Vice-President Jessica Moore Jessica Scott Jordan Kho Amulya Sreekumar Medicine Ninad Oak Jonathan Gallion Jenny Sun Biochemistry & Molecular Biology Jessica Lunsford Integrative Molec & Biomed Sciences Shrenik Mehta Developmental Biology Cameron Landers Molecular and Cellular Biology Amanda Koire Molecular and Human Genetics Marissa Scavuzzo Structural & Computational Biology Chinh Nguyen Molecular Virology & Microbiology vi Neuroscience Immunology Pharmacology Translational Biol. & Molec. MD/PhD 1st year Representative Each year the Graduate Student Symposium of the Graduate School of Biomedical Sciences at Baylor College of Medicine takes us into a new era of research as the School’s trainees present the results of their work in the laboratory. Their work demonstrates two of the College’s important missions – research and education. While each student presents the work he or she has done in the laboratory, that work could not be accomplished without the input of their mentors, the scientists whose presence makes Baylor College of Medicine one of the top research institutions in the United States and, indeed, the world. The work of Baylor’s graduate students is an important part of the output of each basic and translational laboratory in the College. Such work not only advances the mission of Baylor, it also provides promising new information that can be translated into better diagnosis and treatment for some of the most difficult diseases afflicting mankind around the globe. In my tenure as President of Baylor College of Medicine, I have found that my interactions with graduate students are among the most challenging and interesting. Our graduate students hail from Texas and a variety of states and nations around the world, yet they work well together in the College’s laboratories, setting up the possibility of collaborations for decades to come. This is the kind of work and cooperation that results in answers to challenges in disease and biology. Fostering such work and training tomorrow’s scientists is the reason that the Baylor Graduate School of Biomedical Sciences exists. Each year, the School’s leadership and I are honored to take part in this important event that displays our students’ work. On behalf of the Baylor College of Medicine faculty, I congratulate the students, their faculty mentors and the leadership of the Graduate School for creating an outstanding Graduate Student Symposium for 2014. Their continued success is important to the future of Baylor College of Medicine, the Graduate School of Biomedical Sciences and our global community. Paul Klotman, M.D. President & CEO Executive Dean Baylor College of Medicine vii The Graduate Student Research Symposium is a wonderful platform that highlights the enormous breadth of research contributions made by our graduate students at Baylor College of Medicine. It commemorates the hard work and diligence of our students that represent the next generation of scientific leaders. The day fosters stimulating discussions that enhance our students’ ability to articulate their work and presents opportunities for networking and new collaborations. I would like to thank the many individuals that made this Symposium possible. I would like to specifically thank our judges for critiquing the platform presentations and posters, Melissa Houghton for assembling the abstract book, and the Graduate Student Council who planned the scientific program. I am delighted to be part of this special event and hope you will take the time to participate and celebrate the tremendous efforts and productivity of our students and their mentors. Deborah Johnson, Ph.D. Dean, The Graduate School of Biomedical Sciences The William R. Brinkley BRASS Chair Professor, Molecular & Cellular Biology Baylor College of Medicine viii THE JOSEPH L. MELNICK DISTINGUISHED GUEST LECTURER Mina J. Bissell, Ph.D. Faculty Member at University of California, Berkeley, Distinguished Scientist and Senior Advisor at Lawrence Berkeley National Laboratory Dr. Bissell is a visionary and pioneer in the area of the role of extracellular matrix (ECM) and microenvironment in regulation of tissue-specific gene expression with special emphasis in breast cancer, where she has changed a number of established paradigms. She earned an A.B. with honors in chemistry from Harvard College and a Ph.D. in bacterial genetics from Harvard University. She joined the Lawrence Berkeley National Laboratory in 1972, became Director of Cell & Molecular Biology in 1988, and was appointed Director of all of Life Sciences in 1992. Upon stepping down as the Life Sciences Division Director, she was named Distinguished Scientist. She was also the OBER/DOE Distinguished Scientist Fellow in Life Sciences. Dr. Bissell has authored more than 380 publications, is a member of nine international scientific boards, and is on the editorial board of a dozen scientific journals. She has given more than 120 ‘named and distinguished’ lectures and has been both a Fogarty (1983) and Guggenheim (1992) Fellow. Her awards include the E.O. Lawrence Award and Medal (1996), the Mellon Award from the University of Pittsburgh (1998), the Clowes/Eli Lilly Award from AACR (1999), the first “Innovator Award” of the US DOD for breast cancer research (2002), the Brinker Award from Komen Foundation (2003), the Discovery Health Channel Medical Honor and Medal (2004), the H. Lee Moffitt Cancer Center Ted Couch Lectureship and Award (2007), the Pezcoller Foundation–AACR International Award for Cancer Research (2007), the 2007 Inserm/France Foreign Scientist of the Year Award , the 2007 FASEB Excellence in Science Award, the 2008 American Cancer Society's Medal of Honor for Basic Research, a 2009 Rothschild-Mayent Fellowship award by Institut Curie, the 2010 Alexander Bodini Foundation Prize for Scientific Excellence in Medicine from American-Italian Cancer Foundation's The. In 2011 Breast Cancer Research Foundation's Jill Rose Award; the 2011 Susan Bulkeley Butler Leadership Excellence Award, and the 2012 AACR Distinguished Lectureship in Breast Cancer Research as well as the Lifetime Achievement Award, Lawrence Berkeley National Laboratory, and the California State Assembly STEM Woman of the Year (2014) Award. In 2008, University of Porto, Portugal created the Mina J. Bissell Award to be given every 2 years to a person who has changed a field. Dr. Bissell has been elected a Fellow of the AAAS, the Institute of Medicine of the National Academies, the American Academy of Arts and Sciences, the American Philosophical Society, The Royal Society of Chemistry, the National Academy of Sciences, and in 2013 was inducted into the inaugural class of Fellows in the AACR Academy. She has served as President of the American Society of Cell Biology and the International Society of Differentiation. She has received honorary doctorates from Pierre & Marie Curie University in Paris and the University of Copenhagen. The Graduate Student Research Symposium’s Joseph L. Melnick Guest Lectureship is made possible through a generous endowment from the late Joseph L. Melnick, Ph.D., the first Dean of the Graduate School of Baylor College of Medicine. Dr. Melnick served in that capacity from 1968-1991. ix 2013 SYMPOSIUM AWARD WINNERS BECKMAN 1st PLACE PLATFORM SPEAKER WINNER: Ryan Ash, Neuroscience ALUMNI 2ND AND 3RD PLACE PLATFORM SPEAKER AWARDS: Abhisek Bhattacharya, Immunology (2nd place) Viktor Feketa, Molecular Physiology (3rd place) BECKMAN POSTER WINNER: Fatih Semerci, Developmental Biology POSTER PRESENTATION WINNERS: 1st Place William Choi – Developmental Biology Laura Heckman – Molecular & Human Genetics Fatih Semerci– Developmental Biology Liuliu Zheng– Biochemistry and Molecular Biology 2nd Place Shawn Badal – Translational Biology & Molecular Medicine Abhisek Bhattacharya – Immunology Antentor Hinton– Integrative Molecular & Biomedical Sciences Tongchao Li– Developmental Biology rd 3 Place Alexander Herman – Developmental Biology Teng-Wei Huang– Developmental Biology Jennifer Johnson – Neuroscience Stephanie Kyle – Molecular & Human Genetics Honorable Mention Stefanie Alexander – Integrative Molecular & Biomedical Sciences Viktor Feketa – Molecular Physiology/Cardio. Sciences Jeffrey Howard – Translational Biology & Molecular Medicine Amy Ku– Translational Biology & Molecular Medicine Aditya Kulkarni – Biochemistry and Molecular Biology Antonina Kurtova – Integrative Molecular & Biomedical Sciences x SPECIAL AWARDS • 8-STRANDED BETA-BARREL JELLY ROLL AWARD—Established in 1998 these awards are given by the first year graduate students at the end of their first year. The awards are in recognition of outstanding educational achievement in the GSBS Service Curriculum. • PROFESSOR JOHN J. TRENTIN SCHOLARSHIP AWARDS—Established by Dr. John Trentin, former Professor and Head of the Division of Experimental Biology in the Department of Surgery at Baylor from 1960-1992. These awards are given yearly to the graduate students with the top GPA’s upon completion of their first year in graduate school. • DEBORAH K. MARTIN ACHIEVEMENT AWARD IN BIOMEDICAL SCIENCES—Established in 1996, this award is supported by the Deborah K. Martin fund in memory of the late Debbie Martin, former administrator of the Cell and Molecular Biology Program who died of breast cancer. This award is given to a graduate student in recognition of his/her achievements in Graduate School. • MARC DRESDEN EXCELLENCE IN GRADUATE EDUCATION AWARD—in memory of the late Marc Dresden, Ph.D. Dr. Dresden joined the Baylor faculty in 1968 and was appointed Assistant Dean of The Graduate School in 1979. Dr. Dresden was Associate Dean from 1986 until his death in 1990. This award is supported by the Marc Dresden Excellence in Graduate Education Fund. The award recipient is selected each year by the Graduate Student Council from nominations submitted by graduate students. The faculty recipient is chosen for his/her outstanding contributions to graduate education as a distinguished teacher and mentor. xi 2013 AWARD WINNERS MARC DRESDEN EXCELLENCE IN GRADUATE EDUCATION AWARD: John Wilson, Ph.D., Biochemistry & Molecular Biology DEBORAH K. SCIENCES: MARTIN ACHIEVEMENT AWARD IN BIOMEDICAL Kristin Karlin, Integrative Molecular & Biomedical Sciences PROFESSOR JOHN J. TRENTIN SCHOALRSHIP AWARDS: Ryan Gomoto -- Structural & Computational Biology & Molecular Medicine Hsin-I Jen -- Developmental Biology Kathleen Seger -- Integrative Molecular & Biomedical Sciences Heidi Martini-Stoica -- Translational Biology & Molecular Medicine and MD/PhD program Nicholas Neill – Molecular and Human Genetics Victor Onuchic -- Structural & Computational Biology & Molecular Medicine Martin Powers – Developmental Biology Jenny Sun – Neuroscience Edgar Walker – Neuroscience Xing Wei -- Integrative Molecular & Biomedical Sciences Quan Zhou -- Structural & Computational Biology & Molecular Medicine 8-STRANDED BETA-BARREL JELLY ROLL AWARDS: BEST OVERALL COURSE Development BEST TEACHING Dr. Andy Groves, Neuroscience Dr. Joanna Jankowsky, Neuroscience Dr. Steen Pedersen, Molecular Physiology BEST TEACHING ASSISTANT Abhisek Bhattacharya, Immunology xii SYMPOSIUM FUNDING ACKNOWLEDGMENTS We are grateful to the following for their continued support of the Symposium: • JOSEPH L. MELNICK, Ph.D. whose endowment supports The Joseph L. Melnick Distinguished Guest Lecturer and the Symposium Awards Reception. Dr. Melnick was the first Dean of the Graduate School of Baylor College of Medicine and served in that capacity from 1968 to 1991. • HARRIS BUSCH GRADUATE STUDENT SYMPOSIUM FUND—Dr. Harris Busch was the Michael E. DeBakey Professor of the Department of Pharmacology. Dr. Busch was the Chairman of the Department of Pharmacology from July 1, 1960 to June 30, 1998. • MAVIS P. KELSEY AWARDS—The Mavis P. Kelsey fund supports outstanding research by graduate students. This endowment will provide a monetary award for each graduate student who is selected to speak for his or her program. • MILTON GREGORY AWARDS—The Milton Gregory endowment will support the twelve poster awards at the annual symposium. The funds are in honor of outstanding research by graduate students. • BECKMAN BEST POSTER AND BEST PLATFORM TALK AWARDS – Sponsored by Beckman Instruments, Inc. Beckman Instruments of Fullerton, California, operates on a worldwide basis distributing products used in laboratories for biological analysis in all phases of the battle against disease, from pioneering medical research through drug discovery to clinical diagnostics. • GSBS ALUMNI STUDENT SCHOLARSHIP ENDOWMENT -- This endowment was established in 2013 by the BCM Alumni Affairs and will support the student speaker awards. xiii BAYLOR COLLEGE OF MEDICINE Table of Contents Acevedo-Rodriguez, Alexandra ..................................................................................... 1 Department of Neuroscience Advisor: Shailaja Mani, Ph.D. - Department of Molecular & Cellular Biology Benjamin Arenkiel, Ph.D.-Department of Molecular & Human Genetics Adamski, Carolyn Joy ..................................................................................................... 2 Department of Biochemistry & Molecular Biology Advisor: Timothy Palzkill, Ph.D. - Department of Pharmacology Ahmed, Saeed ................................................................................................................ 3 Clinical Scientist Training Program Advisor: Thomas Giordano, M.D. - Department of Medicine Elizabeth Chiao, M.D./M.P.H.-Department of Medicine Akinfenwa, Patricia Y..................................................................................................... 4 Program in Translational Biology & Molecular Medicine Advisor: Richard Hurwitz, M.D. - Department of Pediatrics Donald Parsons, M.D./Ph.D.-Department of Pediatrics Alexander, Stefanie ........................................................................................................ 5 Integrative Program in Molecular and Biomedical Sciences Advisor: Brendan Lee, M.D./Ph.D. - Department of Molecular & Human Genetics Allen, Hunter Marshall ................................................................................................... 6 Department of Neuroscience Advisor: Hui-Chen Lu, Ph.D. - Department of Pediatrics Amin, Samirkumar B ...................................................................................................... 7 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Lynda Chin, M.D. - Genomic Medicine Amin, Viren R ................................................................................................................. 8 Department of Molecular & Human Genetics Advisor: Aleksandar Milosavljevic, Ph.D. - Department of Molecular & Human Genetics Araya, Mussie K ............................................................................................................. 9 Department of Molecular Physiology & Biophysics Advisor: William Brownell, Ph.D. - Department of Otolaryngology-Head and Neck Surgery Arnold, James Michael ................................................................................................. 10 Department of Biochemistry & Molecular Biology Advisor: Arun Sreekumar, Ph.D. - Department of Molecular & Cellular Biology Ash, Ryan Thomas ........................................................................................................ 11 Department of Neuroscience Advisor: Stelios Smirnakis, M.D./Ph.D. - Department of Neurology Atkins, Lisa Michelle .................................................................................................... 12 Department of Molecular Virology & Microbiology Advisor: Joseph Petrosino, Ph.D. - Department of Molecular Virology & Microbiology Atri, Benu ..................................................................................................................... 13 xvi 3 2014 GRADUATE STUDENT SYMPOSIUM Program in Structural and Computational Biology an Molecular Biophysics Advisor: Olivier Lichtarge, M.D./Ph.D. - Department of Molecular & Human Genetics Bachman, Benjamin Judson .......................................................................................... 14 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Olivier Lichtarge, M.D./Ph.D. - Department of Molecular & Human Genetics Badal, Shawn Samson ................................................................................................... 15 Program in Translational Biology & Molecular Medicine Advisor: Farhad Danesh, M.D. - Department of Medicine William Mitch, M.D.-Department of Medicine Bader, David A ............................................................................................................. 16 Department of Molecular & Cellular Biology Advisor: Sean Mcguire, M.D./Ph.D. - Department of Molecular & Cellular Biology Baete, Dillon Patrick ..................................................................................................... 17 Department of Neuroscience Advisor: Mauro Costa-Mattioli, Ph.D. - Department of Neuroscience Bajaj, Lakshya ............................................................................................................. 18 Department of Molecular & Human Genetics Advisor: Marco Sardiello, Ph.D. - Department of Molecular & Human Genetics Balderas, Miriam ......................................................................................................... 19 Department of Molecular Virology & Microbiology Advisor: Anthony Maresso, Ph.D. - Department of Molecular Virology & Microbiology Barrasso, Anthony Patrick ............................................................................................ 20 Integrative Program in Molecular and Biomedical Sciences Advisor: Loning Fu, Ph.D. - Department of Pediatrics Barry, Meagan Amelia .................................................................................................. 21 Program in Translational Biology & Molecular Medicine Advisor: Peter Hotez, M.D./Ph.D. - Department of Pediatrics Laila Woc-Colburn, M.D.-Department of Medicine Benton, Susan Michelle ................................................................................................ 22 Integrative Program in Molecular and Biomedical Sciences Advisor: Jason Heaney, Ph.D. - Department of Molecular & Human Genetics Bertolet, Grant Daniel ................................................................................................... 23 Department of Pathology & Immunology Advisor: Dongfang Liu, M.D./Ph.D. - Department of Pediatrics Birol, Onur ................................................................................................................... 24 Program in Developmental Biology Advisor: Andrew Groves, Ph.D. - Department of Neuroscience Bondar, Vitaliy V .......................................................................................................... 25 Department of Molecular & Human Genetics Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Breaux, Meghan ........................................................................................................... 26 Integrative Program in Molecular and Biomedical Sciences xvii BAYLOR COLLEGE OF MEDICINE Advisor: Nikolaj Timchenko, Ph.D. - Department of Pathology & Immunology Frederick Pereira, Ph.D.-Department of Molecular & Cellular Biology Brewer III, Alex Jawann ............................................................................................... 27 Department of Pharmacology Advisor: Richard De La Garza, Ph.D. - Department of Psychiatry & Behavioral Sciences Brinegar, Amy Elizabeth .............................................................................................. 28 Department of Molecular & Cellular Biology Advisor: Thomas Cooper, M.D. - Department of Pathology & Immunology Brown, Rogers, II Milton .............................................................................................. 29 Program in Developmental Biology Advisor: Andrew Groves, Ph.D. - Department of Neuroscience Burton, Jason Christopher............................................................................................. 30 Integrative Program in Molecular and Biomedical Sciences Advisor: Irina Larina, Ph.D. - Department of Molecular Physiology & Biophysics Byrd, Tiara T................................................................................................................. 31 Program in Translational Biology & Molecular Medicine Advisor: Nabil Ahmed, M.D. - Department of Pediatrics Robert Grossman, M.D.-Department of Neurosurgery Cadwell, Cathryn Rene ................................................................................................. 32 Department of Neuroscience Advisor: Andreas Tolias, Ph.D. - Department of Neuroscience Cai, Mingbo ................................................................................................................. 33 Department of Neuroscience Advisor: David Eagleman, Ph.D. - Department of Neuroscience Campbell, Ian Morgan .................................................................................................. 34 Department of Molecular & Human Genetics Advisor: James Lupski, M.D./Ph.D. - Department of Molecular & Human Genetics Pawel Stankiewicz, M.D./Ph.D.-Department of Molecular & Human Genetics Campbell, James Christopher ....................................................................................... 35 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Choel Kim, Ph.D. - Department of Pharmacology Cao, Zhijuan ................................................................................................................. 36 Program in Cardiovascular Sciences Advisor: Sean Marrelli, Ph.D. - Department of Anesthesiology Carter, Angela N ........................................................................................................... 37 Department of Neuroscience Advisor: Anne Anderson, M.D. - Department of Pediatrics Chaboub, Lesley S.M.................................................................................................... 38 Program in Developmental Biology Advisor: Benjamin Deneen, Ph.D. - Department of Neuroscience Chang, Chi-Hsuan ........................................................................................................ 39 Integrative Program in Molecular and Biomedical Sciences xviii 3 2014 GRADUATE STUDENT SYMPOSIUM Advisor: Jeffrey Rosen, Ph.D. - Department of Molecular & Cellular Biology Chapple, Rich Harrison................................................................................................. 40 Department of Molecular & Human Genetics Advisor: Daisuke Nakada, B.A.Sc. - Department of Molecular & Human Genetics Chen, Bo ...................................................................................................................... 41 Department of Biochemistry & Molecular Biology Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology Chen, Chun-An ............................................................................................................ 42 Department of Molecular & Human Genetics Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Chen, Elaine ................................................................................................................. 43 Department of Molecular & Cellular Biology Advisor: Lawrence Chan, D.Sc. - Department of Medicine Chen, Kuang-Yui Michael ............................................................................................ 44 Department of Biochemistry & Molecular Biology Advisor: Patrick Barth, Ph.D. - Department of Pharmacology Chen, Kuchuan ............................................................................................................. 45 Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Cheng, Jinxuan ............................................................................................................. 46 Department of Biochemistry & Molecular Biology Advisor: Kimberley Tolias, Ph.D. - Department of Neuroscience Chiang, Angie Chi An .................................................................................................. 47 Department of Neuroscience Advisor: Joanna Jankowsky, Ph.D. - Department of Neuroscience Choi, Sungwoo ............................................................................................................. 48 Program in Developmental Biology Advisor: David Moore, Ph.D. - Department of Molecular & Cellular Biology Choi, William Tin-Shing .............................................................................................. 49 Program in Developmental Biology Advisor: Mirjana Maletic-Savatic, M.D./Ph.D. - Department of Pediatrics Chung, Hsiang-Ching .................................................................................................. 50 Integrative Program in Molecular and Biomedical Sciences Advisor: Thomas Westbrook, Ph.D. - Department of Biochemistry & Molecular Biology Ciupek, Andrew Mark Robinson .................................................................................. 51 Program in Translational Biology & Molecular Medicine Advisor: Suzanne Fuqua, Ph.D. - Department of Medicine Mothaffar Rimawi, M.D.-Department of Medicine Ciupek, Sarah Margaret ................................................................................................ 52 Department of Neuroscience Advisor: Daoyun Ji, Ph.D. - Department of Molecular & Cellular Biology Collinson-Pautz, Matthew R ......................................................................................... 53 xix BAYLOR COLLEGE OF MEDICINE Program in Translational Biology & Molecular Medicine Advisor: David Spencer, Ph.D. - Department of Pathology & Immunology Kevin Slawin, M.D.-Department of Urology Conley, Zachary Christopher ........................................................................................ 54 Department of Biochemistry & Molecular Biology Advisor: E. Zechiedrich, Ph.D. - Department of Molecular Virology & Microbiology Cullen, Sean Michael .................................................................................................... 55 Program in Developmental Biology Advisor: Margaret Goodell, Ph.D. - Department of Pediatrics Dai, Hang ..................................................................................................................... 56 Department of Molecular & Human Genetics Advisor: Suzanne Leal, Ph.D. - Department of Molecular & Human Genetics Darrow, Michele C. ...................................................................................................... 57 Department of Biochemistry & Molecular Biology Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology David, Gabriela Riva .................................................................................................... 58 Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Davis, Shaun Michael ................................................................................................... 59 Department of Molecular & Human Genetics Advisor: Herman Dierick, M.D. - Department of Molecular & Human Genetics Dawson, Emily Packard ................................................................................................ 60 Integrative Program in Molecular and Biomedical Sciences Advisor: Jason Heaney, Ph.D. - Department of Molecular & Human Genetics De Maio, Antonia ........................................................................................................ 61 Program in Developmental Biology Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Denfield, George Hilton ............................................................................................... 62 Department of Neuroscience Advisor: Andreas Tolias, Ph.D. - Department of Neuroscience Dharmadhikari, Avinash Vijay ..................................................................................... 63 Program in Translational Biology & Molecular Medicine Advisor: Pawel Stankiewicz, M.D./Ph.D. - Department of Molecular & Human Genetics Ignatia Van Den Veyver, M.D.-Department of Obstetrics & Gynecology Dogruluk, Turgut ......................................................................................................... 64 Department of Molecular & Human Genetics Advisor: Kenneth Scott, Ph.D. - Department of Molecular & Human Genetics Dominguez-Vidana, Rocio .......................................................................................... 65 Integrative Program in Molecular and Biomedical Sciences Advisor: Thomas Westbrook, Ph.D. - Department of Biochemistry & Molecular Biology Dosey, Timothy Lloyd .................................................................................................. 66 Integrative Program in Molecular and Biomedical Sciences xx 3 2014 GRADUATE STUDENT SYMPOSIUM Advisor: Theodore Wensel, Ph.D. - Department of Biochemistry & Molecular Biology Egunsola, Adetutu Taiwo ............................................................................................ 67 Department of Molecular & Human Genetics Advisor: Brendan Lee, M.D./Ph.D. - Department of Molecular & Human Genetics Eisenhofer, Joel David .................................................................................................. 68 Department of Neuroscience Advisor: Richard De La Garza, Ph.D. - Department of Psychiatry & Behavioral Sciences Emerson, Charlene H .................................................................................................... 69 Department of Molecular & Human Genetics Advisor: Alison Bertuch, M.D./Ph.D. - Department of Pediatrics Fachini, Joseph Mario ................................................................................................... 70 Department of Molecular Physiology & Biophysics Advisor: Joel Neilson, Ph.D. - Department of Molecular Physiology & Biophysics Faille, River Uru ........................................................................................................... 71 Department of Pathology & Immunology Advisor: Scott Wenderfer, M.D. - Department of Pediatrics Farinholt, Timothy ....................................................................................................... 72 Department of Biochemistry & Molecular Biology Advisor: Adam Kuspa, Ph.D. - Department of Biochemistry & Molecular Biology Feketa, Viktor .............................................................................................................. 73 Program in Cardiovascular Sciences Advisor: Sean Marrelli, Ph.D. - Department of Anesthesiology Feng, Xiang .................................................................................................................. 74 Department of Pharmacology Advisor: Patrick Barth, Ph.D. - Department of Pharmacology Figard, Lauren Renee .................................................................................................... 75 Integrative Program in Molecular and Biomedical Sciences Advisor: Anna Sokac, Ph.D. - Department of Biochemistry & Molecular Biology Fitch, Olivia Michelle ................................................................................................... 76 Department of Neuroscience Advisor: Michael Friedlander, Ph.D. - Department of Neuroscience Mauro Costa-Mattioli, Ph.D.-Department of Neuroscience Fleet, Tiffany C ............................................................................................................. 77 Program in Translational Biology & Molecular Medicine Advisor: Bert O'Malley, M.D. - Department of Molecular & Cellular Biology Clifford Dacso, M.D./M.P.H.-Department of Molecular & Cellular Biology Foley, Christopher James .............................................................................................. 78 Department of Molecular & Cellular Biology Advisor: Nicholas Mitsiades, M.D./Ph.D. - Department of Medicine Folick, Andrew Kenji ................................................................................................... 79 Program in Developmental Biology Advisor: Meng Wang, Ph.D. - Department of Molecular & Human Genetics xxi BAYLOR COLLEGE OF MEDICINE Fountain, Michael David .............................................................................................. 80 Program in Translational Biology & Molecular Medicine Advisor: Christian Schaaf, M.D./Ph.D. - Department of Molecular & Human Genetics Daryl Scott, M.D./Ph.D.-Department of Molecular & Human Genetics Fousek, Kristen Nicole ................................................................................................. 81 Program in Translational Biology & Molecular Medicine Advisor: Nabil Ahmed, M.D. - Department of Pediatrics Helen Heslop, M.D.-Department of Pediatrics Freire, Pablo Riera ........................................................................................................ 82 Department of Molecular & Cellular Biology Advisor: Orla Conneely, Ph.D. - Department of Molecular & Cellular Biology Froudarakis, Emmanouil .............................................................................................. 83 Department of Neuroscience Advisor: Andreas Tolias, Ph.D. - Department of Neuroscience Fuja, Daniel G ............................................................................................................... 84 Integrative Program in Molecular and Biomedical Sciences Advisor: Jason Yustein, M.D./Ph.D. - Department of Pediatrics Fujiwara, Kenichiro ..................................................................................................... 85 Integrative Program in Molecular and Biomedical Sciences Advisor: Lawrence Donehower, Ph.D. - Department of Molecular Virology & Microbiology Fullerton, Paul Thomas ................................................................................................. 86 Department of Molecular & Human Genetics Advisor: Martin Matzuk, M.D./Ph.D. - Department of Pathology & Immunology Fultz, Robert Steven ..................................................................................................... 87 Integrative Program in Molecular and Biomedical Sciences Advisor: James Versalovic, M.D./Ph.D. - Department of Pathology & Immunology Gala, Upasana .............................................................................................................. 88 Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Galaz-Montoya, Monica Laura ..................................................................................... 89 Department of Biochemistry & Molecular Biology Advisor: Theodore Wensel, Ph.D. - Department of Biochemistry & Molecular Biology Gao, Chunxu ................................................................................................................ 90 Department of Molecular Virology & Microbiology Advisor: James Versalovic, M.D./Ph.D. - Department of Pathology & Immunology Garcia, Courtney Dawn ................................................................................................ 91 Department of Neuroscience Advisor: Dora Angelaki, Ph.D. - Department of Neuroscience Garcia, Melissa Nolan................................................................................................... 92 Clinical Scientist Training Program Advisor: Kristy Murray, Ph.D. - Epidemiology Garno, Sandy Lynn ....................................................................................................... 93 xxii 3 2014 GRADUATE STUDENT SYMPOSIUM Integrative Program in Molecular and Biomedical Sciences Advisor: Weiwei Dang, Ph.D. - Department of Molecular & Human Genetics Gates, Leah Ashley ....................................................................................................... 94 Department of Molecular & Cellular Biology Advisor: Bert O'Malley, M.D. - Department of Molecular & Cellular Biology Gelowani, Violet .......................................................................................................... 95 Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D. - Department of Molecular & Human Genetics Ghosh-Choudhury, Triparna ........................................................................................ 96 Program in Translational Biology & Molecular Medicine Advisor: Matthew Anderson, M.D./Ph.D. - Department of Obstetrics & Gynecology Creighton Edwards, M.D.-Department of Obstetrics & Gynecology Girard, Mary Elizabeth ................................................................................................. 97 Department of Molecular Virology & Microbiology Advisor: Christophe Herman, Ph.D. - Department of Molecular & Human Genetics Godoy, Guilherme ........................................................................................................ 98 Clinical Scientist Training Program Advisor: Seth Lerner, M.D. - Department of Urology Gokulakrishnan, Ganga ............................................................................................... 99 Clinical Scientist Training Program Advisor: Steven Abrams, M.D. - Department of Pediatrics Gramatges, Maria Monica .......................................................................................... 100 Clinical Scientist Training Program Advisor: Alison Bertuch, M.D./Ph.D. - Department of Pediatrics Graves, Joshua Daniel................................................................................................. 101 Integrative Program in Molecular and Biomedical Sciences Advisor: Weei-Chin Lin, M.D./Ph.D. - Department of Medicine Griffin, Deric Maurice ................................................................................................ 102 Program in Translational Biology & Molecular Medicine Advisor: Robia Pautler, Ph.D. - Department of Molecular Physiology & Biophysics Stephanie Abrams, M.D.-Department of Pediatrics Grzeskowiak, Caitlin ................................................................................................. 103 Department of Molecular & Human Genetics Advisor: Kenneth Scott, Ph.D. - Department of Molecular & Human Genetics Haelterman, Nele ....................................................................................................... 104 Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Haines, Katherine ....................................................................................................... 105 Department of Molecular & Human Genetics Advisor: Sharon Plon, M.D./Ph.D. - Department of Pediatrics Haller, Meade ............................................................................................................. 106 Department of Molecular & Cellular Biology xxiii BAYLOR COLLEGE OF MEDICINE Advisor: Dolores Lamb, Ph.D. - Department of Urology Hamilton, Mark Patrick .............................................................................................. 107 Department of Molecular & Cellular Biology Advisor: Sean Mcguire, M.D./Ph.D. - Department of Molecular & Cellular Biology Harrigal, Lindsay Jeanette .......................................................................................... 108 Integrative Program in Molecular and Biomedical Sciences Advisor: Janet Butel, Ph.D. - Department of Molecular Virology & Microbiology Hause, Anne M ........................................................................................................... 109 Program in Translational Biology & Molecular Medicine Advisor: Pedro Piedra, M.D. - Department of Molecular Virology & Microbiology Robert Atmar, M.D.-Department of Medicine He, Zongxiao .............................................................................................................. 110 Department of Molecular & Human Genetics Advisor: Suzanne Leal, Ph.D. - Department of Molecular & Human Genetics Hecksel, Corey W ....................................................................................................... 111 Department of Molecular Virology & Microbiology Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology Hein, Sarah Marie ....................................................................................................... 112 Department of Molecular & Cellular Biology Advisor: Yi Li, Ph.D. - Department of Molecular & Cellular Biology Hendrix, Amanda Y. ................................................................................................... 113 Program in Translational Biology & Molecular Medicine Advisor: Farrah Kheradmand, M.D. - Department of Medicine Anita Sabichi, M.D.-Department of Medicine Herman, Alexander Michael ....................................................................................... 114 Program in Developmental Biology Advisor: Benjamin Arenkiel, Ph.D. - Department of Molecular & Human Genetics Herrera, Jose A. .......................................................................................................... 115 Program in Translational Biology & Molecular Medicine Advisor: Jeffrey Neul, M.D./Ph.D. - Department of Pediatrics Xander Wehrens, M.D./Ph.D.-Department of Molecular Physiology & Biophysics Hilton, Tyler John ....................................................................................................... 116 Integrative Program in Molecular and Biomedical Sciences Advisor: Theodore Wensel, Ph.D. - Department of Biochemistry & Molecular Biology John Wilson, Ph.D.-Department of Biochemistry & Molecular Biology Hinton, Antentor Othrell ............................................................................................ 117 Integrative Program in Molecular and Biomedical Sciences Advisor: Yong Xu, Ph.D. - Department of Pediatrics Ho, Hsing-I ................................................................................................................ 118 Department of Molecular & Human Genetics Advisor: Gad Shaulsky, Ph.D. - Department of Molecular & Human Genetics Ho, Szu-Yu ................................................................................................................ 119 xxiv 3 2014 GRADUATE STUDENT SYMPOSIUM Program in Developmental Biology Advisor: Matthew Rasband, Ph.D. - Department of Neuroscience Holdman, Xue B ......................................................................................................... 120 Integrative Program in Molecular and Biomedical Sciences Advisor: Jeffrey Rosen, Ph.D. - Department of Molecular & Cellular Biology Holt, Matthew Valle ................................................................................................... 121 Department of Biochemistry & Molecular Biology Advisor: Jun Qin, Ph.D. - Department of Biochemistry & Molecular Biology Hong, Chuan .............................................................................................................. 122 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology Hooker, Stanley Eugene ............................................................................................. 123 Department of Molecular & Human Genetics Advisor: Suzanne Leal, Ph.D. - Department of Molecular & Human Genetics Hornstein, Benjamin Daniel ....................................................................................... 124 Department of Molecular Virology & Microbiology Advisor: E. Zechiedrich, Ph.D. - Department of Molecular Virology & Microbiology Hou, Jason K ............................................................................................................... 125 Clinical Scientist Training Program Advisor: Hashem El-Serag, M.D./M.P.H. - Department of Medicine Hryc, Corey F.............................................................................................................. 126 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology Hsu, Hsiang-Ting ....................................................................................................... 127 Department of Pathology & Immunology Advisor: Jordan Orange, M.D. - Department of Pediatrics Hsu, Teng-Kuei .......................................................................................................... 128 Department of Biochemistry & Molecular Biology Advisor: Olivier Lichtarge, M.D./Ph.D. - Department of Molecular & Human Genetics Hsu, Tiffany ............................................................................................................... 129 Integrative Program in Molecular and Biomedical Sciences Advisor: Thomas Westbrook, Ph.D. - Department of Biochemistry & Molecular Biology Huang, Gilbert ........................................................................................................... 130 Department of Biochemistry & Molecular Biology Advisor: Choel Kim, Ph.D. - Department of Pharmacology Huang, Longwen ........................................................................................................ 131 Department of Neuroscience Advisor: Benjamin Arenkiel, Ph.D. - Department of Molecular & Human Genetics Huang, Teng-Wei ....................................................................................................... 132 Program in Developmental Biology Advisor: Jeffrey Neul, M.D./Ph.D. - Department of Pediatrics Huang, Yu-Mei .......................................................................................................... 133 xxv BAYLOR COLLEGE OF MEDICINE Department of Neuroscience Advisor: Matthew Rasband, Ph.D. - Department of Neuroscience Huq, Redwan .............................................................................................................. 134 Department of Molecular Physiology & Biophysics Advisor: Christine Beeton, Ph.D. - Department of Molecular Physiology & Biophysics Hurwitz, Amy Marie ................................................................................................... 135 Program in Translational Biology & Molecular Medicine Advisor: Timothy Palzkill, Ph.D. - Department of Pharmacology Robert Atmar, M.D.-Department of Medicine Hutchinson, Diane Smith ............................................................................................ 136 Program in Translational Biology & Molecular Medicine Advisor: Joseph Petrosino, Ph.D. - Department of Molecular Virology & Microbiology Stephen Pflugfelder, M.D.-Department of Ophthalmology James, Regis Aaron ..................................................................................................... 137 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Chad Shaw, Ph.D. - Department of Molecular & Human Genetics Jarrett, Kelsey Elizabeth ............................................................................................. 138 Integrative Program in Molecular and Biomedical Sciences Advisor: William Lagor, Ph.D. - Department of Molecular Physiology & Biophysics Jen, Hsin-I .................................................................................................................. 139 Program in Developmental Biology Advisor: Andrew Groves, Ph.D. - Department of Neuroscience Jiang, Xiqian .............................................................................................................. 140 Department of Pharmacology Advisor: Jin Wang, Ph.D. - Department of Pharmacology Johnson, Jennifer Leigh .............................................................................................. 141 Department of Neuroscience Advisor: Mauro Costa-Mattioli, Ph.D. - Department of Neuroscience Johnston, Alyssa N ..................................................................................................... 142 Program in Translational Biology & Molecular Medicine Advisor: Yi Li, Ph.D. - Department of Molecular & Cellular Biology Kaelber, Jason T ......................................................................................................... 143 Department of Molecular Virology & Microbiology Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology Karandur, Deepti ........................................................................................................ 144 Program in Structural and Computational Biology an Molecular Biophysics Advisor: B. Pettitt, Ph.D. - Biochemistry Jianpeng Ma, Ph.D.-Department of Biochemistry & Molecular Biology Kaushik, Akash Kumar ............................................................................................... 145 Department of Biochemistry & Molecular Biology Advisor: Arun Sreekumar, Ph.D. - Department of Molecular & Cellular Biology Kee, Sara Elizabeth ..................................................................................................... 146 xxvi 3 2014 GRADUATE STUDENT SYMPOSIUM Department of Neuroscience Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Kelly, Aaron Josef ...................................................................................................... 147 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Tsz-Kwong Man, Ph.D. - Department of Pediatrics Kettner, Nicole M ....................................................................................................... 148 Department of Molecular & Cellular Biology Advisor: Loning Fu, Ph.D. - Department of Pediatrics Kho, Jordan ................................................................................................................ 149 Program in Developmental Biology Advisor: Brendan Lee, M.D./Ph.D. - Department of Molecular & Human Genetics Kim, Cynthia J ............................................................................................................ 150 Department of Molecular & Human Genetics Advisor: Benjamin Arenkiel, Ph.D. - Department of Molecular & Human Genetics Kim, Ik Sun ............................................................................................................... 151 Integrative Program in Molecular and Biomedical Sciences Advisor: Xiang Zhang, Ph.D. - Department of Molecular & Cellular Biology Kim, Maria Hyoun ...................................................................................................... 152 Clinical Scientist Training Program Advisor: Elizabeth Chiao, M.D./M.P.H. - Department of Medicine Knezevic, Jana ........................................................................................................... 153 Department of Molecular & Cellular Biology Advisor: Jeffrey Rosen, Ph.D. - Department of Molecular & Cellular Biology Kong, Kathleen .......................................................................................................... 154 Department of Molecular Virology & Microbiology Advisor: Ronald Javier, Ph.D. - Department of Molecular Virology & Microbiology Kruse, Robert Layne ................................................................................................... 155 Program in Translational Biology & Molecular Medicine Advisor: Karl-Dimiter Bissig, M.D./Ph.D. - Department of Molecular & Cellular Biology Fasiha Kanwal, M.D.-Department of Medicine Ku, Amy Tsu ............................................................................................................... 156 Program in Translational Biology & Molecular Medicine Advisor: Hoang Nguyen, Ph.D. - Department of Molecular & Cellular Biology Abdul Diwan, Ph.D.-Department of Pathology & Immunology Kurbanov, Suhrab ...................................................................................................... 157 Department of Pathology & Immunology Advisor: Leonid Metelitsa, M.D./Ph.D. - Department of Pediatrics Kurtova, Antonina V................................................................................................... 158 Program in Translational Biology & Molecular Medicine Advisor: Keith Chan, Ph.D. - Department of Urology Seth Lerner, M.D.-Department of Urology Kyle, Stephanie Marie ................................................................................................ 159 xxvii BAYLOR COLLEGE OF MEDICINE Department of Molecular & Human Genetics Advisor: Monica Justice, Ph.D. - Department of Molecular & Human Genetics David Nelson, Ph.D.-Department of Molecular & Human Genetics Laitman, Andrew Marc ............................................................................................... 160 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Mirjana Maletic-Savatic, M.D./Ph.D. - Department of Pediatrics Lakshminarasimhan, Janakiraman Kausik ................................................................. 161 Department of Neuroscience Advisor: Dora Angelaki, Ph.D. - Department of Neuroscience Lam, Sharon ............................................................................................................... 162 Department of Pathology & Immunology Advisor: Catherine Bollard, M.B.,B.Ch. - Department of Pediatrics Cliona Rooney, Ph.D.-Department of Pediatrics Laug, Dylan James ...................................................................................................... 163 Program in Developmental Biology Advisor: Benjamin Deneen, Ph.D. - Department of Neuroscience Leach, John Preston .................................................................................................... 164 Department of Molecular Physiology & Biophysics Advisor: James Martin, M.D./Ph.D. - Department of Molecular Physiology & Biophysics Lee, Yi-Chien ............................................................................................................. 165 Integrative Program in Molecular and Biomedical Sciences Advisor: Brendan Lee, M.D./Ph.D. - Department of Molecular & Human Genetics Lee, Yu-Ju .................................................................................................................. 166 Program in Translational Biology & Molecular Medicine Advisor: Weei-Chin Lin, M.D./Ph.D. - Department of Medicine Mothaffar Rimawi, M.D.-Department of Medicine Lemon, Laramie Denise .............................................................................................. 167 Integrative Program in Molecular and Biomedical Sciences Advisor: Alison Bertuch, M.D./Ph.D. - Department of Pediatrics Lesteberg, Kelsey Elizabeth ....................................................................................... 168 Department of Pathology & Immunology Advisor: George Makedonas, Ph.D. - Department of Pediatrics Lewis, Kyle Joseph Edward........................................................................................ 169 Department of Molecular & Cellular Biology Advisor: Nikolaj Timchenko, Ph.D. - Department of Pathology & Immunology Richard Sifers, Ph.D.-Department of Pathology & Immunology Lewis, Phoebe Elnora ................................................................................................. 170 Program in Translational Biology & Molecular Medicine Advisor: Qizhi Yao, M.D./Ph.D. - Department of Surgery Shital Patel, M.D.-Department of Medicine Li, Cheng-Lin ............................................................................................................. 171 Department of Molecular & Human Genetics xxviii 3 2014 GRADUATE STUDENT SYMPOSIUM Advisor: Gad Shaulsky, Ph.D. - Department of Molecular & Human Genetics Li, Lele ....................................................................................................................... 172 Program in Cardiovascular Sciences Advisor: James Martin, M.D./Ph.D. - Department of Molecular Physiology & Biophysics Li, Tongchao .............................................................................................................. 173 Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Andrew Groves, Ph.D.-Department of Neuroscience Li, Zao ........................................................................................................................ 174 Department of Biochemistry & Molecular Biology Advisor: Zheng Zhou, Ph.D. - Department of Biochemistry & Molecular Biology Lian, Hong ................................................................................................................. 175 Department of Molecular & Human Genetics Advisor: Hui Zheng, Ph.D. - Department of Molecular & Human Genetics Liang, Dan .................................................................................................................. 176 Department of Pathology & Immunology Advisor: William Decker, Ph.D. - Department of Pathology & Immunology Lien, Steven Donald ................................................................................................... 177 Department of Neuroscience Advisor: J. Dickman, Ph.D. - Department of Neuroscience Lin, Angelique ........................................................................................................... 178 Integrative Program in Molecular and Biomedical Sciences Advisor: Daisuke Nakada, B.A.Sc. - Department of Molecular & Human Genetics Lin, Chih-Chun .......................................................................................................... 179 Department of Molecular & Human Genetics Advisor: Meng Wang, Ph.D. - Department of Molecular & Human Genetics Lingappan, Krithika ................................................................................................... 180 Clinical Scientist Training Program Advisor: Bhagavatula Moorthy, Ph.D. - Department of Pediatrics Litvinchuk, Alexandra ............................................................................................... 181 Integrative Program in Molecular and Biomedical Sciences Advisor: Hui Zheng, Ph.D. - Department of Molecular & Human Genetics Liu, Kuanqing ............................................................................................................ 182 Department of Molecular Virology & Microbiology Advisor: Jue Wang, Ph.D. - Department of Molecular & Human Genetics Anthony Maresso, Ph.D.-Department of Molecular Virology & Microbiology Liu, Lucy .................................................................................................................... 183 Department of Neuroscience Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Lo, Flora ..................................................................................................................... 184 Integrative Program in Molecular and Biomedical Sciences Advisor: Xiang Zhang, Ph.D. - Department of Molecular & Cellular Biology xxix BAYLOR COLLEGE OF MEDICINE Lo, Yuan-Hung .......................................................................................................... 185 Integrative Program in Molecular and Biomedical Sciences Advisor: Noah Shroyer, Ph.D. - Department of Pediatrics Loehr, James Anthony ................................................................................................ 186 Department of Molecular Physiology & Biophysics Advisor: George Rodney, Ph.D. - Department of Molecular Physiology & Biophysics Lu, Hengyu ................................................................................................................ 187 Integrative Program in Molecular and Biomedical Sciences Advisor: Kenneth Scott, Ph.D. - Department of Molecular & Human Genetics Lu, Hsiang-Chih ......................................................................................................... 188 Program in Developmental Biology Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Lu, Wen ...................................................................................................................... 189 Department of Pathology & Immunology Advisor: David Corry, M.D. - Department of Medicine Lu, Yang .................................................................................................................... 190 Integrative Program in Molecular and Biomedical Sciences Advisor: Jeffrey Rosen, Ph.D. - Department of Molecular & Cellular Biology Lumaban, Jeannette Galande ...................................................................................... 191 Department of Molecular & Human Genetics Advisor: David Nelson, Ph.D. - Department of Molecular & Human Genetics Lunsford, Jessica Leigh .............................................................................................. 192 Department of Pathology & Immunology Advisor: Jonathan Levitt, Ph.D. - Department of Pathology & Immunology Ma, Boxue .................................................................................................................. 193 Department of Biochemistry & Molecular Biology Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology Madan, Simran ........................................................................................................... 194 Program in Translational Biology & Molecular Medicine Advisor: Brendan Lee, M.D./Ph.D. - Department of Molecular & Human Genetics Brett Graham, M.D./Ph.D.-Department of Molecular & Human Genetics Magnan, David Russell ............................................................................................... 195 Integrative Program in Molecular and Biomedical Sciences Advisor: David Bates, Ph.D. - Department of Molecular & Human Genetics Mahadevan, Sangeetha Kaveri ................................................................................... 196 Program in Translational Biology & Molecular Medicine Advisor: Ignatia Van Den Veyver, M.D. - Department of Obstetrics & Gynecology Carlos Bacino, M.D.-Department of Molecular & Human Genetics Mahanic, Christina Susan ........................................................................................... 197 Integrative Program in Molecular and Biomedical Sciences Advisor: Weei-Chin Lin, M.D./Ph.D. - Department of Medicine Majid, Tabassum ........................................................................................................ 198 xxx 3 2014 GRADUATE STUDENT SYMPOSIUM Program in Translational Biology & Molecular Medicine Advisor: Robia Pautler, Ph.D. - Department of Molecular Physiology & Biophysics Rachelle Doody, M.D./Ph.D.-Department of Neurology Mak, Keng Hou .......................................................................................................... 199 Integrative Program in Molecular and Biomedical Sciences Advisor: Jue Wang, Ph.D. - Department of Molecular & Human Genetics Meng Wang, Ph.D.-Department of Molecular & Human Genetics Manning, Kathleen Seger ........................................................................................... 200 Integrative Program in Molecular and Biomedical Sciences Advisor: Thomas Cooper, M.D. - Department of Pathology & Immunology Mao, Dongxue ............................................................................................................ 201 Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Marin, Miguel ............................................................................................................ 202 Department of Neuroscience Advisor: Matthew Rasband, Ph.D. - Department of Neuroscience Martinez, Jarrod Don .................................................................................................. 203 Department of Molecular & Cellular Biology Advisor: Jianming Xu, Ph.D. - Department of Molecular & Cellular Biology Martini-Stoica, Heidi ................................................................................................. 204 Program in Translational Biology & Molecular Medicine Advisor: Hui Zheng, Ph.D. - Department of Molecular & Human Genetics Joshua Shulman, M.D./Ph.D.-Department of Neurology Masand, Ruchi ........................................................................................................... 205 Department of Molecular & Human Genetics Advisor: Brett Graham, M.D./Ph.D. - Department of Molecular & Human Genetics Mata, Melinda ............................................................................................................ 206 Department of Pathology & Immunology Advisor: Stephen Gottschalk, M.D. - Department of Pediatrics Mayle, Allison ........................................................................................................... 207 Department of Molecular & Human Genetics Advisor: Margaret Goodell, Ph.D. - Department of Pediatrics Mayle, Ryan ............................................................................................................... 208 Department of Molecular & Human Genetics Advisor: Grzegorz Ira, Ph.D. - Department of Molecular & Human Genetics Maynard, Janielle P..................................................................................................... 209 Program in Translational Biology & Molecular Medicine Advisor: Sundararajah Thevananther, Ph.D. - Department of Pediatrics John Goss, M.D.-Department of Surgery McCue, Tyler Jordan .................................................................................................. 210 Integrative Program in Molecular and Biomedical Sciences Advisor: Joseph Petrosino, Ph.D. - Department of Molecular Virology & Microbiology xxxi BAYLOR COLLEGE OF MEDICINE Mehta, Pooja Chetan ................................................................................................... 211 Department of Pathology & Immunology Advisor: C Smith, M.D. - Department of Pediatrics Mehta, Shrenik Chetan ............................................................................................... 212 Department of Pharmacology Advisor: Timothy Palzkill, Ph.D. - Department of Pharmacology Meng, Xiangling ........................................................................................................ 213 Department of Neuroscience Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Minnick, Phillip Jay .................................................................................................... 214 Department of Biochemistry & Molecular Biology Advisor: Susan Rosenberg, Ph.D. - Department of Molecular & Human Genetics Minor, Marissa Mie Kehaulani ................................................................................... 215 Department of Molecular Virology & Microbiology Advisor: Betty Slagle, Ph.D. - Department of Molecular Virology & Microbiology Mitchell, Patrick Gerald .............................................................................................. 216 Integrative Program in Molecular and Biomedical Sciences Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology Mitra, Sayantan .......................................................................................................... 217 Department of Biochemistry & Molecular Biology Advisor: B Prasad, Ph.D. - Department of Biochemistry & Molecular Biology Monkkonen, Teresa ................................................................................................... 218 Department of Molecular & Cellular Biology Advisor: Michael Lewis, Ph.D. - Department of Molecular & Cellular Biology Monroe, Tanner Oliver ............................................................................................... 219 Department of Molecular Physiology & Biophysics Advisor: James Martin, M.D./Ph.D. - Department of Molecular Physiology & Biophysics George Rodney, Ph.D.-Department of Molecular Physiology & Biophysics Moore, Jessica M ........................................................................................................ 220 Department of Biochemistry & Molecular Biology Advisor: Susan Rosenberg, Ph.D. - Department of Molecular & Human Genetics Philip Hastings, Ph.D.-Department of Molecular & Human Genetics Morra, Christina Narie ................................................................................................ 221 Integrative Program in Molecular and Biomedical Sciences Advisor: James Versalovic, M.D./Ph.D. - Department of Pathology & Immunology Murray, Stephen Christopher ...................................................................................... 222 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Steven Ludtke, Ph.D. - Department of Biochemistry & Molecular Biology Murry, Jaclyn Nicole .................................................................................................. 223 Department of Molecular & Human Genetics Advisor: Ignatia Van Den Veyver, M.D. - Department of Obstetrics & Gynecology Muscarella, Aaron M .................................................................................................. 224 xxxii 3 2014 GRADUATE STUDENT SYMPOSIUM Integrative Program in Molecular and Biomedical Sciences Advisor: Xiang Zhang, Ph.D. - Department of Molecular & Cellular Biology Myers, Christopher Gary ............................................................................................ 225 Program in Structural and Computational Biology an Molecular Biophysics Advisor: B. Pettitt, Ph.D. - Biochemistry Nair, Amritha ............................................................................................................. 226 Department of Molecular & Human Genetics Advisor: Thomas Westbrook, Ph.D. - Department of Biochemistry & Molecular Biology Namwanje, Maria ....................................................................................................... 227 Department of Molecular & Human Genetics Advisor: Chester Brown, M.D./Ph.D. - Department of Molecular & Human Genetics Nash, Andrea Kathleen ............................................................................................... 228 Department of Molecular Virology & Microbiology Advisor: Joseph Petrosino, Ph.D. - Department of Molecular Virology & Microbiology Neill, Nicholas Jay ...................................................................................................... 229 Department of Molecular & Human Genetics Advisor: Thomas Westbrook, Ph.D. - Department of Biochemistry & Molecular Biology Nguyen, Chinh Thi Quynh .......................................................................................... 230 Department of Molecular Virology & Microbiology Advisor: Anthony Maresso, Ph.D. - Department of Molecular Virology & Microbiology Nguyen, Lena H .......................................................................................................... 231 Department of Neuroscience Advisor: Anne Anderson, M.D. - Department of Pediatrics Nikolai, Bryan C ......................................................................................................... 232 Department of Molecular & Cellular Biology Advisor: Bert O'Malley, M.D. - Department of Molecular & Cellular Biology Novikov, Ilya ............................................................................................................. 233 Department of Biochemistry & Molecular Biology Advisor: Olivier Lichtarge, M.D./Ph.D. - Department of Molecular & Human Genetics O’Neil, Derek Steven .................................................................................................. 234 Program in Translational Biology & Molecular Medicine Advisor: Kjersti Aagaard, M.D./Ph.D. - Department of Obstetrics & Gynecology William Gibbons, M.D.-Department of Obstetrics & Gynecology Oak, Ninad Ramesh .................................................................................................... 235 Department of Molecular & Human Genetics Advisor: Sharon Plon, M.D./Ph.D. - Department of Pediatrics Oakes, Joshua Michael ............................................................................................... 236 Program in Cardiovascular Sciences Advisor: Susan Hamilton, Ph.D. - Department of Molecular Physiology & Biophysics Onuchic, Vitor ........................................................................................................... 237 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Aleksandar Milosavljevic, Ph.D. - Department of Molecular & Human Genetics xxxiii BAYLOR COLLEGE OF MEDICINE Oyola, Mario G ........................................................................................................... 238 Department of Neuroscience Advisor: Shailaja Mani, Ph.D. - Department of Molecular & Cellular Biology Mariella De Biasi, Ph.D.-Department of Neuroscience Ozseker, Ayse-Sena ................................................................................................... 239 Program in Developmental Biology Advisor: Meng Wang, Ph.D. - Department of Molecular & Human Genetics Pankowicz, Francis .................................................................................................... 240 Department of Molecular & Cellular Biology Advisor: Karl-Dimiter Bissig, M.D./Ph.D. - Department of Molecular & Cellular Biology Park, Jiyoung ............................................................................................................. 241 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Stelios Smirnakis, M.D./Ph.D. - Department of Neurology Patel, Maha Praful....................................................................................................... 242 Program in Translational Biology & Molecular Medicine Advisor: Timothy Palzkill, Ph.D. - Department of Pharmacology Robert Atmar, M.D.-Department of Medicine Pejerrey, Sasha McKai ................................................................................................ 243 Department of Molecular & Cellular Biology Advisor: Suzanne Fuqua, Ph.D. - Department of Medicine Pew, Braden Kyle ....................................................................................................... 244 Department of Molecular & Cellular Biology Advisor: Kjersti Aagaard, M.D./Ph.D. - Department of Obstetrics & Gynecology Dennis Bier, M.D.-Department of Pediatrics Piazza, Victor George ................................................................................................. 245 Program in Cardiovascular Sciences Advisor: Mary Dickinson, Ph.D. - Department of Molecular Physiology & Biophysics Pitcher, Meagan Rochelle ........................................................................................... 246 Program in Translational Biology & Molecular Medicine Advisor: Jeffrey Neul, M.D./Ph.D. - Department of Pediatrics Daniel Glaze, M.D.-Department of Pediatrics Pohodich, Amy E ........................................................................................................ 247 Department of Neuroscience Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Polleys, Erica Jean ...................................................................................................... 248 Integrative Program in Molecular and Biomedical Sciences Advisor: Alison Bertuch, M.D./Ph.D. - Department of Pediatrics Pradhan, Geetali ......................................................................................................... 249 Program in Translational Biology & Molecular Medicine Advisor: Yuxiang Sun, M.D./Ph.D. - Department of Pediatrics Susan Samson, M.D.-Department of Medicine Premkumar, Muralidhar Hebbur ................................................................................. 250 xxxiv 3 2014 GRADUATE STUDENT SYMPOSIUM Clinical Scientist Training Program Advisor: Brendan Lee, M.D./Ph.D. - Department of Molecular & Human Genetics Qin, Liying ................................................................................................................. 251 Department of Biochemistry & Molecular Biology Advisor: Choel Kim, Ph.D. - Department of Pharmacology Quick, Ann Pepper ...................................................................................................... 252 Program in Cardiovascular Sciences Advisor: Xander Wehrens, M.D./Ph.D. - Department of Molecular Physiology & Biophysics Quiros, Joel Patrick ..................................................................................................... 253 Integrative Program in Molecular and Biomedical Sciences Advisor: Olivier Lichtarge, M.D./Ph.D. - Department of Molecular & Human Genetics Raghavan, Adithya ..................................................................................................... 254 Department of Molecular & Human Genetics Advisor: William Craigen, M.D./Ph.D. - Department of Molecular & Human Genetics Rajasekharan, Vivek .................................................................................................. 255 Department of Molecular & Cellular Biology Advisor: Frederick Pereira, Ph.D. - Department of Molecular & Cellular Biology Ramachandran, Prasanna ........................................................................................... 256 Department of Molecular & Human Genetics Advisor: Meng Wang, Ph.D. - Department of Molecular & Human Genetics Regenbogen, Sam Julian ............................................................................................. 257 Department of Pharmacology Advisor: Olivier Lichtarge, M.D./Ph.D. - Department of Molecular & Human Genetics Renwick, Alexander ................................................................................................... 258 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Chad Shaw, Ph.D. - Department of Molecular & Human Genetics Reyes, Natalie Michelle .............................................................................................. 259 Integrative Program in Molecular and Biomedical Sciences Advisor: Suzanne Fuqua, Ph.D. - Department of Medicine Rhee, Christopher J ..................................................................................................... 260 Clinical Scientist Training Program Advisor: Kenneth Brady, M.D. - Department of Anesthesiology Ridgeway, Alex David ................................................................................................ 261 Department of Molecular & Cellular Biology Advisor: Dolores Lamb, Ph.D. - Department of Urology Roberts, Justin Michael............................................................................................... 262 Department of Molecular & Cellular Biology Advisor: Nancy Weigel, Ph.D. - Department of Molecular & Cellular Biology Rodriguez, Amanda ................................................................................................... 263 Department of Molecular & Cellular Biology Advisor: Stephanie Pangas, Ph.D. - Department of Pathology & Immunology Rodriguez, Perla Janet ................................................................................................ 264 xxxv BAYLOR COLLEGE OF MEDICINE Integrative Program in Molecular and Biomedical Sciences Advisor: Henry Pownall, Ph.D. - Department of Medicine Roh, Soung Hun ......................................................................................................... 265 Department of Biochemistry & Molecular Biology Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology David Tweardy, M.D.-Department of Medicine Roman-Sanchez, Ramon ............................................................................................ 266 Integrative Program in Molecular and Biomedical Sciences Advisor: John Wilson, Ph.D. - Department of Biochemistry & Molecular Biology Rosewell Shaw, Amanda Nicole................................................................................. 267 Department of Molecular & Human Genetics Advisor: Philip Ng, Ph.D. - Department of Molecular & Human Genetics Ross, Matthew Clayton ............................................................................................... 268 Department of Molecular Virology & Microbiology Advisor: Joseph Petrosino, Ph.D. - Department of Molecular Virology & Microbiology Rubin, Michelle Jenny ................................................................................................ 269 Integrative Program in Molecular and Biomedical Sciences Advisor: Gad Shaulsky, Ph.D. - Department of Molecular & Human Genetics Runquist, Ingrid Sophie .............................................................................................. 270 Department of Molecular & Cellular Biology Advisor: Michael Lewis, Ph.D. - Department of Molecular & Cellular Biology Rutledge, Eric Benjamin ............................................................................................. 271 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Zhandong Liu, Ph.D. - Department of Pediatrics Sabharwal, Jasdeep Singh ........................................................................................... 272 Department of Neuroscience Advisor: Samuel Wu, Ph.D. - Department of Ophthalmology Safdar, Zeenat ............................................................................................................ 273 Clinical Scientist Training Program Advisor: Mark Entman, M.D. - Department of Medicine Saliba, Jason ............................................................................................................... 274 Department of Molecular & Human Genetics Advisor: Sharon Plon, M.D./Ph.D. - Department of Pediatrics Salvo, Jason Scott ....................................................................................................... 275 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Rui Chen, Ph.D. - Department of Molecular & Human Genetics San Martin, Rebeca .................................................................................................... 276 Department of Molecular & Cellular Biology Advisor: David Rowley, Ph.D. - Department of Molecular & Cellular Biology Santhanam, Balaji ...................................................................................................... 277 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Gad Shaulsky, Ph.D. - Department of Molecular & Human Genetics xxxvi 3 2014 GRADUATE STUDENT SYMPOSIUM Satterfield, Laura Louise............................................................................................. 278 Integrative Program in Molecular and Biomedical Sciences Advisor: Jason Yustein, M.D./Ph.D. - Department of Pediatrics Scott, Jessica Diane..................................................................................................... 279 Integrative Program in Molecular and Biomedical Sciences Advisor: David Moore, Ph.D. - Department of Molecular & Cellular Biology Seilheimer, Robert Lionel ........................................................................................... 280 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Samuel Wu, Ph.D. - Department of Ophthalmology Semerci, Fatih ............................................................................................................ 281 Program in Developmental Biology Advisor: Mirjana Maletic-Savatic, M.D./Ph.D. - Department of Pediatrics Senturk, Mumine ........................................................................................................ 282 Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Seymour, Michelle L. ................................................................................................. 283 Department of Molecular & Cellular Biology Advisor: Frederick Pereira, Ph.D. - Department of Molecular & Cellular Biology Shafi, Ayesha ............................................................................................................. 284 Department of Molecular & Cellular Biology Advisor: Nancy Weigel, Ph.D. - Department of Molecular & Cellular Biology Shen, Shan .................................................................................................................. 285 Department of Neuroscience Advisor: Andreas Tolias, Ph.D. - Department of Neuroscience Shen, Ye ..................................................................................................................... 286 Integrative Program in Molecular and Biomedical Sciences Advisor: H Lacorazza, Ph.D. - Department of Pathology & Immunology Shepherd, Jonathan .................................................................................................... 287 Department of Molecular & Cellular Biology Advisor: Powel Brown, M.D./Ph.D. - Department of Molecular & Cellular Biology Shin, Hongsup ............................................................................................................ 288 Department of Neuroscience Advisor: Whee Ma, Ph.D. - Department of Neuroscience Michael Beauchamp, Ph.D.-Department of Neuroscience Shivanna, Binoy ......................................................................................................... 289 Clinical Scientist Training Program Advisor: Bhagavatula Moorthy, Ph.D. - Department of Pediatrics Shum, Thomas ........................................................................................................... 290 Program in Translational Biology & Molecular Medicine Advisor: Cliona Rooney, Ph.D. - Department of Pediatrics Stephen Gottschalk, M.D.-Department of Pediatrics Siehr, Meagan ............................................................................................................ 291 xxxvii BAYLOR COLLEGE OF MEDICINE Department of Molecular & Human Genetics Advisor: Jeffrey Noebels, M.D./Ph.D. - Department of Neurology Simon, Lukas ............................................................................................................. 292 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Chad Shaw, Ph.D. - Department of Molecular & Human Genetics Sinha, Papiya ............................................................................................................. 293 Department of Pathology & Immunology Advisor: Jordan Orange, M.D. - Department of Pediatrics Sivaramakrishnan, Priya ............................................................................................ 294 Department of Molecular & Human Genetics Advisor: Christophe Herman, Ph.D. - Department of Molecular & Human Genetics Soens, Zachry Tore ..................................................................................................... 295 Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D. - Department of Molecular & Human Genetics Sowa, Jessica Nichole ................................................................................................. 296 Department of Molecular & Human Genetics Advisor: Meng Wang, Ph.D. - Department of Molecular & Human Genetics Spike, Aaron Jonathon ................................................................................................ 297 Department of Molecular & Cellular Biology Advisor: Jeffrey Rosen, Ph.D. - Department of Molecular & Cellular Biology Sreekumar, Amulya ................................................................................................... 298 Department of Molecular & Cellular Biology Advisor: Jeffrey Rosen, Ph.D. - Department of Molecular & Cellular Biology Stay, Trace L ............................................................................................................... 299 Department of Neuroscience Advisor: Dora Angelaki, Ph.D. - Department of Neuroscience Stinnett, Gary R. ......................................................................................................... 300 Department of Molecular Physiology & Biophysics Advisor: Robia Pautler, Ph.D. - Department of Molecular Physiology & Biophysics Stoica, Loredana Georgiana ........................................................................................ 301 Department of Molecular & Cellular Biology Advisor: Mauro Costa-Mattioli, Ph.D. - Department of Neuroscience Stojanoski, Vlatko ...................................................................................................... 302 Department of Biochemistry & Molecular Biology Advisor: Timothy Palzkill, Ph.D. - Department of Pharmacology Stone, Adrianne Elayne .............................................................................................. 303 Program in Translational Biology & Molecular Medicine Advisor: Brendan Lee, M.D./Ph.D. - Department of Molecular & Human Genetics Carlos Bacino, M.D.-Department of Molecular & Human Genetics Stuhlsatz, Danielle Dee ............................................................................................... 304 Program in Structural and Computational Biology an Molecular Biophysics Advisor: B. Pettitt, Ph.D. - Biochemistry xxxviii 3 2014 GRADUATE STUDENT SYMPOSIUM Su, Qingtai ................................................................................................................. 305 Integrative Program in Molecular and Biomedical Sciences Advisor: Li Xin, Ph.D. - Department of Molecular & Cellular Biology Sukumar, Ann Tabitha ............................................................................................... 306 Department of Molecular & Human Genetics Advisor: Alison Bertuch, M.D./Ph.D. - Department of Pediatrics Sukumaran, Sujita ...................................................................................................... 307 Program in Translational Biology & Molecular Medicine Advisor: Ann Leen, Ph.D. - Department of Pediatrics William Fisher, M.D.-Department of Surgery Sun, Jenny J ................................................................................................................ 308 Department of Neuroscience Advisor: Russell Ray, Ph.D. - Department of Neuroscience Sun, Jiayi Monika ....................................................................................................... 309 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Ching Lau, M.D./Ph.D. - Department of Pediatrics Sun, Yu ...................................................................................................................... 310 Integrative Program in Molecular and Biomedical Sciences Advisor: Weiwei Dang, Ph.D. - Department of Molecular & Human Genetics Sung, Yun-Min .......................................................................................................... 311 Department of Biochemistry & Molecular Biology Advisor: Theodore Wensel, Ph.D. - Department of Biochemistry & Molecular Biology Szwarc, Maria Magdalena .......................................................................................... 312 Department of Molecular & Cellular Biology Advisor: Bert O'Malley, M.D. - Department of Molecular & Cellular Biology Tackett, Bryan Christopher ......................................................................................... 313 Program in Translational Biology & Molecular Medicine Advisor: Sundararajah Thevananther, Ph.D. - Department of Pediatrics Saul Karpen, M.D./Ph.D.-Department of Pediatrics Tajhya, Rajeev Babu ................................................................................................... 314 Department of Molecular Physiology & Biophysics Advisor: Christine Beeton, Ph.D. - Department of Molecular Physiology & Biophysics Tan, Kai Li ................................................................................................................. 315 Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Tanner, Mark R. .......................................................................................................... 316 Program in Translational Biology & Molecular Medicine Advisor: Christine Beeton, Ph.D. - Department of Molecular Physiology & Biophysics David Corry, M.D.-Department of Medicine Taylor, Aaron Michael ................................................................................................ 317 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Ching Lau, M.D./Ph.D. - Department of Pediatrics xxxix BAYLOR COLLEGE OF MEDICINE Tepe, Burak ................................................................................................................ 318 Program in Developmental Biology Advisor: Benjamin Arenkiel, Ph.D. - Department of Molecular & Human Genetics Terrón-Díaz, María Elisa ............................................................................................ 319 Integrative Program in Molecular and Biomedical Sciences Advisor: Olivier Lichtarge, M.D./Ph.D. - Department of Molecular & Human Genetics Terwilliger, Austen Lee .............................................................................................. 320 Integrative Program in Molecular and Biomedical Sciences Advisor: Anthony Maresso, Ph.D. - Department of Molecular Virology & Microbiology Tian, Gengwen ........................................................................................................... 321 Department of Pathology & Immunology Advisor: Leonid Metelitsa, M.D./Ph.D. - Department of Pediatrics Tian, Lin ..................................................................................................................... 322 Department of Biochemistry & Molecular Biology Advisor: Xiang Zhang, Ph.D. - Department of Molecular & Cellular Biology Tran, Baouyen ............................................................................................................ 323 Department of Neuroscience Advisor: Edward Cooper, M.D./Ph.D. - Department of Neurology Tran, Linda Lien ......................................................................................................... 324 Integrative Program in Molecular and Biomedical Sciences Advisor: David Rowley, Ph.D. - Department of Molecular & Cellular Biology Tsai, Chang-Ru .......................................................................................................... 325 Program in Developmental Biology Advisor: Michael Galko, Ph.D. - Biochemistry and Molecular Biology Tsai, Wei-Chih ........................................................................................................... 326 Department of Molecular Virology & Microbiology Advisor: Richard Lloyd, Ph.D. - Department of Molecular Virology & Microbiology Tu, Yen-Kuei ............................................................................................................. 327 Integrative Program in Molecular and Biomedical Sciences Advisor: Kimberley Tolias, Ph.D. - Department of Neuroscience Tung, Hui-Ying .......................................................................................................... 328 Department of Pathology & Immunology Advisor: David Corry, M.D. - Department of Medicine Ugur, Berrak .............................................................................................................. 329 Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M. - Department of Molecular & Human Genetics Ung, Kevin ................................................................................................................. 330 Program in Developmental Biology Advisor: Benjamin Arenkiel, Ph.D. - Department of Molecular & Human Genetics Vasquez, Yasmin1 Margarita ..................................................................................... 331 Department of Molecular & Cellular Biology Advisor: Francesco Demayo, Ph.D. - Department of Molecular & Cellular Biology xl 3 2014 GRADUATE STUDENT SYMPOSIUM Villanueva, Hugo ....................................................................................................... 332 Department of Molecular & Cellular Biology Advisor: Michael Lewis, Ph.D. - Department of Molecular & Cellular Biology Vogt, Megan Brittany ................................................................................................. 333 Integrative Program in Molecular and Biomedical Sciences Advisor: Rebecca Rico, M.P.H. - Department of Molecular Virology & Microbiology Vue, Zer ..................................................................................................................... 334 Program in Developmental Biology Advisor: Richard Behringer, - Molecular Genetics - M.D. Anderson Walker, Edgar Yasuhiro.............................................................................................. 335 Department of Neuroscience Advisor: Andreas Tolias, Ph.D. - Department of Neuroscience Wang, Chih-Chuan .................................................................................................... 336 Integrative Program in Molecular and Biomedical Sciences Advisor: Matthew Rasband, Ph.D. - Department of Neuroscience Wang, Feng ................................................................................................................ 337 Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D. - Department of Molecular & Human Genetics Wang, Gao T. .............................................................................................................. 338 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Suzanne Leal, Ph.D. - Department of Molecular & Human Genetics Wang, Li .................................................................................................................... 339 Department of Molecular & Human Genetics Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Wang, Mengyu ........................................................................................................... 340 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Ido Golding, Ph.D. - Department of Biochemistry & Molecular Biology Wang, Rui .................................................................................................................. 341 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Wah Chiu, Ph.D. - Department of Biochemistry & Molecular Biology Wang, Yue ................................................................................................................. 342 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Gad Shaulsky, Ph.D. - Department of Molecular & Human Genetics Wang, Yumeng .......................................................................................................... 343 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Patrick Barth, Ph.D. - Department of Pharmacology Wei, Xing ................................................................................................................... 344 Integrative Program in Molecular and Biomedical Sciences Advisor: Li Xin, Ph.D. - Department of Molecular & Cellular Biology Wetendorf, Margeaux ................................................................................................ 345 Integrative Program in Molecular and Biomedical Sciences Advisor: Francesco Demayo, Ph.D. - Department of Molecular & Cellular Biology xli BAYLOR COLLEGE OF MEDICINE White, Joshua James ................................................................................................... 346 Department of Neuroscience Advisor: Roy Sillitoe, Ph.D. - Department of Pathology & Immunology Williams, LaTerrica Chemise ..................................................................................... 347 Program in Translational Biology & Molecular Medicine Advisor: Stephen Gottschalk, M.D. - Department of Pediatrics Xiao-Tong Song, Ph.D.-Department of Pathology & Immunology Wu, Chun-Ting .......................................................................................................... 348 Department of Neuroscience Advisor: Daoyun Ji, Ph.D. - Department of Molecular & Cellular Biology Xia, Jun ...................................................................................................................... 349 Integrative Program in Molecular and Biomedical Sciences Advisor: Susan Rosenberg, Ph.D. - Department of Molecular & Human Genetics Xie, Wei ..................................................................................................................... 350 Integrative Program in Molecular and Biomedical Sciences Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Xu, Mingchu .............................................................................................................. 351 Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D. - Department of Molecular & Human Genetics Xu, Xiaowei ............................................................................................................... 352 Department of Biochemistry & Molecular Biology Advisor: Rachel Schiff, Ph.D. - Department of Medicine Xu, Yang .................................................................................................................... 353 Department of Pathology & Immunology Advisor: Gianpietro Dotti, M.D. - Department of Medicine Xue, Zenghui .............................................................................................................. 354 Department of Biochemistry & Molecular Biology Advisor: Anna Sokac, Ph.D. - Department of Biochemistry & Molecular Biology Yang, Eric Yota .......................................................................................................... 355 Clinical Scientist Training Program Advisor: Christie Ballantyne, M.D. - Department of Medicine Yang, Liubin .............................................................................................................. 356 Department of Molecular & Human Genetics Advisor: Margaret Goodell, Ph.D. - Department of Pediatrics Yatsenko, Dimitri ....................................................................................................... 357 Department of Neuroscience Advisor: Andreas Tolias, Ph.D. - Department of Neuroscience Yeh, Szu-Ying ............................................................................................................ 358 Program in Developmental Biology Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Yen, Shuo-Ting .......................................................................................................... 359 Program in Developmental Biology xlii 3 2014 GRADUATE STUDENT SYMPOSIUM Advisor: Richard Behringer, - Molecular Genetics - M.D. Anderson Yetman, Michael Joseph ............................................................................................. 360 Department of Neuroscience Advisor: Joanna Jankowsky, Ph.D. - Department of Neuroscience Yin, Jiani .................................................................................................................... 361 Department of Molecular & Human Genetics Advisor: Huda Zoghbi, M.D. - Department of Pediatrics Yong, Lin-Kin ............................................................................................................ 362 Program in Translational Biology & Molecular Medicine Advisor: Qizhi Yao, M.D./Ph.D. - Department of Surgery William Fisher, M.D.-Department of Surgery Yosef, Nejla ............................................................................................................... 363 Department of Molecular Physiology & Biophysics Advisor: Mary Dickinson, Ph.D. - Department of Molecular Physiology & Biophysics You, Ran .................................................................................................................... 364 Department of Pathology & Immunology Advisor: Farrah Kheradmand, M.D. - Department of Medicine Young, Melvin Chen................................................................................................... 365 Department of Biochemistry & Molecular Biology Advisor: Patrick Barth, Ph.D. - Department of Pharmacology Yuan, Bo .................................................................................................................... 366 Integrative Program in Molecular and Biomedical Sciences Advisor: James Lupski, M.D./Ph.D. - Department of Molecular & Human Genetics Yuan, Xiaoyi .............................................................................................................. 367 Department of Pathology & Immunology Advisor: Farrah Kheradmand, M.D. - Department of Medicine Yuan, Yuan ................................................................................................................ 368 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Han Liang, Ph.D. - Bio informatics and Computational Biology Zalewski, Zachary A ................................................................................................... 369 Department of Molecular & Human Genetics Advisor: David Nelson, Ph.D. - Department of Molecular & Human Genetics Zaneveld, Jacques Eric Timothy ................................................................................. 370 Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D. - Department of Molecular & Human Genetics Zeng, Huan-Chang ..................................................................................................... 371 Program in Developmental Biology Advisor: Brendan Lee, M.D./Ph.D. - Department of Molecular & Human Genetics Zhai, Yijie .................................................................................................................. 372 Department of Molecular & Human Genetics Advisor: John Belmont, M.D./Ph.D. - Department of Molecular & Human Genetics Zhang, Jing ................................................................................................................. 373 xliii BAYLOR COLLEGE OF MEDICINE Department of Biochemistry & Molecular Biology Advisor: Ido Golding, Ph.D. - Department of Biochemistry & Molecular Biology Zhang, Xiaotian ......................................................................................................... 374 Department of Molecular & Human Genetics Advisor: Margaret Goodell, Ph.D. - Department of Pediatrics Zhao, Li ...................................................................................................................... 375 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Rui Chen, Ph.D. - Department of Molecular & Human Genetics Zhao, Mingkun ........................................................................................................... 376 Integrative Program in Molecular and Biomedical Sciences Advisor: Jin Wang, Ph.D. - Department of Pharmacology Zheng, Liuliu ............................................................................................................. 377 Department of Biochemistry & Molecular Biology Advisor: Anna Sokac, Ph.D. - Department of Biochemistry & Molecular Biology Zhou, Quan ................................................................................................................ 378 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Yongtao Guan, Ph.D. - Department of Pediatrics Zhou, Ying ................................................................................................................. 379 Integrative Program in Molecular and Biomedical Sciences Advisor: David Moore, Ph.D. - Department of Molecular & Cellular Biology Karl-Dimiter Bissig, M.D./Ph.D.-Department of Molecular & Cellular Biology Zhu, Liyuan ................................................................................................................ 380 Department of Molecular Virology & Microbiology Advisor: Yi Li, Ph.D. - Department of Molecular & Cellular Biology Zhu, Wenyi ................................................................................................................ 381 Integrative Program in Molecular and Biomedical Sciences Advisor: Benjamin Deneen, Ph.D. - Department of Neuroscience Zhu, Ying ................................................................................................................... 382 Program in Structural and Computational Biology an Molecular Biophysics Advisor: Fabrizio Gabbiani, Ph.D. - Department of Neuroscience Zollinger, Daniel Richard ........................................................................................... 383 Department of Neuroscience Advisor: Matthew Rasband, Ph.D. - Department of Neuroscience xliv 2014 GRADUATE STUDENT SYMPOSIUM DETERMINING THE ROLE OF OXYTOCIN IN ESTROGEN RECEPTOR BETA’S MODULATION OF STRESS Alexandra Acevedo-Rodriguez Department of Neuroscience Advisor: Shailaja Mani, Ph.D.-Department of Molecular & Cellular Biology Benjamin Arenkiel, Ph.D.-Department of Molecular & Human Genetics Anxiety and mood disorders affect 20% of the population and confer large decrements to the patient’s quality of life. These disorders are frequently associated with dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis which is a system activated in response to stressors. The sex steroids can modulate HPA axis activity which may contribute to gender disparities observed in the prevalences of these disorders. In particular, estrogen receptor β (ERβ) activity has been found to confer anxiolytic effects and to reduce HPA axis reactivity. This receptor is expressed in the paraventricular nucleus (PVN) of the hypothalamus which is a key component of the HPA axis. Within the PVN, ERβ is highly expressed in oxytocin neurons, and in vitro studies find that ERβ interacts with the oxytocin promoter to regulate oxytocin transcription. Oxytocin is being researched as a potential therapeutic for numerous psychiatric disorders, and peripheral oxytocin administration has been found to decrease HPA axis reactivity and anxiety related behaviors. With ERβ’s expression on oxytocin neurons and ERβ and oxytocin’s shared anxiolytic behavior, this suggests that ERβ’s anxiolytic effect may be due to modulating oxytocin signaling. To determine if oxytocin modulates ERβ mediated anxiolysis, oxytocin null and wild-type animals will perform a behavioral battery to measure anxiety related behaviors and social aversion. To determine the locus of oxytocin signaling necessary for ERβ mediated anxiolysis, oxytocin receptors will be ablated from the PVN, central amygdala, and the median raphe, regions important in stress circuitry. This will be done by utilizing stereotaxic injection of an adeno-associated viral (AAV) vector associated with Cre-recombinase, which will knockdown oxytocin receptor expression in infected cells of OxtrloxP/loxP animals. In order to determine if ERβ binds to the oxytocin promoter in vivo and if stress alters the level of this interaction, wild-type animals will be injected with ERβ agonist or vehicle and then undergo restraint stress or be sacrificed from their home cage. The PVN will be microdissected out and in vivo chromatin immunoprecipitation will be performed on this tissue to determine if ERβ interacts with the oxytocin promoter. For all experiments, male and female animals will be gonadectomized, to prevent endogenous sex steroid interactions with ERβ, and treated with R-DPN or vehicle. These experiments will illustrate the interaction between ERβ and oxytocin and how this interaction relates to physiological stress and anxiety related behaviors. Contributors: Acevedo-Rodriguez, Alexandra; Oyola, Mario; Malysz, Anna; Kudwa, Andrea; Carbone, David; Handa, Robert; Mani, Shaila 1 BAYLOR COLLEGE OF MEDICINE KEY DETERMINANTS IN THE β-LACTAMASE-MEDIATED MECHANISM OF BACTERIAL RESISTANCE TO CEPHALOSPORIN ANTIBIOTICS Carolyn Joy Adamski Department of Biochemistry & Molecular Biology Advisor: Timothy Palzkill, Ph.D.-Department of Pharmacology A comprehensive understanding of the enzymes mediating resistance to the widely prescribed cephalosporin antibiotics is necessary to address the growing need for new antimicrobial therapies. The CTX-M-14 β-lactamase enzyme, one of the most common sources of resistance to cephalosporin antibiotics, was used in this study to identify residues that are necessary for hydrolysis of cefotaxime, an important cephalosporin drug. The roles of residues Ser237 and Arg276 in the activity of CTX-M14 β-lactamase were investigated using site-directed mutagenesis, enzyme kinetic analysis and X-ray crystallography. Ser237 and Arg276 were mutated to alanine and the catalytic activity of the single and double mutant combinations was evaluated. The kinetic analysis revealed that, together, Ser237 and Arg276 residues are key determinants of cefotaxime hydrolysis by the CTX-M-14 β-lactamase. High-resolution crystal structures of the CTX-M-14 variants alone and in complex with cefotaxime illustrated the roles of Ser237 and Arg276 in cefotaxime hydrolysis. When both residues were mutated to alanine several significant changes were seen in the active site including a compromised hydrogen bonding network, a narrower active site, and a 180° rotation in the orientation of the aminothiazole ring on cefotaxime. These results identify Ser237 and Arg276 as key determinants of cefotaxime activity in the CTX-M-14 βlactamase and elucidate their role in catalysis. Furthermore, these studies will help to inform future drug development studies by highlighting these residues as potential targets for β-lactamase inhibitor design. Contributors: Adamski, Carolyn; Cardenas, Ana Maria; Brown, Nicholas; Horton, Lori; Sankaran, Banumathi; Prasad, B.V.V.; Palzkill, Timothy 2 2014 GRADUATE STUDENT SYMPOSIUM IMPROVED IDENTIFICATION AND ENROLLMENT INTO CARE OF HIV-EXPOSED AND INFECTED INFANTS AND CHILDREN FOLLOWING A COMMUNITY HEALTH WORKER INTERVENTION IN LILONGWE, MALAWI Saeed Ahmed Clinical Scientist Training Program Advisor: Thomas Giordano, M.D.-Department of Medicine Elizabeth Chiao, M.D./M.P.H.-Department of Medicine Background: Early identification and prompt entry into care is critical to reducing morbidity and mortality in children with HIV. This report describes the impact of the Tingathe program, which utilizes dedicated community health workers (CHWs) to improve identification and enrollment into care of HIV-exposed and infected infants and children at three Ministry of Health centers in Lilongwe, Malawi. Methods: Three program phases are described. During the first phase, Mentorship-Only (March 2007 to February 2008), on-site clinical mentorship on pediatric HIV care was provided. In the second phase, Tingathe-Basic (March 2008 to February 2009), CHWs provided HIV testing and counseling to improve case finding of HIVexposed and infected children. In the final phase, Tingathe-PMTCT (March 2009 to February 2011), in addition to Tingathe-Basic activities, CHWs were also assigned to HIV-infected pregnant women to improve mother-infant retention in PMTCT care. We reviewed routinely collected program data from March 2005 to March 2011. Results: During the Mentorship-Only phase, 42 children (38 HIV-infected and 4 HIV-exposed) were active in care. During the three years covering the Tingathe-Basic and Tingathe-PMTCT periods, CHWs conducted 44,388 rapid HIV tests, 7658 (17.3%) in children 18 months to 15 years of age. Of children tested, 351 (4.6%) tested HIVpositive. During the Tingathe-Basic phase, 238 HIV-infected children were enrolled, representing a six-fold increase in rate of enrollment from 3.2 to 19.8 per month. The number of HIV-exposed infants increased from 4 to 118. During the Tingathe-PMTCT phase, 526 HIV-infected children were enrolled over 24 months, at a rate of 21.9 patients per month. 1667 HIV-exposed infants were enrolled. Following the addition of PMTCT activities, there was a greater than seven-fold increase in average number of exposed infants enrolled per month (9.5 to 70 patients per month), resulting in 1667 enrolled with a younger median age at enrollment (5.2 vs 2.5 months; p <.001). Conclusion: A multipronged approach utilizing CHWs to conduct health center and home-based HIV testing, link HIV-infected children into clinical care, and provide adherence support to PMTCT mothers can dramatically improve the identification and enrollment into care of HIV-exposed and infected children. Contributors: Saeed Ahmed, MD*1,2§, Maria H. Kim, MD*1,2, Amanda C. Dave, MPH2, Rachael Sabelli2, Kondwani Kanjelo2, Geoffrey Preidis, MD,PhD1, Thomas P. Giordano, MD3, Elizabeth Chiao3, MD, Mina Hosseinipour, MD4,5, Peter N. Kazembe, MBChB1,2, Frank Chimbwandira, MBChB6, and Elaine J. Abrams, MD7. 3 BAYLOR COLLEGE OF MEDICINE VERSICAN, AN INTRAOCULAR PROTEOGLYCAN, ENHANCES VIRAL TRANSGENE EXPRESSION USING A JAK/STAT DEPENDENT PATHWAY Patricia Y Akinfenwa Program in Translational Biology & Molecular Medicine Advisor: Richard Hurwitz, M.D.-Department of Pediatrics Donald Parsons, M.D./Ph.D.-Department of Pediatrics Understanding host pathways regulating the expression of transgenes delivered by adenoviral and AAV vectors could result in improved gene therapy strategies. Although most therapies testing systemic administration have been unsuccessful, gene therapy protocols have been particularly successful in the eye. Our group previously reported the enhancement of AdV transgene expression (TGE) in the presence of vitreous, the gelatinous material that fills the posterior eye. Vitreous has no effect on the efficiency of vector internalization but results in increased transgene mRNA levels. We found that hyaluronan (HA) contributes to the enhancement of AdV TGE, but another at least one other mediator is involved. The project aim was to determine other vitreous component(s) and the signaling pathway(s) responsible for enhancing TGE. Versican, a hyaluronan-binding proteoglycan highly expressed in vitreous, is proteolyzed into two domains (G1 and G3), both have been shown to have functional effects in cells. We hypothesize that versican mediates signaling that enhances AdV transgene expression. Versican is secreted from ACHN cells, a renal adenocarcinoma cell line. Versicanenriched supernatant (VES) was produced by culturing cells for 7 days and concentrating proteins <300kDa using centrifugal filtration. Recombinant versican G1 and G3 supernatants were isolated in a similar fashion. Enrichment was verified by silver stain and Western blot analysis. Y79 or Weri-Rb retinoblastoma cells were transduced with AdV/CMV-Luc and treated with VES, recombinant G1 or G3 and/or kinase inhibitors and effects on AdV TGE were measured using a luciferase reporter assay. STAT3/5 phosphorylation was measured by flow cytometry. The VES and recombinant G1 and G3 resulted in a 2-3-fold enhancement of luciferase activity, similar to enhancement with vitreous treatment. Vitreous and versican treatment increased pSTAT3 and not pSTAT5. Small-molecule inhibition of JAK1/2 using 1µM of Ruxolitinib and STAT3/5 using 10µM C188-9 eliminated the enhancement of TGE mediated by vitreous or versican, suggesting that enhancement is dependent upon JAK/STAT signaling. Results support our hypothesis that versican mediates the enhancement of AdV TGE. This regulation seems to be dependent upon transcriptional activation through JAK/STAT signaling. In a theoretical model, the G1 or G3 domain of versican binds a cell surface receptor, CD44 or EGFR, respectively. The signal transduces through the JAK/STAT pathway, mediating enhanced TGE. These results are the first to implicate versican as an enhancer of AdV TGE, a finding which could be applied to improve the efficiency of adenoviral-based gene therapy protocols. Contributors: Akinfenwa, Patricia; Bond, Wesley; Hurwitz, Mary; Hurwitz, Richard 4 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF SMALL LEUCINE-RICH PROTEOGLYCANS IN OSTEOGENESIS IMPERFECTA Stefanie Alexander Integrative Program in Molecular and Biomedical Sciences/M.D.-Ph.D. Program Advisor: Brendan Lee, M.D./Ph.D.-Department of Molecular & Human Genetics Osteogenesis imperfecta (OI) is a debilitating genetic osteodysplasia that results in low bone mass, bone deformity, and bone fractures. Most cases of OI are caused by mutations in the structural protein type I collagen (dominant OI) or in protein complexes that post-translationally modify type I collagen (recessive OI); both types of mutations can lead to biochemical overmodification. Type I collagen is important for structural integrity of bone and extracellular matrix (ECM) cell signaling. It extensively interacts with ECM components that regulate bioavailability of signaling molecules. Due to the phenotypic similarities between OI mouse models and TGFβ overexpression models, we hypothesized that altered TGFβ signaling could contribute to OI pathogenesis in both recessive (Crtap-/-) and dominant (G610C OI) OI mouse models. In support of this, we found increased mRNA expression of TGFβ target genes with quantitative PCR and an increased ratio of phosphorylated Smad2 to total Smad2 with western blot in bone samples from both OI mouse models suggesting increased TGFβ signaling. In a bone marrow stromal cell (BMSC) in vitro system, which mimics osteoblast differentiation, we found that osteoblasts from both OI mouse models demonstrated increased TGFβ signaling and increased free TGFβ in the media, suggesting altered TGFβ bioavailability. Treatment of both OI mice with a TGFβ antibody (1D11) significantly rescued the OI bone phenotype. Together, these experiments demonstrate that increased TGFβ signaling contributes to the pathogenesis of OI. Small leucine-rich proteoglycans (SLRPs), ECM components that regulate bioavailability of signaling molecules, are the only ECM components that can bind both type I collagen and TGFβ. I hypothesize that changes in type I collagen in OI disturb binding to the SLRPs, which in turn alters TGFβ availability in the ECM. Surface plasmon resonance (SPR) demonstrated that the SLRP decorin (Dcn) binds Crtap-/- type I collagen 45% less effectively than wildtype type I collagen. Altered binding could modulate TGFβ signaling by preventing proper sequestration to type I collagen. I will use SPR to study binding of other SLRPs to type I collagen from both OI mouse models. My initial data studying the interaction between SLRPs and altered TGFβ signaling using the BMSC in vitro system demonstrates that SLRP mRNA expression increases significantly during osteoblast differentiation in both OI models compared to WT. This increase correlates with the increased expression of TGFβ target genes. Further studies will be done to investigate the relationship between the increased TGFβ signaling, increased SLRP expression, and the binding of SLRPs to the ECM. Ultimately, establishing the molecular mechanisms behind altered ECM signaling in OI will lead to more specific treatments for this disease. Contributors: Alexander, Stefanie; Grafe, Ingo; Lee, Brendan 5 BAYLOR COLLEGE OF MEDICINE LEVELS OF NMNAT2, A MOLECULAR CHAPERONE, LINK TO PATHOLOGY IN ALZHEIMER’S DISEASE Hunter Marshall Allen Department of Neuroscience Advisor: Hui-Chen Lu, Ph.D.-Department of Pediatrics The efficacy of nicotinamide mononucleotide adenylyl transferases (NMNATs) for neuroprotection in a diverse range of neurodegenerative conditions makes them promising therapeutic targets. NMNAT2 is the major NMNAT isoform in mammalian brain and is a key neuronal maintenance factor. Nmnat2 mRNA expression is reduced in many neurodegenerative diseases. Elevating NMNAT2 in mammals, or Drosophila NMNAT (dNMNAT) in flies, reduces Tau burden and ameliorates tauopathy model phenotypes. Molecular chaperones aid in the clearance or refolding of non-native proteins in neurodegenerative diseases. Here we report a novel chaperone function requiring a unique C-terminal ATP site that is independent from NMNAT2’s NAD synthase activity. We found that NMNAT2’s chaperone, but not enzymatic, function is required to ameliorate Ataxin1-82Q- and Tau-mediated proteinopathies. In the brains of Alzheimer’s disease (AD) patients, NMNAT2 is greatly reduced at both the mRNA and protein levels. The residual NMNAT2 in AD brains shifts its solubility and co-localizes with aggregated Tau. Similarly to heat shock proteins (HSP), NMNAT2 complexes with Tau (P301L). Thus, these multiple lines of evidence suggest that NMNAT2 acts as a chaperone to reduce proteotoxic stress and maintain neuronal health. Contributors: Ali, Yousuf O.; Allen, Hunter M.; Hatcher, Asante; Bjorklund, Nicole; Taglialatela, Giulio; Lu, Hui-Chen 6 2014 GRADUATE STUDENT SYMPOSIUM ROLE OF LONG NONCODING RNAs IN EPIGENETIC REMODELING IN TRIPLE WILD TYPE MELANOMA Samirkumar B Amin Program in Structural and Computational Biology and Molecular Biophysics Advisor: Lynda Chin, M.D.-Genomic Medicine The major focus of this study is to uncover emerging functional role of long noncoding RNA (lncRNA) in the development and progression of cancer. Although structural alterations like mutations in BRAF, NRAS, etc. are found to be driver events in most of melanoma cases, the cancer genome atlas (TCGA) melanoma working group have also shown around 15% (n=39) of melanoma samples having none of these driver events. Given likely absence of structural events in these triple wild type (WT) samples, epigenetic remodeling may play a dominant role in driving oncogenic transcriptional program. At the transcription core, such remodeling can take place by differential interaction patterns of TF with other TFs, co-regulators or availability of TF DNA-binding site by actions of chromatin remodelers. However, it is unknown whether these TF-coregulator-DNA interactions at known cancer gene sites are coordinated by an abundance of sequence-specific tethering elements. In this context, our hypothesis underscores both mechanistic and functional role of lncRNAs in cancer. Based on recent research, we hypothesize that abundance of lncRNAs harboring sequencespecific motif or structural motif coordinate TF-DNA interactions at promoter regions of known cancer genes and thus, drive carcinogenesis. Using data from TCGA melanoma project, we have characterized differential expression of several lncrRNAs in triple WT melanoma as compare to BRAF/NRAS/NF1 mutant melanoma subtypes. Further, this subtype specific overexpression of lncRNAs show strong association with normal-like CpG island methylation (CIMP) phenotype, possibly indicating altered methylation at promoter regions secondary to lncrna mediated interactions. We also observe abundance of a few transposable elements from AluYc and LINE-1 families in coding region of these lncRNAs and promoter regions of expressed consensus cancer genes. Further, using ChIP-seq data on histone marks, H3K36me3 and H3K27ac on four melanoma samples, we are integrating epigenetic data with known gene and microrna expression data to understand potential shift in transcription secondary to expressed lncrnas in triple WT subtype. Finally, our approach and resulting methods pipeline will be scalable and extensible in understanding mechanistic and functional role of lncRNA in other cancers. Contributors: Amin, Samir B. ; Akdemir, Kadir ; Wu, Chang-Jiun ; Rai, K ; Chin, L. 7 BAYLOR COLLEGE OF MEDICINE EPIGENOMIC FOOTPRINTS ACROSS 111 REFERENCE EPIGENOMES REVEAL TISSUE-SPECIFIC EPIGENETIC REGULATION OF LINCRNAS Viren R Amin Department of Molecular & Human Genetics Advisor: Aleksandar Milosavljevic, Ph.D.-Department of Molecular & Human Genetics Tissue-specific expression of lincRNAs suggests developmental and cell-type specific functions, yet tissue specificity has been established for only a small fraction of this abundant gene category. By analyzing 111 reference epigenomes from the NIH Roadmap Epigenomics project we identify tissue-specific epigenetic regulation for a total of 3,753 (69% examined) lincRNAs, with 54% active in only one of fourteen cell/tissue clusters and 15% in up to three. The tissue-specific lincRNAs are strongly linked to tissue-specific pathways, supporting their important role in development or celltype specific processes. A larger fraction of lincRNA TSSs shows enhancer-like (H3K4me1) than promoter-like (H3K4me3) tissue-specific activation and a significant fraction shows a combined pattern. Polycomb-regulated lincRNAs reside in bivalent state in embryonic stem cells. The exquisitely tissue-specific epigenetic regulation of lincRNAs and the assignment of a majority of lincRNAs to specific tissue types will inform future mechanistic and genetic studies of this newly discovered class of genes. Contributors: Amin, Viren; Harris, Alan; Onuchic, Vitor; Jackson, Andrew; Charnecki, Tim; Paithankar, Sameer; Subramanian, Sai Lakshmi; Riehle, Kevin; Coarfa, Cristian; Milosavljevic, Aleksandar 8 2014 GRADUATE STUDENT SYMPOSIUM NEUROMECHANICS OF HEARING Mussie K Araya Department of Molecular Physiology & Biophysics Advisor: William Brownell, Ph.D.-Department of Otolaryngology-Head and Neck Surgery Hearing requires precise detection and coding of the acoustic signal by the inner ear and equally precise communication of the information through the auditory brain stem. A membrane based motor in the outer hair cell lateral wall contributes to the transformation of sound to a precise neural code. Structural, molecular and energetic similarities between the outer hair cell and auditory brainstem neurons suggest neuromechanics contributes to hearing. The rapid initiation of action potential by cooperative activation of voltage gated ion channels enhances neuronal temporal processing and increases the upper frequency limit for phase locking. We explore the possibility of electrically evoked mechanical forces mediated by changes in the physical properties of the lipid bilayer in synchronizing the gating of voltage gated ion channels for rapid spike generation, thereby introducing the concept of mechanical force in gating of voltage gated ion channels. The rapid speed of membrane electromechanical signaling can supports a simple and direct mechanism of ion channel cooperativity. Using optical tweezers based force bio-sensor; we find that it takes 2-3 times more pulling force to form membrane tethers from the AIS of hippocampal pyramidal neurons than from the soma indicating the AIS membrane has a stronger membranecytoskeleton adhesion force. Previous experiments in cultured neurons showed that channels in soma (which are not anchored by Ank-G) are removed from the membrane by endocytosis. Our results together with previous experiments suggest that channels are firmly anchored to the actin–spectrin–AnkG-based cytoskeleton at AIS. Since cooperative activation is highly dependent on the density of Na+ channels indicating that the inter-channel coupling is distance dependent, the clustering of Na+ channels at the AIS by the actin-spectrin-AnkG network can set the stage for cooperative gating between ion channels. Currently membrane tethers are being pulled under voltage clamp to a standard length and allowed to relax to an equilibrium force. The electromechanical coefficient will be measured. The effect of manipulating membrane material properties on the coefficient and simultaneously measured membrane currents will be determined. Membrane currents will resolve channel function. A greater electromechanical coefficient in the AIS would be consistent with a lower threshold for ion channel gating and support a contribution of electromechanics to ion channel gating. The manipulations that alter electromechanics also alter ion channel kinetics. Further support for the membrane’s role in ion channel gating would be for the coefficient and channel function to vary concomitantly. Contributors: Mussie K. Araya1 and William E. Brownell1,2 1Molecular Physiology and Biophysics 2Otolaryngology – H&N Surgery Baylor College of Medicine, Houston, Tx, USA 9 BAYLOR COLLEGE OF MEDICINE FOXM1 AS A REGULATOR OF BREAST CANCER LIPID METABOLISM James Michael Arnold Department of Biochemistry & Molecular Biology Advisor: Arun Sreekumar, Ph.D.-Department of Molecular & Cellular Biology Breast cancer is a significant public health concern and there remain unmet challenges in the diagnosis and treatment of triple negative breast cancer (TNBC). Gene expression profiling has revealed that TNBC is composed of a diverse set of disease states, which serve as an imperfect predictor of response to therapy, thus there is a need for a more integrated approach to describe phenotypic subtypes within TNBC. Metabolites are the final products of cell signaling pathways, and the relative levels of metabolites can be considered a direct readout of the current phenotypic state of the cell. In a recently published study, our lab profiled 536 metabolites across 67 matchedtumor-normal pairs of breast tissue, all containing matched Affymetrix-derived gene expression profiles and epidemiological data. Our results suggest altered levels of specific metabolites within a subset of TNBC, predominantly of African American (AA) descent. These include elevated levels of unsaturated fatty acids, lipids, and carnitines. Interestingly, this same group of TNBC tumors is highly enriched for the expression of FOXM1, an oncogenic transcription factor associated with cell cycle progression known to be associated with aggressive breast cancer subtypes. Datamining of public datasets and preliminary siRNA knockdown studies in vitro suggest FOXM1 may regulate key lipid catabolism genes ACSL3 and HADHB, thus playing a critical role in the regulation of tumoral lipid profiles and fatty acid beta-oxidation. Furthermore, knockdown of FOXM1 in multiple TNBC cell lines results in an accumulation of neutral lipids, observable by lipidtox staining, which further supports the concept that FOXM1 plays an important role in directing lipid catabolism. These preliminary results suggest FOXM1 may play a significant role in the regulation and maintenance of lipid metabolism in breast cancer. Contributors: Arnold, James; Konde, Sai Aparna; Gu, Franklin; Rao, Arundhati; Sreekumar, Arun 10 2014 GRADUATE STUDENT SYMPOSIUM STABILIZATION OF DENDRITIC SPINE CLUSTERS AND HYPERACTIVE RASMAPK SIGNALING PREDICT ENHANCED LEARNING IN THE MOUSE MODEL OF MECP2 DUPLICATION SYNDROME Ryan Thomas Ash Department of Neuroscience/M.D.-Ph.D. Program Advisor: Stelios Smirnakis, M.D./Ph.D.-Department of Neurology Methyl-CpG-binding-protein-2 (MECP2) duplication syndrome is a progressive Xlinked disorder of intellectual disability and autism. Interestingly, the mouse model of MECP2 duplication syndrome exhibits enhanced motor and contextual fear learning in addition to stereotyped behaviors and social avoidance. We hypothesized that a bias towards increased synaptic stability could lead to abnormally enhanced memory consolidation, reminiscent of savant-like behaviors occasionally associated with autism. Chronic 2-photon microscopy revealed that more dendritic spines are formed and stabilized during motor learning in apical corticospinal dendritic arbors of MECP2 duplication mice, compared to controls. Learning-associated spines formed clusters whose number predicted enhanced motor performance in mutants. The Ras-MAPK signaling pathway was found to be hyperactive specifically after training in MECP2 duplication mouse motor cortex. Pharmacologic inhibition of MAPK signaling normalized both motor and contextual-fear learning in mutants. We conclude that pathologically stable learning-associated dendritic spine clusters and hyperactive Ras-MAPK signaling drive abnormal learning phenotypes in this form of syndromic autism. Contributors: Ash,Ryan; Buffington, Shelly; Costa-Mattioli, Mauro; Zoghbi, Huda; Smirnakis, Stelios 11 BAYLOR COLLEGE OF MEDICINE A GENOMICS-DRIVEN APPROACH TO UNDERSTANDING MECHANISMS OF ATTENUATION AND VIRULENCE IN FRANCISELLA TULARENSIS Lisa Michelle Atkins Department of Molecular Virology & Microbiology Advisor: Joseph Petrosino, Ph.D.-Department of Molecular Virology & Microbiology F. tularensis (Ft) is a Gram-negative, facultative intracellular coccobacillus and the etiological agent of the zoonotic disease tularemia. Subspecies tularensis (type A) and holarctica (type B) are responsible for 100% of tularemia fatalities in the United States, with type A infections resulting in significantly higher mortality (24%) compared to type B (7%). Ft is classified as a Tier One select agent due to its low infectious dose (<15 CFU), high morbidity, and lack of a licensed vaccine. An attenuated Live Vaccine Strain (LVS) was empirically derived in the 1950s from repeated passage of a type B strain. While it was later shown that LVS is less effective against inhalational type A, it is to date the only vaccine for which formal efficacy data in humans exists. However, LVS remains unlicensed for public use, and the exact mechanism of attenuation is unknown. Comparative genomics revealed 51 syntenic block rearrangements in type A compared to type B genomes, which complicates genetic comparison studies between LVS and type A strains. Therefore, we aligned the LVS genome to virulent type B strains and identified 17 genes that are disrupted in LVS, implicating these genes in LVS attenuation and Ft virulence. These candidate genes are not well described and their respective roles in pathogenesis are poorly understood. We propose that the disruption of a subset of candidate genes in virulent type B will result in attenuation as determined by reduced intracellular replication, increased macrophage activation and decreased virulence in mice. Preliminary studies to establish attenuation assays in LVS show that primed macrophages display increased pro-caspase-1 protein levels compared to uninfected controls and undergo cell death earlier than unprimed macrophages, effectively removing the replicative niche for Ft. In contrast, virulent type B replicates to higher numbers in both human and murine macrophages compared to LVS. Additionally, macrophages infected with virulent type B secrete diminished levels of Th1 cytokines compared to LVS, thus establishing valid thresholds in order to evaluate candidate gene disruptions. Characterizing the mechanism of LVS attenuation may lead to the development of attenuated vaccines, in type A or type B backgrounds, that are genetically defined and more efficacious against type A. Contributors: Atkins, Lisa; Ayvaz, Tulin; Petrosino, Joseph 12 2014 GRADUATE STUDENT SYMPOSIUM FUNCTIONAL EFFECTS OF RESIDUE POSITION AND SIZE OF SUBSTITUTION IN E.COLI RECA Benu Atri Program in Structural and Computational Biology and Molecular Biophysics Advisor: Olivier Lichtarge, M.D./Ph.D.-Department of Molecular & Human Genetics Different substitutions impact the function of a protein in different ways. While some substitutions cause a large effect, others are neutral or harmless. In light of widespread exome sequencing, it is critical to distinguish deleterious substitutions from harmless ones in a clinical setting. We hypothesize that the impact of a substitution on phenotype depends on two factors: the positional importance of the residue and size of substitution. To test this, we chose bacterial RecA protein that plays a central role in homologous recombination and regulates DNA damage repair. RecA is also a key component of the bacterial SOS response where it controls the expression of many other DNA damage repair genes. We quantitatively measured the impact of a mutation on RecA by assaying for DNA damage repair function and recombination function. The results of these assays reveal a correlation between residue position and the size of substitution on the functions of RecA. These experimental results complement prior correlations in retrospective analyses, including the leading performance in independent evaluations of blind predictions. Together these data shed new light on the relationship between genotype and phenotype variations. Contributors: Katsonis, Panagiotis; Adikesavan, K. Anbu; Lichtarge, Olivier 13 BAYLOR COLLEGE OF MEDICINE MACHINE LEARNING FROM SOCIAL NETWORKS APPLED TO PROTEINPROTEIN INTERACTIONS: NETFLIX, FACEBOOK, AND P53 Benjamin Judson Bachman Program in Structural and Computational Biology and Molecular Biophysics Advisor: Olivier Lichtarge, M.D./Ph.D.-Department of Molecular & Human Genetics Popular websites such as Netflix and Facebook enhance their users’ experiences by creating recommendations of items such as “potential friends” and “movies you might like” to their customers. They do so by using various machine learning tools to predict interactions. For example, a common method for predicting social interactions is by looking at the proximity of two users in a network. If they have many mutual friends, it is likely that they themselves are friends, but simply haven’t connected with each other online. With Netflix, a common method for predicting a user’s enjoyment of a movie is to build a model to represent how much each user likes various genres of movies and how well each movie fits into a particular genre using a method known as collaborative filtering. Here, it will be shown that these same concepts can be applied to molecular biology, with a focus on predicting interactions of tumor suppressor p53. Rather than having a social network or a matrix of user-movie ratings, the information used will be large-scale biological data, such as protein-protein interaction information and gene coexpression data. In the case of proximity-based predictions, it will be shown that novel p53 kinases can be found by looking at the set of known p53 kinases and identifying their potential “friends”, nearby kinases in the network. With the Netflix-style method, we build a model of the protein-protein interaction network to predict previously unknown p53 interactions. A meta-algorithm combining these methods is also built to optimize prediction quality. The predictions made by these machine learning tools are then tested experimentally to show their prediction accuracy. In addition to predicting that these interactions exist, it is shown that the model can be used to predict something about the nature of the interaction. With this study, it will be shown that social network models can be applied to biological data to successfully predict novel biology, thus speeding our advancement of knowledge. Contributors: Adikesavan, Anbu K; Wilkins, Angela D; Lisewski, Andreas; Regenbogen, Sam; Pickering, Curtis; Donehower, Lawrence; Lichtarge, Olivier 14 2014 GRADUATE STUDENT SYMPOSIUM MIR-93 LINKS METABOLIC CUES TO CHROMATIN REMODELING IN DIABETIC NEPHROPATHY Shawn Samson Badal Program in Translational Biology & Molecular Medicine Advisor: Farhad Danesh, M.D.-Department of Medicine William Mitch, M.D.-Department of Medicine Despite recent progress in identifying microRNAs (miRNAs) with crucial roles in the development of diabetic nephropathy (DN), the functional validation and downstream effectors of these miRNAs in vivo are frequently lacking. Here, we show that miR-93 plays an important role in the development and progression of DN in vivo. First, a tissue-specific, tamoxifen-inducible model of miR-93 overexpression was generated to selectively induce miR-93 overexpression only in podocytes (Pod-miR-93). At 24 weeks of age, diabetic db/db mice with forced expression of Pod-miR-93 exhibited improved kidney function as measured by a significant reduction in albuminuria, when compared to diabetic controls. Additionally, these mice exhibited a significant reduction in kidney fibrosis compared to controls as measured by Periodic-Acid-Schiff’s (PAS) staining. Importantly, micrographs of kidney cortices prepared for transmission electron microscopy revealed reduced podocyte detachment, and improved glomerular basement membrane thickness in diabetic Pod-miR-93 mice, similar to that of nondiabetic control mice. Second, restoring miR-93 expression via the in vivo delivery of miR-93 mimic in diabetic mice rescued DN phenotype in a similar fashion. RNASequencing and bioinformatics analysis revealed a potential role for miR-93 as an epigenetic regulator through a novel target, Msk2. We observed high glucose induced activation of Msk2 leads to global changes in H3S10 phosphorylation levels, which acts to alter chromatin states. We propose that miR-93 serves as a relaying link between the metabolic state and the chromatin remodeling in the diabetic milieu mainly through its regulatory effect on phosphorylation of histone H3 at serine 10 (H3S10). These findings uncover a novel role of miRNAs as critical metabolic/epigenetic switches, which has implications for targeting of miRNAs in DN. Contributors: Shawn S. Badal, Yin Wang, Jianyin Long, Antony Rodriguez, Benny H. Chang, and Farhad R. Danesh 15 BAYLOR COLLEGE OF MEDICINE FUNCTIONAL INVESTIGATION OF THE MITOCHONDRIAL PYRUVATE CARRIER IN PROSTATE CANCER David A Bader Department of Molecular & Cellular Biology/M.D.-Ph.D. Program Advisor: Sean Mcguire, M.D./Ph.D.-Department of Molecular & Cellular Biology Androgen receptor (AR)-induced de-novo lipid synthesis enables the growth and survival of hormone sensitive and castrate-resistant prostate cancer cells, but the metabolic alterations mediated by AR to support lipogenesis in prostate cancer are poorly understood. Lipogenesis requires pyruvate to generate lipid precursors (e.g. citrate) and recent in-vivo imaging strategies have demonstrated that, in contrast to virtually all other tumors, prostate tumors take up pyruvate more rapidly and reliably than glucose. Further, a subunit of the mitochondrial pyruvate carrier (MPC), MPC2, is a direct AR target gene, suggesting a link between AR action pyruvate trafficking, and lipogenesis. The overall objective of our current project is to determine the mechanism by which AR alters pyruvate trafficking to fuel lipogenesis in prostate cancer. The overarching central hypothesis is that AR regulates the MPC to increase pyruvate flux into mitochondria to enable oncogenic growth by increasing lipogenic capacity of tumor cells. Characterization of the mechanism underlying increased pyruvate trafficking in prostate cancer will likely enable the development of strategies to therapeutically manipulate pyruvate flux. Such treatments could be applied to both hormone sensitive and treatment refractory castrate-resistant prostate cancer. In order to accomplish the overall objective, the central hypothesis will be tested by pursuing the following three specific aims: 1) Determine the impact of altered MPC subunit expression on pyruvate flux by experimentally modifying MPC subunit composition and measuring a variety of metabolic endpoints in addition to using mass-spectroscopy-based methods to perform 13C pyruvate tracing. 2) Define MPC-dependent tumor properties using in-vitro measurements of tumor cell proliferation, invasive capacity, and lipogenic capacity. 3) Assess the impact of MPC perturbation on prostate tumor growth in-vivo by transplanting nude mice with prostate cancer cells harboring stable modifications to MPC subunit expression or treating xenografted mice with the MPC inhibitor UK5099. Completion of our research plan will define the metabolic and oncogenic consequences of the altered MPC subunit expression pattern observed in prostate cancer. The significance of this contribution derives from the expected translational opportunities made possible by the characterization of lipogenic substrate trafficking in prostate cancer, disruption of which represents a rational therapeutic strategy in all stages of the disease. The proposed research is innovative because it focuses on understanding and disrupting tumor-specific metabolic substrate trafficking rather than enzymatic activities common to normal tissues and tumor cells alike. It is expected this expansion of focus to include substrate trafficking will broaden the approach to metabolic targeting in cancer.Contributors: Bader, David; Coarfa, Cristian; Hartig, Sean; Sreekumar, Arun; O’Malley, Bert; McGuire, Sean 16 2014 GRADUATE STUDENT SYMPOSIUM ELEVATION OF MTORC2 ACTIVITY IS RESPONSIBLE FOR ASD-LIKE PHENOTYPES IN PTEN-MUTANT MICE Dillon Patrick Baete Department of Neuroscience Advisor: Mauro Costa-Mattioli, Ph.D.-Department of Neuroscience Autism spectrum disorder (ASD) is a heterogeneous group of diseases collectively characterized by impaired social interaction and communication, repetitive behaviors, and cognitive deficits. Although the etiology of the disease is unknown, several genes have been identified as mutated in individuals with ASD, including the phosphatase and tensin homolog gene (PTEN). PTEN encodes for the eponymous PTEN protein, a tumor suppressor and inhibitor of the PI3K pathway. PTEN directly antagonizes the actions of PI3K, dephosphorylating PIP3, thereby preventing activation of the pathway. Mutation of PTEN causes overactivation of the pathway, leading to an increase in mTOR Complex 1 (mTORC1) activity and also, through an unknown mechanism, increased mTOR Complex 2 (mTORC2) activity. Pharmacological reduction of both complexes can rescue many abnormalities associated with PTEN mutation; however, to date it is unclear which abnormalities are associated with which complex. We have generated forebrain-specific knockout mice with loss of PTEN and loss of rictor, a key mTORC2 component. We found that deletion of rictor abrogates tonic-clonic seizures, anti-social behavior, and deficient learning in memory exhibited by PTEN knockout mice. In addition, deletion of rictor nearly doubles the lifespan of PTEN knockout mice. While deletion of rictor ablates mTORC2 activity, mTORC1 activity remains elevated in PTEN knockout. These results suggest that upregulated mTORC2 activity, and not mTORC1 activity, is responsible for the epileptiform activity and ASDlike phenotypes exhibited by PTEN knockout mice. Contributors: Baete, Dillon; Huang, Wei; Costa-Mattioli, Mauro 17 BAYLOR COLLEGE OF MEDICINE ROLE OF CLN6 IN THE PATHOGENESIS OF NEURONAL CEROID LIPOFUSCINOSIS-6 (NCL-6) Lakshya Bajaj Department of Molecular & Human Genetics Advisor: Marco Sardiello, Ph.D.-Department of Molecular & Human Genetics Neuronal Ceroid Lipofuscinoses (NCLs) are the most commonly inherited neurodegenerative disorders of childhood. They are a group of autosomal recessive, progressive encephalopathies characterized by the accumulation of auto-fluorescent ceroid lipopigment in various tissues, notably in neurons. Current analyses indicate that subunit C of the mitochondrial ATP synthase and sphingolipid activator proteins (SAPs) are the two major aggregates found in the neuronal cells. The clinical course includes progressive dementia, seizures, progressive visual failure and, death by the second or third decade of life. CLN6 is an ER transmembrane protein, mutations in which are known to cause vLINCL. Similar to other NCLs, deposits of ceroid lipopigments are found in the cells. Neurons are particularly vulnerable to damage caused by lipopigments and more susceptible to cell death. The progressive death of neurons contributes to the signs and symptoms of late-infantile NCL. However, it is unclear how mutations in the CLN6 gene are involved in the buildup of lipopigments and its function in disease pathogenesis. The ongoing experiments in our lab strongly hinted that CLN8, another protein causing a very similar form of NCL is acting as a cargo transporter or a chaperone for various lysosomal hydrolases. As the defects in CLN8 and CLN6 are majorly lysosomal and these proteins result in two very similar diseases phenotypically, we postulated that CLN6 might also be acting as cargo transporters or chaperones for various lysosomal enzymes or maybe is a part of a complex with CLN8 aiding in the delivery of various lysosomal enzymes. Working towards this hypothesis we performed a mass-spectrometry analysis on the lysosomal fraction extracted from livers of onemonth-old CLN6 KO mice and we found depletion of various lysosomal hydrolases in the lysosomal compartment. We further confirmed the depletion of certain lysosomal enzymes by performing immuno-blot analysis. We investigated the difference in the enzyme activity between CLN6 KO and WT mice by performing enzyme assay on a battery of hydrolases. We are also working towards finding the mechanism by which CLN6 is aiding in the delivery of these lysosomal hydrolases. To check for the same we performed high throughput direct-interaction analysis between N-terminus YFP tagged CLN6 acting as bait and the C-terminus YFP tagged various lysosomal hydrolases in a Bimolecular florescent complementation (BiFC) assay screen using Flow-cytometry. The candidate interactors are undergoing an unbiased check using CoImmunoprecipitation (CO-IP). Using Bi-FC we have also been able to show that CLN6 homodimerizes and heterodimerizes with CLN8, the latter interaction has been further confirmed by CO-IP. Taking a lead from these results we are curious to see the relationship between CLN6 and CLN8 and are currently in a process to generate CLN6/-; CLN8-/- KO mice. Contributors: Bajaj, Lakshya; Di Ronza, Alberto; Palmieri, Michela; Lotfi, Parisa; Sharma, Jaiprakash; Sardiello, Marco 18 2014 GRADUATE STUDENT SYMPOSIUM CHARACTERIZATION OF HAL AND THE USE OF NEAT-CONTAINING PROTEINS AS A PROTEIN SUBUNIT VACCINE FOR ANTHRAX DISEASE Miriam Balderas Department of Molecular Virology & Microbiology Advisor: Anthony Maresso, Ph.D.-Department of Molecular Virology & Microbiology Iron is essential trace element for nearly all forms of life and its sequestration plays an important role in mammalian hosts defense against bacterial infection by limiting bacterial replication. The mammalian ionic iron level is far too low (10-18) to sustain normal bacterial growth, further to counteract free iron availability most of the mammalian iron is compartmentalized in heme, which is further sequestered in hemoglobin (Hb). Thus, successful bacterial pathogens have evolved mechanisms to access heme-iron pools to successfully replicate. Bacillus anthracis, a Gram-positive, spore forming bacteria and the causative agent of anthrax, has evolved mechanisms to capture sequestered iron mediated by a structural module called the near-iron transporter (NEAT) domain. In B. anthracis NEAT-containing proteins form a network of transport proteins that function to import and catabolize heme. In this study, hal (hemeacquisition leucine-rich repeat protein), a gene recently implicated in anthrax disease progression, was characterized. Hal encodes a single NEAT domain and several leucine-rich repeats. The purified recombinant NEAT domain of Hal was shown to bind heme, despite lacking a stabilizing tyrosine common to the NEAT superfamily of hemoproteins. The NEAT domain also bound Hb and acquired heme from Hb in solution. Deletion of hal resulted in bacilli unable to grow efficiently on heme or hemoglobin as an iron source and yielded the most significant phenotype relative to other putative heme uptake systems, a result that suggests this protein plays a prominent role in the replication of B. anthracis in hematogenous environments. Finally, studies using a novel vaccine, composed of the conserved protein NEAT domains of B. anthracis, suggest the NEAT subunits confer resistance and provide protective immunity to anthrax disease in a murine model of infection. This project proposes to move forward the development of NEAT domains as a vaccine for anthrax disease. These studies advance our understanding of heme acquisition by this dangerous pathogen and justify efforts to determine the mechanistic function of this novel protein for vaccine or inhibitor development. Contributors: Balderas, Miriam; Maresso, Anthony 19 BAYLOR COLLEGE OF MEDICINE THE ROLE OF SYMPATHETIC NERVOUS SYSTEM DYSFUNCTION IN HEPATOCARCINOGENESIS Anthony Patrick Barrasso Integrative Program in Molecular and Biomedical Sciences Advisor: Loning Fu, Ph.D.-Department of Pediatrics The sympathetic nervous system (SNS) is classically thought to activate the “fight-or-flight” response as a reaction to external stress. However, the SNS also plays an important role in maintaining the homeostasis of diverse physiological functions in vivo. The SNS releases epinephrine and norepinephrine, which bind α- and βadrenergic receptors (AR) to regulate many key cellular processes in peripheral tissues. There are three sub-types of β-AR (β1, 2, and 3), all of which are known to activate cell proliferation pathways. Recent studies have linked uncontrolled β-AR intracellular signaling to the initiation and progression of several cancers in both humans and mouse models. Especially, β2-AR deregulation, which has been identified as a potential prognostic marker for cancer in humans. We are specifically interested in the role of βAR signaling in hepatocellular carcinogenesis. We propose to generate a mouse model lacking all three β-ARs in the liver by first generating mice carrying floxed alleles of βAR1-3 genes via CRISPR/Cas genome editing. These mice will then be crossed with the transgenic mice expressing cre recombinase under the control of albumin gene promoter (Alb-cre). We expect to obtain the liver-specific β-less mice within 6 months. Contributors: Barrasso, Anthony; Fu, Loning 20 2014 GRADUATE STUDENT SYMPOSIUM A THERAPEUTIC NANOPARTICLE VACCINE AGAINST TRYPANOSOMA CRUZI IN A MOUSE MODEL OF CHAGAS DISEASE Meagan Amelia Barry Program in Translational Biology & Molecular Medicine/M.D.-Ph.D. Program Advisor: Peter Hotez, M.D./Ph.D.-Department of Pediatrics Laila Woc-Colburn, M.D.-Department of Medicine Chagas disease is a neglected tropical disease of great importance in the Americas, with 7-8 million people infected. The causative agent is Trypanosoma cruzi (T. cruzi), and results in an acute febrile illness that progresses to chronic chagasic cardiomyopathy in 30% of patients. In endemic areas, Chagas disease is the leading cause of cardiovascular death between ages 30-50. Current pharmacological treatments are plagued by significant side effects, poor efficacy, and are contraindicated in pregnancy. There is an urgent need for new treatment modalities. A therapeutic vaccine for Chagas disease has potential advantages that include cost savings, reduced adverse effects, and the potential to be used as a replacement for current therapies or when paired with chemotherapy. Prior work in mice has identified an efficacious T. cruzi antigen (Tc24). To elicit a protective cell-mediated immune response to the Tc24 protein, we have utilized a nanoparticle delivery system in conjunction with CpG motif-containing oligodeoxynucleotides (ODN) as an immunomodulatory adjuvant. When tested in a BALB/c mouse model, a dose response study demonstrated a positive relationship between dose of vaccine and Tc24-specific IFN-γ response. Our nanoparticle vaccine, comprised of Tc24 and CpG ODN encapsulated in poly(lactic-coglycolic acid) (PLGA) nanoparticles, produced the most robust TH1- mediated CD8+ T cell immune response. When tested for therapeutic efficacy in T. cruzi infected BALB/c mice, improved survival was seen in the vaccine group compared to the control groups. Additionally, there was a significant reduction in the number of parasites in the cardiac tissue of the vaccine group compared to the PBS sham vaccine group, indicating protection from parasite-driven cardiac damage. The mice that survived to the end of the study had almost undetectable numbers of parasites in the cardiac tissue. These data demonstrate the immunogenicity and efficacy of a Tc24/CpG ODN nanoparticle vaccine and are convincing evidence for a potential new therapeutic vaccine against Chagas disease. Contributors: Barry, Meagan; Wang, Qian; Beaumier, Coreen; Jones, Kathryn; Keegen, Brian; Bottazzi, Maria Elena; Heffernan, Michael; Hotez, Peter. 21 BAYLOR COLLEGE OF MEDICINE THE ROLE OF TRANSLATION INITIATION FACTOR 2 IN COLORECTAL CANCER Susan Michelle Benton Integrative Program in Molecular and Biomedical Sciences Advisor: Jason Heaney, Ph.D.-Department of Molecular & Human Genetics Colorectal cancer (CRC) is the third most common cancer and the second highest cause of cancer related deaths in the U.S. Mutations inactivating the gene Adenomatous Polyposis Coli (Apc) are found in familial adenomatous polyposis and account for approximately 80% of all sporadic human colorectal cancers. The Apc multiple intestinal neoplasia (Apcmin/+) mouse model is commonly used to model tumor initiation and as a tool to discover genetic modifiers of CRC risk. Cancer requires the up-regulation of oncogenes to grow and metastasize; in fact key elements of the translational machinery are up-regulated in several cancers. Several oncogenes (e.g. Cyclin D1, Myc) have been shown to be highly dependent on efficient translation initiation for their protein expression, and are highly sensitive to changes in the rate of translation initiation. Our preliminary data show that partial deficiency for the beta subunit of the translation initiation factor eIF2 (Eif2s2) significantly reduces both polyp number and size in Apcmin/+ mice, suggesting that adenoma development is influenced by the rate of translation. Using both mouse models of intestinal cancer and human colorectal cancer cell lines we will determine how modulation of translation by eIF2 affects the hallmarks of tumorigenesis, mechanism by which modulation of eIF2 affects tumorigenesis, and if drugs that target translation initiation can be used as a colorectal cancer treatment. Contributors: Maywald, Becky; Heaney, Jason 22 2014 GRADUATE STUDENT SYMPOSIUM DETERMINING HOW PD-1 DIFFERENTIALLY INFLUENCES SYNAPSE FORMATION IN CD4+ VS CD8+ T CELL SUBSETS Grant Daniel Bertolet Department of Pathology & Immunology Advisor: Dongfang Liu, M.D./Ph.D.-Department of Pediatrics T cells are a subtype of white blood cells that can be broadly divided into two groups based on the expression of one of two different surface proteins: either CD4 or CD8. CD8+ T cells are also known as “killer T cells”, as their primary function is to kill infected or malignant cells. CD4+ T cells are known as T helper cells. Their role is more complex, but they generally serve to direct and augment the immune response through the secretion of different sets of chemical messenger proteins known as cytokines. For both subsets, in order to carry out their functions they must first form what is known as an immunological synapse (IS)– a stable interface between the T cell and another cell in which biochemical information about the state of the infection is exchanged and the T cell is accordingly programmed for future action. PD-1 is a cell surface receptor expressed on both subsets of T cells, and whose main function is to temper cell activation. PD-1’s expression is known to be upregulated on virus-specific T cells during chronic viræmia and on tumor infiltrating lymphocytes in cancer. Blockade of PD-1 signalling has been repeatedly shown to improve the antitumoral and antiviral functions of so-called “exhausted” T cells, which otherwise have impaired ability to carry out necessary immune function. This demonstrates that PD-1 signalling is an important component of exhaustion. However, it is not completely understood how exactly PD-1 signalling contributes to exhaustion. One of the ways that PD-1 appears to influence T cell responses is through alteration of the IS. PD-1 co-localizes with the T cell receptors (TCRs) in the center of the IS and attenuates signals transmitted from them. This attenuation comes in the form of dephosphorylation of important TCR-proximal signalling mediaries such as ZAP-70 and PKCθ. Curiously enough, the ultimate effect of this signal-dampening differs between CD4+ and CD8+ T cells in regards to their synapse formation. In CD4+ T cells, PD-1 signalling causes disruption of the synapse, whereas in CD8+ T cells, PD-1 signalling actually strengthens the synapse. It is unknown how PD-1 does this. It is known, however, that there are innate differences in synapse formation between CD4+ and CD8+ T cells, independent of PD-1. CD8+ T cells are known to form stronger, more stable synapses than CD4+ T cells, and this is due to the activity of PKCθ. PKCθ is known to disrupt synapse formation, and CD4+ T cells recruit more PKCθ to their synapses than do CD8+ T cells. We thus hypothesize that PD-1 differentially influences synapse formation in CD4+ and CD8+ T cells by acting on the different levels of PKCθ at the synapse in both subsets. Contributors: Liu, Dongfang 23 BAYLOR COLLEGE OF MEDICINE DECIPHERING THE ROLE OF FOXI3 IN MOUSE DEVELOPMENT Onur Birol Program in Developmental Biology Advisor: Andrew Groves, Ph.D.-Department of Neuroscience Foxi3 is a Forkhead transcription factor that is expressed early in chick and mouse in the preplacodal region. This region gives rise to multiple craniofacial placodes, such as lens, trigeminal or epibranchial in addition to otic placode, the precursor to the inner ear. Inner ear induction is triggered by FGF signaling, and competence to induce otic placode markers in response to FGFs correlates with the expression of preplacodal markers such as Foxi3. Our lab has knocked out Foix3 in mice and Foxi3 mutants completely lack all ear structures and also have additional craniofacial defects. Ear induction appears to fail at the very earliest stages, as we see no markers of the otic placode, such as Pax2, induced in Foxi3 mutants. Foxi3, like other Forkhead transcription factors, has a DNA binding domain that resembles H1 and H5 linker histones and it is believed that due to this structural resemblance it may serve as a pioneer factor that can bind to condensed chromatin and help to make it transcriptionally accessible. This in turn may induce competence for the cell to respond to future inducing signals. At one extreme, Foxi3 might simply confer competence for the expression of inner ear genes after preplacodal ectoderm receives FGF signals. Foxi3 might also provide competence to preplacodal ectoderm by regulating genes (such as FGF receptors) that to respond to FGF signals. At the other extreme, Foxi3 might be required to induce preplacodal genes in the first place. We are analyzing the mutant mice to pinpoint, where exactly the Foxi3 plays role in FGF signaling since the mutant mice lack Pax2 induction, which is one of the first responsive genes in ear. Our results indicate to an earlier role of Foxi3 because we have observed that some of the preplacodal markers are not expressed in mutant mice ectoderm. Moreover, the FGF pathway genes analyzed to date have revealed no difference in expression between mutants and wild types. This suggests that the preplacodal tissue is receiving the inducing FGF signaling; however, due to defects on earlier gene expressions, it is unable to interpret it to turn on the ear development program. On the other hand, Foxi3 is also expressed early embryonically in some neural tissue and later in preplacodal region that is very close to the presumptive neural crest tissues. We are not sure whether these expressions are biologically relevant. In some Foxi3 mutant mice, the embryos exhibit exencephaly phenotype. Furthermore, the branchial arches that are normally filled with neural crest cells fail to form. Considering all these, we hypothesize that Foxi3 is biologically significant for some neural tissue and neural crest cells. In order to investigate this part, we plan to perform fate mapping using Foxi3-CreER mouse line and tissue specific knock out studies. 24 2014 GRADUATE STUDENT SYMPOSIUM ELUCIDATING ROLE OF ATXN1L IN SCA1 Vitaliy V Bondar Department of Molecular & Human Genetics Advisor: Huda Zoghbi, M.D.-Department of Pediatrics Spinocerebellar Ataxia type 1 (SCA1) is a dominantly inherited fatal neurodegenerative disorder for which there is no available treatment. It is caused by the expansion of a trinucleotide repeat that encodes a polyglutamine (polyQ) tract in ATAXIN-1 (ATXN1), rendering the protein more stable and resulting in toxic accumulation of mutant ATXN1. Investigation into molecular mechanisms of SCA1 pathogenesis revealed that mutant ATXN1 exerts deleterious effect through its interacting partners that include Capicua (CIC), a transcriptional repressor that forms stable complexes with ATXN1. Previously we showed that in presence of expanded polyQ ATXN1, CIC significantly increases transcriptional repression of its target genes. Furthermore, haploinsufficiency of Cic rescues neurodegeneration in SCA1 mouse model through reduction of aberrant repression. Another interacting partner of ATXN1 is ATAXIN-1-Like (ATXN1L), which is a paralog of ATXN1 with high amino acid sequence identity. Yeast-two-hybrid screen revealed that ATXN1 and ATXN1L share many common interactors such as CIC. Although loss of either Atxn1 or Atxn1L in mice does not cause lethality, double knock out of Atxn1 and Atxn1L results in perinatal lethality and thus providing further evidence for their shared function. Interestingly, duplication of Atxn1L in mice rescues SCA1 by out-competing mutant Atxn1 from its interacting partners. Given importance of ATXN1L interacting partners in understanding SCA1 pathogenesis we aim to identify interacting network for ATXN1L, in vivo. We propose that identifying endogenous protein interacting partners of Atxn1L may reveal crucial players in SCA1 pathogenesis. To accomplish this we engineered an Atxn1L knock-in mouse (Atxn1L-V mice, V for Venus) with C-terminal tags for which good monoclonal antibodies are available. Further biochemical studies revealed that epitope tag in Atxn1L does not interfere with its binding ability. Currently we are performing immunoprecipitation followed by mass spectrometry to identify Atxn1L interacting partners in different brain regions. Contributors: Jefar-Nejad, Paymaan; Tan, Qiumin; Zoghbi, Huda 25 BAYLOR COLLEGE OF MEDICINE THE ROLE OF P300 IN KEY ASPECTS OF LIVER BIOLOGY Meghan Breaux Integrative Program in Molecular and Biomedical Sciences Advisor: Nikolaj Timchenko, Ph.D.-Department of Pathology & Immunology Frederick Pereira, Ph.D.-Department of Molecular & Cellular Biology Epigenetic control is a key process involved in liver regulation. Recent studies have shown that several age-related liver dysfunctions are associated with the change of chromatin structure. These dysfunctions include development of hepatic steatosis (fatty liver), impaired liver proliferation and age-associated liver cancer. Previous studies in Dr. Timchenko’s lab revealed that two members of C/EBP family, C/EBP and C/EBP , are altered by age and are involved in alteration of chromatin structure in the liver. These observations were obtained in several animal models of aging liver including C/EBP -S193D knockin mice and recently generated transgenic mice expressing dominant negative p300, dnp300 mice. We have found that histone acetyltransferase p300 co-operates with C/EBP proteins in the epigenetic control of liver functions and that age dramatically changes p300-C/EBP pathways in the liver. The dnp300 mice have altered expression of C/EBP proteins compared to WT and higher basal proliferation. Upon challenges to the liver using several methods including Carbon Tetrachloride treatment and Partial Hepatectomy, we observe that dnp300 mice have higher rates of proliferation and are more resistant to liver injury. Therefore, the main goal of my project is to determine the role of p300-dependent epigenetic control in the key functions of the liver. Contributors: Breaux, Meghan; Jin, Jingling; Iakova, Polina; Jiang, Yanjun 26 2014 GRADUATE STUDENT SYMPOSIUM MODAFINIL + LISDEXAMFETAMINE FOR COCAINE USE DISORDER Alex Jawann Brewer III Department of Pharmacology Advisor: Richard De La Garza, Ph.D.-Department of Psychiatry & Behavioral Sciences AIMS: The dopamine transporter (DAT) has been implicated in the subjective and reinforcing effects produced by cocaine. Modafinil has shown some efficacy in reducing these effects. The aim of this study was to determine if the ability of modafinil to reduce the subjective and reinforcing effects produced by cocaine could be enhanced by combination with another drug, lisdexamfetamine, that is also known to interact with the DAT. METHODS: Non-treatment seeking, cocaine-dependent individuals had a history of cocaine use via the smoked or intravenous route, were 18-55 years old, were not dependent on other drugs of abuse excluding nicotine, and did not have unstable medical conditions or psychiatric diagnoses. Eligible individuals were randomized to one of four treatments: placebo, modafinil (200 mg, p.o.), lisdexamfetamine (30 mg p.o.), or modafinil (200 mg, p.o.) + lisdexamfetamine (30 mg, p.o.) for four days. On the fourth day, individuals participated in two double-blind self-administration sessions for the opportunity to receive cocaine or saline. Upon receiving a contingent dose of cocaine (20 mg, iv) or saline, individuals had the opportunity to make 5 subsequent choices for either another infusion or keep $1 USD. The sessions were counterbalanced so that some individuals received cocaine in the morning and saline in the afternoon session, and vice versa. Subjective effects and heart rate and blood pressure were recorded prior to and throughout each session. RESULTS: Participants (n = 27) were mostly male (74%), mostly African American (78%), and non-Hispanic (85%). Expectedly, compared to saline, cocaine 20 mg significantly increased heart rate and systolic blood pressure, and positive subjective effects of cocaine, including “high”, “like drug”, “good effects”, and “stimulated” (all p-values < 0.001). Compared to the other treatments, modafinil attenuated subjective ratings of “high”, “desire”, “any drug effects”, “good effects”, and “stimulated” , but the combination of modafinil and lisdexamfetamine did not produce significant attenuations of any subjective or physiological responses induced by cocaine (all p-values > 0.05). CONCLUSIONS: These data support previous findings that modafinil is effective for treating cocaine dependence, yet it does not appear to be more effective when combined with lisdexamfetamine. Contributors: Brewer III, Alex J.; Mahoney III, James J.; Newton, Thomas F., De La Garza II, Richard 27 BAYLOR COLLEGE OF MEDICINE IDENTIFICATION OF NOVEL ALTERATIVE SPLICING NETWORKS DURING POSTNATAL SKELETAL MUSCLE DEVELOPMENT Amy Elizabeth Brinegar Department of Molecular & Cellular Biology Advisor: Thomas Cooper, M.D.-Department of Pathology & Immunology Alternative splicing increases proteome diversity by producing isoforms that help define tissue specificity. During development, alternative splicing is highly regulated and the loss of regulation causes mis-splicing, which can lead to pathogenesis of some diseases. Postnatal heart development has a well-established global switch in alternative splicing due to changes in the protein levels of splicing regulators. Skeletal muscle has a similar switch in alternative splicing regulators during development that also results in a global switch in alternative splicing. The postnatal period, encompassing the first four weeks after birth, is a very dynamic time in skeletal muscle development with many changes at the cellular and molecular level. The skeletal muscle cells transitions from a highly proliferative population of mononucleated cells to quiescent terminally differentiated myofibers. Also during this time, innervation of myofibers matures through neuronal pruning and sarcomeres mature to ensure proper muscle function. Although some muscle-specific splicing switching has been identified during postnatal development, very few isoforms have a known biological function related to muscle physiology. To identify novel transcriptome changes, particularly alternative splicing events, that affect muscle physiology, I have performed RNA-seq analysis across different time points of postnatal development. To narrow my scope and prioritize alternative splicing events, I have also performed GO analysis. From here, I will determine the biological significance of the splicing events by redirecting particular splicing events in vivo that will be examined by muscle-related assays. With this, we will better understand the impact that alternative splicing has on normal muscle biology. If these splicing events are also mis-regulated in disease, these splicing events can be targeted to potentially correct muscle function. Contributors: Brinegar, Amy; Xia, Zheng; Manning, Kathleen; Li, Wei; Cooper, Thomas 28 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF NOTCH SIGNALING IN THE DEVELOPMENT OF THE VESTIBULAR SYSTEM OF THE INNER EAR Rogers Milton Brown, II Program in Developmental Biology Advisor: Andrew Groves, Ph.D.-Department of Neuroscience The inner ear is a complex and specialized vertebrate organ deeply embedded in the function of the auditory sensory pathway as well as in the detection of linear and angular acceleration. The ability of the inner ear to do so is dependent on the six sensory organs contained within the structure. In general, these sensory organs consist of apically ciliated sensory epithelial cells called hair cells and the supporting cells that surround them. There is much that is known about the function of these hair cells, as well as their morphology, but there is less that is known about the developmental cascade by which hair cells arise and differentiate. It is thought that Notch signaling plays a key role in inner ear development at three stages: first during neurogenesis to generate delaminating neuronal precursors, later to promote the pro-sensory domains that will give rise to the sensory organs of the inner ear, and finally to establish the pattern of hair cells and supporting cells during hair cell differentiation through lateral inhibition. Previous studies performed to analyze the role of Notch in these processes revolved around removing key ligands or receptors in the Notch signaling pathway like Jag1 and Jag2, Notch1, and Dll1. The phenotypes observed in these experiments demonstrated a range of results which suggests that when some of these elements are removed from the pathway, other receptors or ligands may be able to compensate in certain situations. Through the use of a dominant-negative Mastermind-like (dnMAML) and a conditional knock out of RBPJ-K (RBPJ CKO), we hope to observe the effect of the complete loss of Notch signaling on the development of the pro-sensory domains that generate the hair cells and supporting cells of the vestibular sensory organs. By attacking the “bottleneck” of the pathway we hope to successfully abolish all Notch signaling in the inner ear. Through the use of paint fillings to understand the threedimensional development of these mutants, and both RNA in situ hybridization and fluorescent immunochemistry for markers of pro-sensory development, we hope to obtain a clearer picture of the function the Notch pathway plays in pro-sensory development. Contributors: Brown, Rogers II; Basch, Martin; Groves, Andrew 29 BAYLOR COLLEGE OF MEDICINE CILIA FUNCTION AND OOCYTE TRANSPORT IN THE MURINE OVIDUCT Jason Christopher Burton Integrative Program in Molecular and Biomedical Sciences Advisor: Irina Larina, Ph.D.-Department of Molecular Physiology & Biophysics Investigation of pre-implantation events in the murine oviduct is crucial in understanding normal reproductive functions; however, detailed analysis of the ciliated layer of the oviduct has been restricted, due to the complexity of the transport system and the lack of proper imaging tools. We introduce an innovative approach for threedimensional imaging and tracking of oocytes through the oviduct with optical coherence tomography (OCT), as well as a novel Functional-OCT imaging modality to analyze the dynamics of motile cilia in the oviduct. Imaging is performed with a spectral-domain OCT system operating at 70 kHz A-scan rate with a measured resolution near 5 microns. Oocytes were tracked in vivo from ovulation as they transit through the oviduct towards the uterus. The movement of the oocyte can be clearly visualized and the velocity vectors can be quantified based on direct mapping of the trajectories. Dynamics of cilia beat frequency and location of ciliated cells were extracted from twodimensional OCT images using a Fourier Transform. Extracted frequency information was mapped back onto OCT images to highlight ciliated epithelial layers. Motile cilia are responsible for transport of the pre-implantation embryos and a quantitative analysis of their activity during different stages of pregnancy increase our knowledge of dynamic events during early pregnancy. To the best of our knowledge, this is the first application of OCT to the study of dynamic events in the murine oviduct and the first attempt to develop Functional-OCT techniques that allow for analysis of motile cilia. OCT is capable of producing detailed structural information of in vivo processes that allows for comprehensive studies of dynamic pre-implantation events. This study will increase our understanding of pregnancy events and may lead to further investigations of tubal and cilia defects and their impact on fertility. Contributors: Burton, Jason C.; Wang, Shang ; Larina, Irina V. 30 2014 GRADUATE STUDENT SYMPOSIUM TEM8 SPECIFIC T CELLS TARGET THE TUMOR CELLS AND TUMOR ASSOCIATED VASCULATURE IN TRIPLE NEGATIVE BREAST CANCER Tiara T Byrd Program in Translational Biology & Molecular Medicine Advisor: Nabil Ahmed, M.D.-Department of Pediatrics Robert Grossman, M.D.-Department of Neurosurgery Background: Tumor endothelium marker 8 (TEM8) was discovered by St Croix et al as one of nine gene products preferentially upregulated in the tumor-associated vs. normal endothelium. Interestingly, TEM8 has also been identified as a tumor restricted antigen in triple negative breast cancer (TNBC), a clinical entity associated with a particularly poor prognosis. Being null for HER2, estrogen and progesterone receptors, targeted therapies for TNBC are quite limited. Purpose: To use T cells expressing TEM8-specific chimeric antigen receptors (CAR) as a novel approach to target both TNBC cells and their tumor-associated vasculature. Methods/Results: We used in silico design to construct a novel TEM8-specific CAR molecule. The antigen recognition exodomain consisted of a single chain variable fragment based on the TEM8-specific monoclonal antibody, L2. The signaling endodomain consisted of the costimulatory molecule CD28 and CD3-zeta chain. The encoding DNA was codon optimized, synthesized and then sequence verified. We used a retroviral transduction system to express the TEM8 CAR transgene on HEK 293T, then on primary T cells. Approximately 90% of primary human T cells expressed the TEM8 CAR, as detected by flow cytometry. The expression of TEM8 was characterized using flow cytometry and western blot on a battery of TNBC lines, TEM8 transduced (modest and high expressers) cell lines as well as TEM8 negative cell lines. TEM8 CAR T cells recognized and killed TEM8 positive target cells in an antigen-dependent fashion in 51Cr release assays and secreted immunostimulatory cytokines upon encounter of TEM8 positive cells. There was no reactivity against TEM8 negative cell lines. No cytotoxicity or cytokine release was exhibited by T cells expressing an irrelevant CAR or non-transduced T cells from the same blood donor. We are currently testing this strategy in a vascularized orthotopic breast cancer murine model. Conclusion: TEM8 specific CAR T cells could serve as a tumor and vascular targeted immunotherapeutic modality for triple-negative breast cancer. Contributors: Byrd, Tiara; Fousek, Kristen; Pignata, Antonella; Wakefield, Amanda; St Croix, Bradley; Fletcher, Bradley; Hegde, Meenakshi and Ahmed, Nabil 31 BAYLOR COLLEGE OF MEDICINE DEVELOPMENT OF NEOCORTICAL CIRCUITS Cathryn Rene Cadwell Department of Neuroscience/M.D.-Ph.D. Program Advisor: Andreas Tolias, Ph.D.-Department of Neuroscience The mammalian neocortex gives rise to complex cognitive processes such as perception and decision-making. Despite tremendous progress in understanding the physiology and cell biology of individual neurons in the cortex, the process by which networks of neurons become wired together during development and organize into functional circuits remains poorly understood. Recent studies suggest that cell lineage influences the connectivity and functional properties of excitatory pyramidal neurons in the neocortex, but it remains unclear to what extent cell lineage influences circuit assembly. We have developed a tamoxifen-inducible Cre/lox system for lineage tracing in the neocortex that allows us to sparsely label neural progenitors and trace their daughter cells. When progenitors are labeled at the onset of neurogenesis, they give rise to columns of clonally related pyramidal neurons spanning all six cortical layers. Interestingly, we find that sister cells in ontogenetic columns show enhanced connectivity across cortical layers but not within the same cortical layer. By utilizing 3D random-access multiphoton (3D-RAMP) imaging, we are also able to study the functional organization of ontogenetic columns in the visual cortex. These experiments will shed light on how functional networks are established during development and could provide a circuit level foundation to study neurodevelopmental disorders. Contributors: Cadwell, Cathryn; Jiang, Xiaolong; Froudarakis, Emmanouil; Yatsenko, Dimitri; Tolias, Andreas 32 2014 GRADUATE STUDENT SYMPOSIUM PERCEIVED DURATION IS REDUCED BY REPETITION, BUT NOT BY HIGH-LEVEL EXPECTATION Mingbo Cai Department of Neuroscience Advisor: David Eagleman, Ph.D.-Department of Neuroscience A repeated stimulus is perceived as briefer than a novel one. It has been suggested that this duration illusion is an example of a more general phenomenon, namely that a more expected stimulus is perceived as briefer than a less expected one. To test this hypothesis, we manipulated high-level expectation through the probability of a stimulus sequence, through the regularity of the preceding stimuli in a sequence, or by violating an overlearned sequence. We found that perceived duration is not distorted by these manipulations. Repetition of stimuli, on the other hand, consistently reduces perceived duration across our experiments. Our findings suggest that estimates of subsecond duration are largely the result of low-level processing and independent of highlevel expectation. Contributors: Cai, Mingbo; Eagleman, David M.; Ma, Wei Ji 33 BAYLOR COLLEGE OF MEDICINE PARENTAL SOMATIC MOSAICISM IS UNDERRECOGNIZED AND INFLUENCES RECURRENCE RISK OF GENOMIC DISORDERS Ian Morgan Campbell Department of Molecular & Human Genetics/M.D.-Ph.D. Program Advisor: James Lupski, M.D./Ph.D.-Department of Molecular & Human Genetics Pawel Stankiewicz, M.D./Ph.D.-Department of Molecular & Human Genetics New human mutations are thought to originate in germ cells, thus making a recurrence of the same mutation in a sibling exceedingly rare. However, increasing sensitivity of genomic technologies has anecdotally revealed mosaicism for mutations in somatic tissues of apparently healthy parents. Such somatically mosaic parents might also have germline mosaicism that can potentially cause unexpected intergenerational recurrences. Here, we show that somatic mosaicism for transmitted mutations among parents of children with simplex genetic disease is more common than currently appreciated. Using the sensitivity of individual-specific breakpoint PCR, we prospectively screened 100 families with children affected by genomic disorders due to rare deletion copy-number variants (CNVs) determined to be de novo by clinical analysis of parental DNA. Surprisingly, we identified four cases of low-level somatic mosaicism for the transmitted CNV in DNA isolated from parental blood. Integrated probabilistic modeling of gametogenesis developed in response to our observations predicts that mutations in parental blood increase recurrence risk substantially more than parental mutations confined to the germline. Moreover, despite the fact that maternally transmitted mutations are the minority of alleles, our model suggests that sexual dimorphisms in gametogenesis result in a greater proportion of somatically mosaic transmitting mothers who are thus at increased risk of recurrence. Therefore, somatic mosaicism together with sexual differences in gametogenesis might explain a considerable fraction of unexpected recurrences of X-linked recessive disease. Overall, our results underscore an important role for somatic mosaicism and mitotic replicative mutational mechanisms in transmission genetics. Contributors: Campbell Ian; Yuan Bo; Robberecht Caroline; Pfundt Rolph; Szafranski Przemyslaw; McEntagart Meriel; Nagamani Sandesh; Erez Ayelet; Bartnik Magdalena; Wisniowiecka-Kowalnik Barbara; Plunkett Katie; Pursley Amber; Kang Sung-Hae; Bi Weimin; Lalani Seema; Bacino Carlos; Vast Mala; Marks Karen; Patton Michael; Olofsson Peter; Patel Ankita; Veltman Joris; Cheung Sau Wai; Shaw Chad; Vissers Lisenka; Vermeesch Joris; Lupski James; Stankiewicz Pawel 34 2014 GRADUATE STUDENT SYMPOSIUM CYCLIC NUCLEOTIDE RECOGNITION MECHANISM OF CYCLIC GMP DEPENDENT PROTEIN KINASE II James Christopher Campbell Program in Structural and Computational Biology and Molecular Biophysics Advisor: Choel Kim, Ph.D.-Department of Pharmacology Membrane bound type II cGMP dependent protein kinase (PKG II) is a central mediator of the cGMP signaling cascade, which regulates circadian rhythmicity, intestinal water secretion, bone growth and renal function. PKG II contains a N-terminal regulatory (R)-domain, and a C-terminal catalytic (C)-domain. The R-domain contains tandem cyclic nucleotide binding domains (CNB-A and B) each with different affinities for cGMP, the second messenger that regulates kinase activity of PKG II. Once PKG II is active by cGMP it phosphorylates pharmaceutically relevant protein target, but due in part to a lack of structural information drug discovery efforts on have been slow. While it is known that PKG II needs to be highly selective for cGMP over cAMP to prevent cross-talk little is known about its cyclic nucleotide selectivity mechanism and the allosteric activation of PKG II. We seek to understand the cyclic nucleotide selectivity of PKG II’s CNB domains and their role in the activation of PKG II. In this pursuit, we characterized the CNB domain’s cyclic nucleotide selectivity. Our findings are that CNBB imparts an almost 500 fold preference for cGMP, while CNB-A only offers a 10 fold preference. We then solved atomic resolution crystal structures of the two CNB domains of PKG II, to gain insight into the molecular basis of the selectivity seen. We have discovered a unique ligand recognition mechanism in CNB-B and tested its role in the activation of PKG II. Contributors: Campbell, James; Li, Kevin; Huang, Gilbert; Reger, Albert; Sankaran, Banumathi; Link, Todd; Ladbury, John; Kim, Choel 35 BAYLOR COLLEGE OF MEDICINE LONG-TERM NEUROPROTECTION WITH PHARMACOLOGICAL HYPOTHERMIA BY TRPV1 AGONISM IN ISCHEMIC STROKE Zhijuan Cao Program in Cardiovascular Sciences Advisor: Sean Marrelli, Ph.D.-Department of Anesthesiology Background: Therapeutic hypothermia is a promising strategy for neuroprotection following stroke. However, current widely available methods are not compatible with conscious patients and produce unwanted physiological stress. We have recently demonstrated that pharmacological hypothermia (PH) through transient receptor potential vanilloid 1 (TRPV1) channel agonism is effective and safe in the conscious mouse and provides significant neuroprotection following ischemic stroke at 24 hours reperfusion. We now evaluate whether this method of PH provides sustained neuroprotection through one month reperfusion. Methods: Three experimental groups were evaluated: sham operated (Sham), stroke with normothermia (Stroke/NT), and stroke with PH (Stroke/PH) (n=8, 10, 7). Stroke was induced by transient occlusion of left distal middle cerebral artery (MCA) plus left common carotid artery for two hours. For the Stroke/PH group, hypothermia (32-34ºC) was induced by infusion of TRPV1 agonist (dihydrocapsaicin) for 8 hours; beginning 90 minutes after reperfusion. We evaluated several behavioral tests from prestroke up to 28 days reperfusion. Total cerebral infarct was then evaluated in Nisslstained brain sections at 30 days reperfusion. Results: The foot fault test was the most sensitive in demonstrating extended functional deficit after stroke. With this test, Stroke/NT demonstrated prolonged deficiency (through 21 days reperfusion) compared to Sham. PH treatment (Stroke/PH) demonstrated significant improvement in function compared to Stroke/NT. Other tests demonstrated non-statistically significant performance reduction of Stroke/NT in swimming time, grip side preference, turning and climbing time. Histological analysis demonstrated more severe ipsilateral necrosis and hemisphere shrinkage in Stroke/NT brains compared to Stroke/PH brains. Conclusions: In summary, we demonstrate long-term neuroprotection with TRPV1-mediated PH following stroke. These studies support the therapeutic potential of TRPV1 agonism for promoting hypothermia in the conscious stroke subject. Supported by NIH R21NS077413 & AHA PRE16900006. Contributors: Balasubramanian, Adithya; Marrelli, Sean P 36 2014 GRADUATE STUDENT SYMPOSIUM A SINGLE SEIZURE RESULTS IN DEFICITS IN LONG-TERM LEARNING AND MEMORY AS WELL AS ALTERED SIGNALING IN mTOR SIGNALING Angela N Carter Department of Neuroscience Advisor: Anne Anderson, M.D.-Department of Pediatrics RATIONALE: Research using animal models of epilepsy has implicated hyperactive signaling of the mammalian target of rapamycin (mTOR) pathway as a candidate mechanism underlying the pathophysiology of epilepsy and the associated co-morbidities. In physiological conditions, mTOR signaling regulates protein synthesis and is necessary for learning and memory. However, how hyperactive mTOR signaling in epilepsy results in learning and memory deficits is still unclear. In addition, how seizures affect learning and memory has yet to be fully elucidated. METHODS: To evaluate the effects of a single seizure on learning and memory as well as mTOR signaling, we administered either saline (Sham) or the convulsant pentylenetetrazole (PTZ; 75mg/kg, i.p.) in rats. Our pilot studies show that this dose was sufficient to induce a generalized seizure. To determine the effects of a single seizure on learning and memory, animals were subjected to the Fear Conditioning (FC) task. To evaluate how a single seizure affects mTOR signaling, PTZ was administered and tissue collected at 3 different time points and homogenized for western blotting. The time points were: 1-, 3-, and 24- hours post induction. Antibodies against Phospho (P)S6 and P-eEF2 were used as downstream markers of mTOR activation. To determine the effect of mTOR inhibition on FC as well as P-S6 and P-eEF2, the mTOR inhibitor rapamycin (Rap) or vehicle was given immediately post seizure. To verify inhibition of mTOR signaling, tissue was collected for western blotting. RESULTS: Following a PTZ-induced seizure, neither the Sham nor the PTZ animals displayed short-term memory deficits in the FC task. When tested for long-term memory however, the PTZ animals exhibited deficits in contextual and associative memory (n=7-11, p<0.01) compared to Sham rats. Biochemical analyses reveal that at 1 hour following a PTZ-induced seizure, the levels of P-S6 and P-eEF2 were significantly increased (n=4-6, p<0.001). At 3-hours post seizure, P-eEF2 remained elevated while P-S6 levels returned to baseline (n=4-6, p<0.01). Finally, both P-S6 and P-eEF2 returned to baseline levels 24-hours post seizure. The increased P-S6 levels (n=3-5, p<0.05) were blocked with Rap treatment however the P-eEF2 levels remained elevated (n=3-5) and Rap did not rescue the deficits in FC (n=6-11). CONCLUSIONS: These data confirm that a single PTZ-induced seizure leads to long-term memory deficits. Additionally, our data show that the deficits observed in FC are not rescued by inhibition of mTOR signaling. Studies are underway to further characterize PTZ-induced seizures and to determine other signaling pathways that may underlie the seizure related memory deficits. Contributors: Dao, A.T, Muyco, M. A., Lee, W.L., Anderson, A.E. 37 BAYLOR COLLEGE OF MEDICINE ARHGEF4 INFLUENCES OLIGODENDROCYTE PRECURSOR CELLS RECRUITMENT AT LESION SITE IN A WHITE MATTER INJURY MODEL Lesley S.M. Chaboub Program in Developmental Biology Advisor: Benjamin Deneen, Ph.D.-Department of Neuroscience While very diverse in their origins and presentations, demyelination disorders of the central nervous system (CNS) share common cellular events. Recruitment of oligodendrocyte precursor cells (OPCs) at the lesions site, which normally function to remyelinate axons, is an especially important step in proper remyelination, and has been shown to be defective in at least two disorders; multiple sclerosis and hypoxic ischemic encephalopathy (HIE). In order to understand how these cellular events occur during the disease state, it is imperative to understand how these same processes are regulated during normal development. Therefore, identification of novel genes involved in glial cell development might shed light into demyelination disorders, especially white matter injury (WMI). In a developmental screen performed in the spinal cord, we recently identified Arhgef4 as a novel gene expressed in both glial precursor cells and mature astrocytes. Arhgef4 is a guanine exchange factor for the Rho GTPase family of protein and has been shown to regulate migration in other systems. During spinal cord development, we showed that Arhgef4 influences glial precursor cells migration. We hypothesize that Arhgef4 would also influence migration of precursor cells in disease state. Lysolecithin injection into the ventral white matter region of the spinal cord is a well-recognized model of WMI where the kinetics and cellular events of remyelination are well established. Using this model, we showed that Arhgef4 is expressed in the lesion area, validating the use of this technique to further our understanding the role of Arhgef4 in WMI. Lesions performed on Arhgef4 germline knock-out (KO) animals appear to have delayed remyelination kinetics when compared to heterozygous littermate animals. Furthermore, Arhgef4 KO lesions present with a significant decrease in the number of OPCs present inside the lesion area, suggesting that the recruitment of OPCs at lesion sites is inhibited in the absence of Arhgef4. We will utilize in vitro OPC cultures to differentiate whether Arhgef4 influences the migration of OPCs into the lesion site or their proliferation. Ongoing studies will focus on further characterizing Arhgef4 KO lesions as well as performing misexpression of Arhgef4 into the lesion to assess whether it can accelerate remyelination. In addition we will assess the effect of Arhgef4 expression in a mouse WMI hypoxic model that closely mimics HIE. This study identified a novel player in OPCs recruitment at the lesion site during remyelination events. Guanine exchange factors, such as Arhgef4, are potentially good target for therapies since they affect critical cellular pathways while being tissue specific. Contributors: Chaboub, Lesley; Lee, Hyun-Kyoung and Deneen, Benjamin. 38 2014 GRADUATE STUDENT SYMPOSIUM THE MECHANISMS OF DNA DAMAGE RESPONSE IN MAMMARY STEM CELLS AND TUMOR INITIATING CELLS FROM A p53-NULL BREAST CANCER MODEL Chi-Hsuan Chang Integrative Program in Molecular and Biomedical Sciences Advisor: Jeffrey Rosen, Ph.D.-Department of Molecular & Cellular Biology The DNA damage response (DDR) includes activation of cell cycle checkpoints, apoptosis, and DNA repair. To repair DNA double strand breaks, cells may undergo homologous recombination (HR) or non-homologous end joining (NHEJ). Adult stem cells tend to have different DDR mechanisms compared to differentiated cells since they reside in the tissues for extended periods and continuously ensure tissue homeostasis. Studies have shown that mouse hair follicle bulge stem cells and hematopoietic stem cells have higher NHEJ activity and lower apoptosis, resulting in their resistance to radiation. However, little is known about the DDR in mammary stem cells (MaSCs). In addition, previous studies from the clinic and mouse models have demonstrated that tumor initiating cells (TICs) from mammary tumors have an increased resistance to conventional therapies. However, the mechanisms underlying the DDR to this therapeutic resistance are still mainly undiscovered. Using a syngeneic p53-null tumor model, our lab has identified a Lin-CD29HCD24H subpopulation in these tumors with functional TICs properties, and these TICs are more resistant to radiation. To study the specific mechanisms that contribute to this resistance, we hypothesized that TICs from p53-null tumors exhibit increased DDR and elevated NHEJ activity to repair DNA damage, which might be the similar DDR mechanisms used in MaSCs. To date, we have shown that MaSCs and basal cells have lower apoptosis as compared to luminal cells in mammary glands. In cell cycle analysis, a significant increase of G2/M phase was observed in MaSCs after irradiation, indicating stem cells have increased G2 checkpoint activation as compared to basal and luminal cells. In addition, using a high-throughput NHEJ assay, we showed that basal cells, which contain MaSCs, exhibit higher NHEJ activity as compared to luminal cells. Furthermore, using Reverse Phase Protein Array (RPPA) analysis, we were able to show that in stem cells, some stress-responsive proteins were up-regulated, such as p-p38, p-p44, and conversely that most pro-apoptotic genes were down-regulated, indicating that MaSCs are intrinsically more resistant to stress and apoptosis. In tumor studies, TICs have more efficient DNA repair according to γH2AX foci staining and neutral Comet assay. We also showed that TICs exhibit decreased apoptosis and higher NHEJ activity. Ongoing studies are designed to identify the “Achilles heel” of these DDR pathways in TICs, which may act as therapeutic targets. Contributors: Chang, Chi-Hsuan; Zhang, Mei; Lu, Tao; Coarfa, Cristian; Rosen, Jeffrey 39 BAYLOR COLLEGE OF MEDICINE LOSS OF THE PROTO-ONCOGENE BMI1 FACILITATES HEMATOPOIETIC STEM CELL DEPLETION AFTER ESTROGEN EXPOSURE Rich Harrison Chapple Department of Molecular & Human Genetics Advisor: Daisuke Nakada, B.A.Sc.-Department of Molecular & Human Genetics The hematopoietic system is a highly dynamic tissue that exhibits remarkable plasticity and is characterized by high cell turnover. To circumvent the constant depletion of these constituents, multipotent hematopoietic stem cells (HSCs) are infrequently recruited out of quiescence wherein they become proliferative cells. In this capacity, HSCs are capable of generating new blood cells to promote tissue homeostasis. However, entry into the cell cycle must be tightly regulated in order to maintain stem cell integrity and preserve HSC function. One such gene involved in the regulation of HSC division is B cell-specific Moloney murine leukemia virus integration site 1 (Bmi1). Bmi1 is a member of the Polycomb repressive complex 1 (PRC1), and is involved in epigenetic silencing of the Cdkn2a tumor suppressor locus1. Disrupting the expression of Bmi1 has deleterious consequences on HSC function – overexpression of Bmi1 is associated with poor prognosis in AML2, and deletion of Bmi1 impairs HSC selfrenewal and long-term reconstitution potential3. Interestingly, we have recently observed sex differences in the HSC response after Bmi1 deletion. More specifically, HSC frequency in Bmi1-KO females become progressively depleted compared to that of male counterparts. We surmised that the female sex hormone estrogen (E2) was responsible for the observed differences, and subjected male Bmi1-KO animals to an estrogen regimen. After E2 treatment, a significant reduction in HSC frequency was observed in Bmi1 deficient animals. The results of these experiments raise the possibility that Bmi1 deficiency may sensitize HSCs to division-promoting stimuli. We are now focusing our efforts to determine the molecular underpinnings of E2-mediated HSC division through NGS applications. The results of these data will not only be used for further analysis in our Bmi1-KO model, but also will be used for the discovery of mechanisms that induce HSC division. 1. Sparmann, A. & Van Lohuizen, M. Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer 6, 846–856 (2006). 2. Chowdhury, M. et al. Expression of Polycomb-group (PcG) protein BMI-1 predicts prognosis in patients with acute myeloid leukemia. Leukemia 21, 1116-1122 (2007). 3. Park, I. et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature 423, 302-305 (2003). Contributors: Chapple, Richard; Takeichi, Makiko; Kitano, Ayumi; Saitoh, Yusuke; Lin, Angelique; Nakada, Daisuke 40 2014 GRADUATE STUDENT SYMPOSIUM BINDING TO HUMAN ΓD-CRYSTALLIN CAUSES PARTIAL CLOSURE OF CIS-RING AND SYMMETRY-BROKEN FEATURES OF TYPE II CHAPERONIN Bo Chen Department of Biochemistry & Molecular Biology Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology Chaperonin is a class of protein that plays an essential role in protein folding for all cells from bacteria, archaea and eukaryotic cells. Recently it is reported that a type II chaperonin from archaea Methanococcus Marapaludis (Mm-Cpn) could assist refolding of human γD-crystallin to its native conformation. Human γD-crystallin is a lens protein which is associated with the onset of cataract when it partially unfolds. We are interested in understanding the structural mechanism of how type II chaperonin recognizes the γD-crystallin. Here, cryo-electron microscopy (cryo-EM) single particle analysis method was applied to resolve the structure of Mm-Cpn and human γD-crystallin during their initial recognition step. A challenge in studying the chaperonin-substrate structure is the compositional and conformational heterogeneity in the reaction product. We used a multi-model refinement protocol to sort out particle images according to their structural uniformity. Our analysis showed three conformations: 33% of the particles (Subset II) resembled the apo state Mm-Cpn conformation, while 39% of particles (Subset I) had one-ring less open, one-ring open conformation and 28% of the rest particles (subset III) did not yield a reliable map. The control cryo-EM map from the Mn-Cpn showed both ring open which was similar to the map from the subset II of the Mm-Cpn and human γD-crystallin complex. Based on the Statistical variance analysis, we conclude that the subset I corresponds to the Mm-Cpn population with substrate binding. High levels of variances are observed inside the cis-rings. Subsequent symmetry-free reconstructions of subset I particle images converged to one-ring less open and one-ring open conformation. Furthermore, differential conformations of each of the 8 subunits in the less open cisrings were observed to form a tetramer of dimers while both rings in subset II appeared to have a good 8-fold symmetry. Interestingly the 8-fold symmetries on apical domain and equatorial domain are largely broken, while intermediate domain still maintain good 8-fold symmetry. It seems that apical domain and equatorial domain work cooperatively to recognize the unfolded substrate. In conclusion, our results demonstrate the conformational changes and symmetry broken features of type II chaperonin upon binding to γD-crystallin. Contributors: Chen, Bo1; Sergeeva, Oksana2; Goulet, Daniel2; Knee, Kelly2; Joanita, Jakana3; King, Johnathan2; Chiu, Wah1,3* 1: The Verna and Marrs McLean Department of Biochemisty and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA 77030 2: Biology Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, USA 02139 3: National Center for Macromolecular Imaging, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030 41 BAYLOR COLLEGE OF MEDICINE THE ROLE OF TMPRSS9 IN INTELLECTUAL DISABILITY AND AUTISTIC DISORDER Chun-An Chen Department of Molecular & Human Genetics Advisor: Huda Zoghbi, M.D.-Department of Pediatrics Using whole genome sequencing, our lab identified compound heterozygous nonsense mutations in a novel gene, TMPRSS9, in a patient with developmental regression at 2.5 years of age, leading to severe intellectual disability and autism spectrum disorder. TMPRSS9 encodes for polyserase-1, a transmembrane serine protease that is poorly studied. To date, the physiological role of TMPRSS9 is unknown and its endogenous substrates or protein interaction partners have not been identified. Using qPCR analysis, we showed that TMPRSS9 is expressed in various brain regions, including cortex, cerebellum, hippocampus, amygdala, brainstem, and hypothalamus. To test whether loss of function mutations in TMPRSS9 cause developmental regression leading to intellectual disability and autism spectrum disorder, we generated a TMPRSS9 knockout mouse model by removing exon 2 using the Cre/loxP recombination system. Deletion of exon 2 generates a frameshift from exon1 to exon 3, with a premature stop codon in exon 3. A battery of behavioral tests was used to evaluate social approach activity, learning and memory, anxiety-like responses, and motor activity of homozygous knockout mice. The results indicated that knockout mice display increased body weight, impaired social interaction (three-chamber test), reduced motor coordination (rotarod), and abnormal sensorimotor gating (prepulse inhibition). To dissect the physiological function of polyserase-1, we performed immunoprecipitaion experiments to identify protein-protein interaction partners. The serase-1 domain of polyserase-1, tagged with HA and FLAG, was expressed in HEK cells. Subsequently, the purified protein was incubated with mouse brain lysate and subsequently subjected to tandem affinity purification followed by mass spectrometry. Three putative proteinprotein interactors, ATP1A1, SCAMP3, and NCLN, were furthered validated by coimmunoprecipitation studies and colocalization studies of overexpressed proteins in HEK cells. Future work will focus on the identification of the endogenous substrates of polyserase-1. Contributors: CHEN, CHUN-AN; YIN, JIANI; SABO, ANIKO; RICHARD GIBBS, ZOGHBI, HUDA; SCHAAF, CHRISTIAN 42 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF PLIN2 IN ENDOPLASMIC RETICULUM STRESS-INDUCED DIABETES AND PANCREATIC β-CELL Elaine Chen Department of Molecular & Cellular Biology Advisor: Lawrence Chan, D.Sc.-Department of Medicine The endoplasmic reticulum (ER) is an important organelle that performs several critical functions, including the posttranslational modification, folding, and assembly of the newly synthesized proteins. Disruption of these physiological functions leads to accumulation of improperly folded proteins in the ER and the activation of unfolded protein response (UPR). Perilipin 2 (Plin2) is a protein found tightly associated to lipid droplets (LD) and plays an important role in intracellular lipid metabolism. Plin2 is present in multiple tissues, including the liver and the pancreas. We previously found that when we injected tunicamycin (TM), an ER stress inducer, into WT and Plin2-/- mice, the liver of Plin2-/mice recovered faster from TM-induced hepatosteatosis compared with the liver of WT mice. The mechanism whereby Plin2 alleviates the ER stress is not completely understood. In this study, we first investigated whether the deletion of Plin2 has similar protective effect in tissues other than livers. An ER stress-induced diabetic mouse model, Akita mice, was used. Akita mice are heterozygous for a mutation in Ins2 that results in substitution of tyrosine for cysteine, which prevents normal folding and secretion of insulin. The Akita mice exhibit markedly elevated glucose levels and a decline in plasma insulin concentration. Genetic ablation of Plin2 in Akita mice resulted in ameliorated hyperglycemia and increased β cell mass. Mouse islets with Plin2 ablation showed less UPR activation, indicating that deletion of Plin2 also protects pancreatic β cell from ER stress. It is believed that autophagy is activated for cell survival under ER stress. Our data suggest that Plin2 regulates ER stress at least partly by affecting autophagy activity. Contributors: Chen, Elaine; Tsai, Luke; Chang, Benny; Chan, Lawrence 43 BAYLOR COLLEGE OF MEDICINE TRAPPING G PROTEIN-COUPLED RECEPTORS IN ACTIVE SIGNALING STATES THROUGH PROTEIN DESIGN Kuang-Yui Michael Chen Department of Biochemistry & Molecular Biology Advisor: Patrick Barth, Ph.D.-Department of Pharmacology G protein-coupled receptors (GPCRs) are one of the largest families of membrane-embedded receptors that transduce extracellular stimuli into cytoplasmic responses. Increasing evidence from structural, mutational and spectroscopic studies on model GPCRs (e.g. rhodopsin, beta-2-adrenergic receptor) indicate that these receptors signal through long-range conformational changes. However, the atomic-level sequence/structure/energetic relationships governing such allosteric transitions in this large receptor family remain poorly understood, thereby hindering the development of more effective therapeutics (i.e. allosteric regulators) targeting these receptors. We have developed an integrated homology modeling/multistate design/experimental approach to reprogram the signaling properties of structurally uncharacterized GPCRs by redesigning conformational switches in transmembrane (TM) and loop regions. The method was applied to switch the function of the structurally uncharacterized dopamine D2 receptor (DRD2) towards the active state. Inactive and active states of DRD2 were modeled by conformational ensembles using the homology modeling mode of RosettaMembrane. By manipulating the receptor conformational energy landscape and rewiring the networks of energetically coupled residues, multistate design of the DRD2 TM region resulted in receptor variants exhibiting up to an eight-fold increase in basal activity compared to wild type receptor. In agreement with the predictions, biophysical and pharmacological characterizations indicate that the designed receptor variants adopt different local conformations exhibiting various levels of basal and ligand-induced activities. To our knowledge, this is the first computational modeling and design approach that can reprogram the signaling properties of structurally uncharacterized GPCRs by rationally designing novel residue conformational switches and energetic couplings between intra-receptor allosteric regions. Contributors: Chen, Kuang-Yui Michael; Barth, Patrick 44 2014 GRADUATE STUDENT SYMPOSIUM IMPAIRED INTRACELLULAR IRON AND SPHINGOLIPID HOMEOSTASIS LEADS TO NEURODEGENERATION IN A DROSOPHILA MODEL OF FRIEDREICH’S ATAXIA Kuchuan Chen Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics Friedreich’s ataxia (FRDA), the most prevalent form of recessive cerebellar ataxia, is characterized by neurodegeneration and cardiomyopathy. FRDA is caused by mutations in Frataxin (FXN), which is highly conserved across evolution. It has been shown that FXN is localized to the mitochondria, and its deficiency leads to impaired iron-sulfur (Fe-S) cluster biosynthesis and iron accumulation in the mitochondria. However, evidence to support that either or both defects contribute to the pathogenesis of FRDA is still a matter of debate. It is generally believed that increased reactive oxygen species (ROS), which are generated by impaired mitochondria, causes toxicity and lead to degeneration, However, antioxidant therapy only show limited benefits in patients. In a forward genetic screen designed to identify mutations in genes that cause neurodegeneration in Drosophila, we identified the first mutation in frataxin homolog (fh), a Drosophila homolog of mammalian FXN. fh mutants exhibit an age dependent neurodegeneration, and the phenotype can be rescued by human FXN, suggesting human and fly FXN play a conserved role in maintaining neuronal function. Consistent with previous studies, mitochondrial function is impaired in fh mutants, but ROS levels are normal compare to control, and overexpression of ROS scavengers cannot rescue degeneration. Interestingly, both ferrous and ferric irons accumulate in the nervous system and other tissues in these mutants, and reducing iron levels in the food or genetically chelating free irons by overexpressing ferritin can suppress and delay degeneration in fh mutants, suggesting that iron toxicity contributes to neurodegeneration. Furthermore, we observed an accumulation of lipid droplets in the glia cells, and feeding myriocin, a drug that inhibits sphingolipid synthesis, delays the demise of neurons in fh mutants. We argue that accumulated iron triggers impaired sphingolipid homeostasis and leads to neurodegeneration. We are currently investigating this pathway in fh mutants. Contributors: Chen KC; Lin G; Haelterman N; Li Z; Sandoval H; Jaiswal M; Bayat V; Xiong B; Zhang K; David G; Duraine L; Yamamoto S; Charng WL; Graham B; Bellen HJ 45 BAYLOR COLLEGE OF MEDICINE CONTROL OF NEURONAL DEVELOPMENT BY THE RAC-GEFS TIAM1 AND TIAM2 Jinxuan Cheng Department of Biochemistry & Molecular Biology Advisor: Kimberley Tolias, Ph.D.-Department of Neuroscience Neurons undergo a number of highly regulated processes during development that are required to form a functional nervous system, including migration, axonal and dendritic outgrowth, and synapse formation and remodeling. The small Rho-family GTPases Rac1, RhoA and Cdc42 are vital regulators of nervous system development and perturbations in Rho GTPase signaling are associated with neurological diseases, such as Intellectual Disabilities, Amyotrophic Lateral Sclerosis and Autism. Like all GTPases, Rac1 cycles between a GTP-bound activate state and a GDP-bound inactive state. Active Rac1 binds to effector proteins and stimulates signaling pathways that induce actin and microtubule remodeling, which promotes directed cell migration, the growth and branching of dendrites, and the formation and maturation of spines. To function properly, Rac1 activity needs to be tightly regulated in space and time. Rac1 is activated by the guanine nucleotide exchanger factor (GEF) Tiam1 and its closely related homolog Tiam2. Tiam1 (T-lymphoma invasion and metastasis 1) is a large multi-domain protein, which has been shown in vitro to regulate different aspects of neuron development. Tiam2 (STEF, SIF and Tiam1-like exchange factor) is a closely related homolog of Tiam1. Tiam2 is required for neurite outgrowth and growth cone morphology in cultured hippocampal neurons. Furthermore, Tiam1 and Tiam2 are both components of the evolutionary conserved PAR polarity complex (Par3, Par6, aPKC), which is essential for establishing and maintaining cell polarity during processes such as directed cell migration, axon initiation, and synapses formation. The expression levels of Tiam1 and Tiam2 are high in the brain during development, and remain high in some regions that undergo plasticity. Tiam2, which is known to regulate cell migration in cell culture, is highly expressed in the cortex during embryogenesis when neuronal migration happens. This evidence suggests that Tiam1 and Tiam2 may play important roles in nervous system development. However, most of the studies investigating Tiam1 and Tiam2 have been done in vitro, and little is known about their roles in vivo. Here we describe the generation of Tiam1 and Tiam2 conditional knockout mice and our attempts to use these mice to determine the roles of Tiam1 and Tiam2 in neuronal migration and synapse development in vivo. Our preliminary data indicate that Tiam1 and Tiam2 knockout mice show decreased spine density, defects in astrocytes and interneuron migration. My future work will be using in utero electroporation to study neuron migration in the Tiam1 and Tiam2 knockout mice, and characterize the role of Tiam1 and Tiam2 in regulating these processes. Contributors: Cheng, Jinxuan; Niu, Sanyong; Firozi, Karen; Tolias, Kimberley 46 2014 GRADUATE STUDENT SYMPOSIUM TESTING THE ROLE OF MICROGLIA IN ANTIBODY-MEDIATED TREATMENT OF ALZHEIMER’S DISEASE Angie Chi An Chiang Department of Neuroscience Advisor: Joanna Jankowsky, Ph.D.-Department of Neuroscience New therapies for Alzheimer’s disease directly target amyloid-β (Aβ), the main component of pathological plaques. Immunization with an anti-Aβ antibody (clone Ab9) combined with Aβ suppression significantly reduces amyloid load. However, the mechanism by which combination therapy works remains unclear. Optimizing the outcome of immunotherapy requires understanding how antibody treatment lowers amyloid burden. To address this, we are testing whether microglia, the resident phagocytes of the brain, are necessary for antibody-mediated plaque clearance. We are crossing CD11b-HSVTK mice, a mouse model in which microglia can be selectively ablated from the brain with the administration of ganciclovir, to tet-off APP double transgenic mice, a mouse model for Alzheimer’s amyloidosis that allows for temporal control of the transgenic expression of APP. By using the tet-off system to suppress transgenic APP and Aβ synthesis during administration of an anti-Aβ antibody, microglia are allowed to work from a static amyloid plaque load. Following treatment with ganciclovir, microglia cells will induce cell death. Histological assessment of amyloid burden and microglial ablation will be used for quantification. By understanding the role of microglia in amyloid clearance, the results of this study may be useful for the development of better therapies and the avoidance of harmful consequences. Contributors: Chiang, Angie CA ; Jankowsky, Joanna L. 47 BAYLOR COLLEGE OF MEDICINE THE ROLE OF SRC-2 IN THE ANTI-DIABETIC AND ANTI-LIPOGENIC EFFECTS OF LRH-1 Sungwoo Choi Program in Developmental Biology Advisor: David Moore, Ph.D.-Department of Molecular & Cellular Biology Type 2 diabetes is a metabolic disease characterized by a high level of blood glucose due to insulin insensitivity. This disorder is tightly correlated with increased fat accumulation in the liver. It has been proposed that a vicious cycle of increased fat accumulation and insulin insensitivity drives the early stages of this disease. In the forward direction of this cycle serum insulin levels rise in response to obesity. The elevated insulin results in increased lipogenesis and lipid deposition in the liver, which drives further whole body insulin resistance. This in turn results in even higher serum insulin levels and further liver fat accumulation. Several nuclear receptors are able to reverse this cycle by directly repressing lipogenesis in the liver. In this case, nuclear receptor activation suppresses liver fat accumulation and thereby promotes whole body insulin sensitivity. The resulting decrease in serum insulin feeds forward to further decrease liver fat accumulation, with the net result of reversing hepatic steatosis and insulin resistance, even in the absence of effects on overall body weight. My project focuses on Liver Receptor Homolog-1 (LRH-1), which can be activated by Dilauroyl Phosphatidylcholine (DLPC). The Moore lab has linked the ability of this LRH-1 agonist to reverse insulin resistance in mouse models of type 2 diabetes to decreased expression of the key lipogenic transcription factor SREBP-1. But the specific mechanisms involved remain unknown. My goal is to understand the mechasnism of action of the anti-diabetic effects of DLPC. The transcriptional effects of nuclear receptors are mediated by coactivators and corepressors, and recent results from the O’Malley laboratory have identified the coactivator Streroid Receptor Complex-2 (SRC-2) as a key metabolic regulator (PMIDs 21195347, 19039140). Additional preliminary results show that SRC-2 can coactivate LRH-1 transactivation, and indicate extensive overlap between genome wide LRH-1 and SRC-2 binding sites in the liver. In vivo, there are also phenotypic similarities between SRC-2 binding sites in the liver. In vivo, there are also phenotypic similarities between SRC-2 liver specific knockouts and LRH-1 liver specific knockouts in terms of lipid metabolism and bile acid metabolism. Thus, my specific hypothesis is that SRC-2 is an essential mediator of the anti-diabetic and anti-lipogenic effects of DLPC. I am now trying to determine the impact of both acute and chronic DLPC treatments in SRC-2 liver specific knockout mice and the littermate controls. Contributors: Choi,Sungwoo; Kim,Mi-Sun; Kim,Seung-Whan; Lee,Jae Man; Moore,David 48 2014 GRADUATE STUDENT SYMPOSIUM LIPID BIOMARKER OF NEURAL STEM AND PROGENITOR CELLS AFFECTS NR2E1 (TLX) TRANSCRIPTIONAL ACTIVITY William Tin-Shing Choi Program in Developmental Biology/M.D.-Ph.D. Program Advisor: Mirjana Maletic-Savatic, M.D./Ph.D.-Department of Pediatrics The fate of the cell, whether it divides, differentiates, enters a transient (quiescence) or permanent (senescence) growth arrest, or triggers a suicidal mechanism of death, demands a finely-tuned sequential activation and deactivation of both biosynthetic and energy generating metabolic pathways. Lipid molecules are important regulators of stem cell proliferation, including neural stem and progenitor cells (NPCs). Our previous study of NPCs using proton magnetic resonance spectroscopy (H-NMR) showed an enrichment of molecule(s) that resonate at 1.28-ppm, characteristic of –CH2– methylene lipid moiety. The amplitude of the 1.28-ppm signal correlated with NPC proliferation both in vitro and in vivo, suggesting that the respective molecule(s) are associated with NPC fate. Further investigations of the chemical composition of these molecules identified them as a specific group of lipids, some of which were unique to NPCs as compared to astrocytes. We then asked if these molecules were functionally important for NPCs, specifically as signaling molecules regulating NPC cell cycle. Lipid modifiers are known ligands for nuclear receptors, transcription factors which can be modulated by ligand binding. Transcript analysis of 48 known nuclear receptors found 25 to be expressed in NPCs, 11 of which were orphan receptors with unknown ligands. Interestingly, 3D-reconstruction and structure comparison of known ligand—nuclear receptor combinations predicted a high probable interaction of the 1.28-ppm molecule(s) with the ligand-binding domain of six orphan nuclear receptors expressed in NPCs. One of those was Nr2e1 (also known as Tlx), already known to be important for NPC proliferation and self-renewal. Interestingly, transcriptional activity of Nr2e1 was altered when it was exposed to molecules that resonate at 1.28-ppm, as shown by mammalian one-hybrid luciferase assay. Altogether, these data suggest that the NPC fate may involve the interaction of Nr2e1 and the endogenous lipid molecules enriched in them, thus illuminating a possible mechanism controlling NPC behavior. Contributors: Choi, William; Ma Lihua, Thakkar, Aarohi; Gopakuma, Sricharan; Cerda-Smith, Christian; Young, Damian; MacKenzie, Kevin; Maletić-Savatić, Mirjana 49 BAYLOR COLLEGE OF MEDICINE THE ROLE OF CUL5 IN MODULATING DRUG SENSITIVITY IN TRIPLE-NEGATIVE BREAST CANCER Hsiang-Ching Chung Integrative Program in Molecular and Biomedical Sciences Advisor: Thomas Westbrook, Ph.D.-Department of Biochemistry & Molecular Biology Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is recalcitrant to target therapies. Previous studies in our lab uncovered the codependency of TNBC survival and progression on multiple receptor tyrosine kinases (RTKs) including MET and PDGFRβ. Notably, combined inhibition of these RTKs with tyrosine kinase inhibitors (TKis), sunitinib and crizotinib (or S+C) effectively suppresses TNBC tumor growth across many independent patient-derived xenograft (PDX) models of TNBC in vivo. However, not all TNBC PDX models are sensitive to S+C.(unpublished data) To better understand the molecular determinants of S+C sensitivity and resistance, we performed a genetic (RNAi) screen for modifiers of S+C response in multiple TNBC models. CUL5 was a prominent candidate from these multiple genetic screens. We validated that knockdown of CUL5 greatly enhances cell fitness upon S+C treatment, suggesting CUL5 function is a critical determinant of TNBC drug sensitivity. It is known that CUL5 functions as a scaffold protein for cullin-based ubiquitin E3 ligases, however, the role(s) of CUL5 E3 ligases in TNBC pathogenesis and modulating drug resistance has not been explored (especially resistance to tyrosine kinase inhibitors, the largest class of anti-cancer targeted therapies). The characterization of how CUL5 controls TNBC drug response represents an unprecedented opportunity to investigate the role of ubiquitylation in modulating drug response. Identifying genetic or epigenetic determinants of resistance may enable us to predict resistance mechanisms to S+C in TNBC patients, and will allow for improved efficacy of target therapies. More broadly, these discoveries may provide insights or principles governing patient response to other tyrosine kinase inhibitors (TKIs) used in other cancers. Contributors: Sun, Tingting; Nair, Amritha; Thomas Westbrook 50 2014 GRADUATE STUDENT SYMPOSIUM ROLE OF ANDROGEN RECEPTOR IN HORMONE RESISTANCE OF ESTROGEN RECEPTOR ALPHA-POSITIVE BREAST CANCER Andrew Mark Robinson Ciupek Program in Translational Biology & Molecular Medicine Advisor: Suzanne Fuqua, Ph.D.-Department of Medicine Mothaffar Rimawi, M.D.-Department of Medicine Background: We have previously developed a novel model of hormone therapy resistance involving shRNA knockdown of Rho GDI(. Although it is known that androgen receptor (AR) is frequently expressed along with estrogen receptor (ER) ( in breast tumors, AR’s role in resistance is unknown. Here we explore the role of AR in the resistance of the Rho GDI( knockdown model. Materials and Methods: shRNA knock down was used to block expression of Rho GDI( in the ER(-positive breast cancer cell lines MCF-7 and ZR-75-B. Reverse phase protein arrays (RPPA) were used to analyze global changes in growth factor pathway expression. Microarray analysis was used with tamoxifen (Tam) treated cells to explore pathways promoting Tam resistance. Western blot analysis was used to look at activation of the AR, ER, and epidermal growth factor receptor (EGFR) pathways. The effects of AR and EGFR inhibitors and agonists on anchorage independent growth and proliferation in the presence of Tam were measured in soft agar colony formation assays and MTT growth assays. Results: MCF-7 Rho GDI( knockdown cells were resistant to the growth inhibitory effects of Tam when grown as xenografts in vivo. We discovered that AR was over expressed in the knockdown cells as confirmed by both western blot and RPPA. Microarray analysis showed an activation of EGFR and MAPK signaling in Tam treated ZR-75-B knockdown cells. In addition, western blot analysis showed that rapid activation of EGFR in these cells could be blocked by both AR and EGFR inhibitors. In MCF-7 and ZR-75-B knockdown cells, increased basal levels of anchorage independent growth, proliferation, and Tam stimulated growth were seen which could be blocked by inhibition of AR with bicalutimide and MDV3100 or the EGFR inhibitors AG1478 and Gefitinib. Discussion: Since AR was significantly over expressed with the Tam-resistant phenotype and AR agonists/inhibitors modulated the phenotype, we hypothesize that AR plays a role in the process. Also, given that EGFR is activated and EGFR inhibitors block the Tam-resistant phenotype, EGFR also likely contributes to the resistance. Since an AR inhibitor can block rapid activation of EGFR we hypothesize that AR may promote Tam-resistance through activation of EGFR signaling. Our results suggest that AR may represent a new clinical target for hormone therapy resistance of ER( positive breast cancer. Contributors: Ciupek A, Brusco L, Covington KR, Weigel NL, Lu Y, Mills G, and Fuqua AW 51 BAYLOR COLLEGE OF MEDICINE SPATIAL MEMORY ENCODING IN A MOUSE MODEL OF TAU-MEDIATED NEURODEGENERATION Sarah Margaret Ciupek Department of Neuroscience Advisor: Daoyun Ji, Ph.D.-Department of Molecular & Cellular Biology Improper aggregation of tau protein occurs in many neurodegenerative dementias, including Alzheimer’s disease. A major target of degeneration in these tauopathies is the hippocampus, a classical brain region for memory. In the normal hippocampus, spatial memory is encoded by place cells, a class of pyramidal neurons that fire preferentially when the animal is in a specific location in space. Additionally, proper memory encoding is facilitated by stereotyped oscillations in the local field potential (LFP) and entrainment of neuronal firing to those oscillations. However, we do not know which aspects of hippocampal memory processing are altered in tauopathies and when in disease alterations occur. Such knowledge would bridge our understanding of the molecular and behavioral level changes that occur in these disease states. Here, we study this question using the rTg4510 (Tau) mouse line, in which human tau with the P301L mutation found in patients with frontotemporal dementia is expressed in the mature forebrain. In this mouse, tau becomes hyperphosphorylated and begins to aggregate in the hippocampus as young as 2.5 months of age. Neurodegeneration begins soon after, with 60% of the CA1 layer of the hippocampus already degenerated by 5.5 months of age. Previous experiments in our laboratory have shown deficits in place cell stability in older Tau mice (age 7-9 months) that already exhibit profound neurodegeneration. These older mice also show altered LFP oscillations as well as impaired firing synchrony during LFP events. However, this previous study only looked at older animals, when the effects of neurodegeneration cannot be distinguished from those of tau phosphorylation alone. To test which, if any, of these network level changes occur prior to neurodegeneration, we have recorded in vivo from the hippocampus of Tau mice at 2 to 5 months of age as they run through a linear track, explore an open field, and sleep. Our results show only minor deficits in place cell stability and size compared to WT mice prior to major degeneration. In contrast, these young mice do exhibit LFP and synchrony changes. Specifically, there is a decrease in the amplitude of sharp-wave ripples (high-frequency LFP oscillations associated with memory consolidation) during slow-wave sleep in Tau mice and a corresponding unusual pattern of cell firing during ripple events. When graphed by age, the ripple amplitude phenotype gets progressively worse with age in Tau mice but stays constant in WT mice, with a starting average ripple amplitude higher than even the youngest Tau mice. These results allow us an insight into the network level alterations that lead to the debilitating memory loss symptoms seen in patients with various types of tauopathies, an aspect of these diseases that is poorly understood. Contributors: Cheng, Jingheng; Ji, Daoyun 52 2014 GRADUATE STUDENT SYMPOSIUM DELIVERY OF A GENETIC CANCER VACCINE CONTAINING A SMALL MOLECULE INDUCIBLE IMMUNE ADJUVANT BY IN VIVO ELECTROPORATION (EVACIDE) Matthew R Collinson-Pautz Program in Translational Biology & Molecular Medicine Advisor: David Spencer, Ph.D.-Department of Pathology & Immunology Kevin Slawin, M.D.-Department of Urology The use of dendritic cell vaccines to treat cancer has significant potential, however that potential has yet to be achieved in treating cancer patients. Fortunately, pioneering biotechnologies are paving the way for more effective cancer vaccines. Our lab has developed a drug inducible MyD88/CD40 (iMC) composite adjuvant that promotes robust cytotoxic T cell priming by iMC activated DCs. Despite the iMC innovation, its implementation in patient-tailored ex vivo DC vaccines is impractical for widespread use, due to issues of scalability, distribution, and cost. We have therefore begun to address the delivery of DC vaccines as “off-the-shelf” therapies using in vivo electroporation (EP) of plasmid DNA (pDNA) encoding both the iMC adjuvant and tumor antigen (eVaCIDe). Intradermal EP of pDNA encoding the model antigen LacZ in mice primed LacZ-specific CD8+ T cell responses, as mice receiving LacZ + EP had significantly greater portions of antigen responsive, IFNγ secreting T cells than mice that received that same LacZ vector without EP. Mice vaccinated prophylactically with LacZ +EP were either protected from establishment of B16/LacZ tumors or demonstrated significantly slower tumor growth compared to controls. In mice bearing pre-established B16/LacZ tumors, LacZ-eVaCIDe, but not EP with LacZ antigen alone significantly reduced tumor burden compared to negative controls. Lastly, eVac encoding the human Prostate cancer antigen, PSMA, was able to stimulate significant antigen-specific CD8+ T cell and serum antibody responses. Taken together, these data indicate that eVac with iMC-antigen is an effective “off-the-shelf” cancer vaccine and potentially a platform for a wide range of disease targets. These findings warrant elucidation of the underlying immunological mechanisms responsible for the anti-tumor responses observed with eVac treatment to guide further refinement of the vaccine in a pre-clinical model. Contributors: Collinson-Pautz, M; Lu, An; Decker, W; Spencer, D; Levitt, J 53 BAYLOR COLLEGE OF MEDICINE GENE DELETION SCREENING SUGGESTSCANDIDATE GENE APPROACHES REVEAL NEW TARGET PATHWAYS FOR TREATING ANTIBIOTIC-RESISTANT GRAM-NEGATIVE BACTERIAL INFECTIONS Zachary Christopher Conley Department of Biochemistry & Molecular Biology Advisor: E. Zechiedrich, Ph.D.-Department of Molecular Virology & Microbiology New antibiotics are needed because resistance has rendered many existing drugs ineffective. We discovered that the antifungal drug, ciclopirox, prevents growth of problematic, multidrug-resistant clinical isolates, although the drug target remains elusive (Carlson-Banning et al. 2013 PLoS One 8:e69646). We showed that both sugar metabolism and the availability of free iron in the growth medium affect ciclopirox inhibition in of E. coli. Additionally, iron-acquiring siderophore production in P. aeruginosa was increased in the presence of ciclopirox, implying a disruption of iron acquisition. To examine which We performed a literature search and identified 103 genes involved in specific iron-utilization and sugar metabolism pathways. To test mightfor those that are be affected by ciclopirox, we we screened gene deletion strains 103 gene deletion strains for increased sensitivity to ciclopirox, . Hhoping to identify ciclopirox-specific targets, . Towe remove filtered out general antibiotic effects, we by comparingred our data to a study that measured the antibiotic susceptibility of 22 antibiotics forof the entire Keio collection of E. coli gene deletions and removed from our list any that were affected by any antibiotic (Liu et. al. 2010 Antimicrob. Agents and Chemother. 54:139310.1128/AAC.00906-09vol, p). To remove filter out genes involved in general iron chelation affect, as ciclopirox can bind free iron, we screeneding our 29(how many?) gene sets for increased sensitivity to the known iron chelator 1, 10-phenanthroline. This stratification resulted in 18 gene deletions with increased susceptibility to ciclopirox alone. The majority Most (n) of these genes encode proteins identified are involved with the synthesis of the surface glycolipid enterobacterial common antigen. Additionally,In support of the idea that ciclopirox affects cell outer membrane, we had already shown that O-antigen producing bacteriaE. coli grown in sub-inhibitory concentrations of ciclopirox display less O-antigen and less high molecular weight lipopolysaccharide (CarlsonBanning et al. 2013 PLoS One 8:e69646). We hypothesize that one ciclopirox target exists in the construction of glycolipids that are attached to the bacterial outer membrane. The next largest group ofOther stratified genes identified isencode proteins involved in the uptake of the siderophore enterobactin, a gram-negative-specific siderophore with the highest known iron binding affinity of any siderophore. We hypothesize that ciclopirox drug targets are not restricted to one pathway. Here, we identified the construction of glycolipids that are attached to the bacterial outer membrane as one pathway, and iron uptake through siderophores ais another pathway affected by ciclopirox. Perhaps the existence of multiple essential targeted pathways explains why this drug is effective against antibiotic-resistant bacteria. These data are an important step toward developing ciclopirox or new derivatives of ciclopirox, as drugs against multidrug-resistant gram-negative pathogens for which few therapeutic options exist currently.. Contributors: Conley, Zachary C.; Carlson-Banning, Kimberly M.; Carter, Ashley; Chou, Andrew; Hamill, Richard J.; Song, Yongcheng; and Zechiedrich, Lynn 54 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF DNMT3A IN MAINTAINING DNA METHYLATION IN STEM CELL POPULATIONS Sean Michael Cullen Program in Developmental Biology/M.D.-Ph.D. Program Advisor: Margaret Goodell, Ph.D.-Department of Pediatrics Our lab recently published the first hematopoietic stem cell (HSC) methylome. One feature identified was over 1,000 large, unmethylated regions, named “canyons” (>3.5kb, <10% average methylation, >5 CpG dinucleotides/kb). These canyons were largely conserved across cell type and species, but were also observed to possess subtle, dynamic differences between these groups. For example, in mouse embryonic stem cells (ESCs), 839 canyons were identified, 82% of which were shared with HSCs, but differed slightly overall in size and methylation levels. As for the purpose of these canyons, a high number and concentration of HSC-specific TF binding sites were found within the confines of these canyons. Deletion of DNA methyltransferase 3a (Dnmt3a), previously shown to be essential for proper HSC differentiation (Challen 2012), was found to change methylation significantly at canyon borders, but, unexpectedly, both expanded and contracted the size of many HSC canyons (Jeong 2013). This suggests Dnmt3a functions to maintain methylation at canyon borders, which leads to our central hypothesis that direct Dnmt3a binding at canyon edges is essential for canyon stability. Using an ESC line capable of doxycycline-induced expression of biotinylatedDnmt3a, we performed Chromatin Immunoprecipitation-Sequencing (ChIP-Seq) to identify the genome-wide binding sites of Dnmt3a. Dnmt3a binding was largely excluded from ESC canyons, but was found at the edges of these canyons, often at high levels. The cell line was then used to generate a mouse model capable of examining the similarities and differences of Dnmt3a DNA-binding in HSCs and ESCs, to determine how accurately binding differences mimic canyon changes between stem cell populations. Contributors: Cullen, Sean; Luo, Min; Jeong, Mira; Lin, Xueqiu; Li, Wei; Goodell, Margaret 55 BAYLOR COLLEGE OF MEDICINE CLINICAL IMPLICATIONS AND FREQUENCY OF VARIANTS IN NONSYNDROMIC HEARING IMPAIRMENT GENES IN A POPULATION-BASED SAMPLE OF AFRICAN-AMERICANS AND EUROPEAN-AMERICANS Hang Dai Department of Molecular & Human Genetics Advisor: Suzanne Leal, Ph.D.-Department of Molecular & Human Genetics A large number of studies on nonsyndromic hearing impairment (NSHI) have been performed using samples collected from the Indian subcontinent, Middle East and Europe. There has been limited study of NSHI in African-Americans (AA) and subSaharan Africans. We evaluated the frequency of previously reported “pathogenic” variants in NSHI genes using data from the NHLBI-Exome Sequencing Project (ESP) which is a population-based study of AA (N=2203) and European-Americans [EA (N=4300)]. Out of 201 observed variant sites which are pathogenic according to ClinVar and/or the Deafness Variation Database, we reclassified 121 (60.2%) variant sites as likely non-pathogenic based on literature, high allele frequencies in ESP and bioinformatics tools. In ESP, 80 likely pathogenic variant sites were observed in 5 autosomal dominant (AD) and 14 autosomal recessive (AR) NSHI genes of which 24 of the variant sites in 7 of these genes cause syndromic hearing impairment (HI), e.g. MYO7A variants cause both NSHI and Usher syndrome. Of these variant sites, 49 (91 alleles) were found only in EA, 18 (23 alleles) only in AA, and 13 (194 alleles) in both EA and AA. GJB2 c.35delG was the ARNSHI variant site with the highest allele frequency in EA [N=89; 1.09% (95%CI: 0.88%, 1.34%)] but with a much lower allele frequency in AA [N=4; 0.094% (95%CI: 0.03%, 0.2%)]. Additionally OTOF c.2348delG was identified in 4 AA alleles [0.095% (95%CI: 0.03%, 0.2%)] but is very rare in EA [N=1; 0.01% (95%CI: 0.0003%, 0.07%)]. For EA, pathogenic variant sites were identified in 11 AR and 5 AD NSHI genes including variant sites in 7 NSHI genes which cause syndromic HI, while for AA, pathogenic variant sites were identified in 13 AR and 2 AD NSHI genes, of which 6 genes have variant sites which cause syndromic HI. MYO7A c.3764delA which was reported to cause Usher syndrome, is homozygous in 1 AA and 3 EA individuals. The use of EVS in order to exclude nonpathogenic variants must be done cautiously due to NSHI-causal variants within EVS. Knowledge of the frequency of NSHI variants in large population samples from different ethnic backgrounds is not only important to evaluate clinical significance but also aids in evaluating pathogenicity. Although population-based samples of AA aids in evaluating population-specific frequency of previously reported pathogenic variants, AA and sub-Saharan Africans with HI need to be studied to better understand the genes and variants underlying disease etiology. Contributors: Dai, Hang; Wang, Gao; Santos-Cortez, Regie; Leal, Suzanne 56 2014 GRADUATE STUDENT SYMPOSIUM ELUCIDATING THE INTERACTIONS OF MUTANT HUNTINGTIN WITH THE TRiCLIKE CCT5 COMPLEX Michele C. Darrow Department of Biochemistry & Molecular Biology Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology Huntington’s disease, a neurodegenerative disorder characterized by loss of striatal neurons, is linked to an expanded and unstable CAG trinucleotide repeat, which translates as a polyglutamine repeat in the protein product. 40+ trinucleotide repeats have been described as pathological. Mutant huntingtin exon 1 with 46 polyglutamine repeats was purified via thioredoxin tag and incubated with the TRiC chaperonin subunit CCT5 homo-oligomer complex, markedly slowing mHtt fibril aggregation, as shown using a filter trap assay and cryo-electron microscopy. Cryo-electron tomography and averaging was also performed on these samples with the goal of identifying the structure of CCT5 homo-oligomer in complex with mHtt fibrils and oligomers. Results indicate that CCT5 homo-oligomer interacts with mHtt at the tips of the fibrils, and by encapsulating oligomers. This study provides further information about how a single subunit of TRiC chaperonin interacts with mHtt fibrils and oligomers to slow aggregation, identifying a potential target for therapeutics. Contributors: Sergeeva, Oksana A.; Isas, Jose M.; Galaz-Montoya, Jesus; King, Jonathan A.; Langen, Ralf; Chiu, Wah 57 BAYLOR COLLEGE OF MEDICINE WACKY IS A NOVEL REGULATOR OF THE MTOR PATHWAY Gabriela Riva David Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics The mTOR signaling pathway senses and integrates environmental signals, cellular nutrition, and energy status to regulate cell growth and metabolism. This pathway plays a role in many major cellular processes, and its dysregulation has been implicated in diseases that include neurodegeneration and cancer. Through a forward genetic screen on the Drosophila X chromosome for essential genes that play a role in the development, function, and maintenance of the nervous system, we have isolated a novel component of the mTOR pathway: the Drosophila homolog of WW domain-containing adapter with coiled-coil region (WAC), which we have named Wacky. wacky loss-of-function (LOF) mutant eye clones exhibit functional defects and morphological degeneration in the adult retina. Looking at the subcellular defects of wacky LOF mutant clones in the eye and fat body, we observe increase in autophagic vesicles and lysosomes. Since autophagy is misregulated in wacky mutant cells, we looked at the activity level of mTOR, which negatively regulates autophagy. We found that, in wacky mutant larval fat bodies, the phosphorylation levels of downstream mTOR kinase targets, S6K, 4E-BP, and Akt, are dramatically decreased. Consistent with an increase in basal autophagy that results from a decrease in mTOR activity, knocking down WAC in HEK293 cells leads to an increase in the degradation of betaine homocysteine methyltransferase, a substrate of autophagy. To determine how loss of Wacky leads to a decrease in mTOR signaling, we performed immunoprecipitation followed by mass spectrometry to identify protein interactors of Wacky. From this experiment, we identified four mTOR pathway components as Wacky physical interactors: mTOR, Vha100-2, Pontin, and Reptin. Currently, we are focusing on dissecting the interaction of Wacky with these mTOR pathway components to determine the mechanism by which Wacky positively regulates this signaling pathway. Because of its role in the mTOR pathway, Wacky could be a novel target for the modulation of the mTOR pathway, and further deciphering its functions may lead to the development of effective therapies for neurodegeneration and cancer. Contributors: David, Gabriela; Xu, Zhen; Rui, Yan-Ning; Charng, Wu-Lin; Jaiswal, Manish; Yamamoto, Shinya; Zhang, Ke; Xiong, Bo; Bayat, Vafa; Duraine, Lita; Zhang, Sheng; Bellen, Hugo 58 2014 GRADUATE STUDENT SYMPOSIUM COMPLEMENTARY GENETIC APPROACHES TO ELUCIDATE MOLECULAR MECHANISMS REGULATING AGGRESSION IN DROSOPHILA Shaun Michael Davis Department of Molecular & Human Genetics Advisor: Herman Dierick, M.D.-Department of Molecular & Human Genetics Aggressive behavior is widespread in the animal kingdom. However, its mechanisms remain poorly understood, and the degree of molecular conservation between distantly related species is unknown. Here, we used two complementary genetic approaches to identify the mechanisms that regulate this behavior. In mice, loss-of-function of the transcriptional repressor Nr2e1 causes extreme aggression. We used a candidate gene approach and show that knock-down of tailless (tll), a fly ortholog of Nr2e1, increases aggression in Drosophila melanogaster. Tll localizes to the adult pars intercerebralis (PI), which is composed of neurosecretory cells with similarity to the mammalian hypothalamus. Knock-down of tll in the PI is sufficient to increase aggression and is rescued by co-expressing human NR2E1. Knock-down of atrophin, which encodes a co-repressor of Tll, also increases aggression and both proteins physically interact in the PI. The tll-knock-down induced aggression phenotype is fully suppressed by blocking neuropeptide processing or release from the PI, showing that aggression is dependent upon a neuropeptide-based mechanism. In addition, genetically activating PI neurons, which leads to neuropeptide release in flies, increases aggression and mimics the aggression-inducing effect of hypothalamic stimulation in mammals. Together our results show that Tll regulates aggression in flies through a neuropeptide-based mechanism in the neurosecretory cells of the adult PI. In addition to the candidate gene approach, we developed a forward genetic screen to identify novel genes that regulate aggressive behavior. One consequence of excessive aggression is increased physical damage. We found a positive correlation between number of lunges and percentage of wings with damage. Wild-type male flies were mutagenized with EMS to perform an X-chromosome forward genetic screen. Over 1,500 different chromosomes were analyzed; the average percent wing damage was 7% and only lines with more than 30% damaged wings were kept for further analysis. Of these 42 lines, 5 showed an increased aggression phenotype. These lines are now being sequenced to identify the causal mutation. This unbiased approach can identify novel components and/or pathways that regulate aggression behavior in Drosophila. Much like the tll-regulated neuropeptide-based mechanism, these results can provide insight into human aggression. Contributors: Thomas, Amanda; Nomie, Krystle; Gnerer, Joshua; Huang, Longwen 59 BAYLOR COLLEGE OF MEDICINE A CRITICAL ROLE FOR CYCLIN D1 IN TESTICULAR GERM CELL TUMORIGENESIS Emily Packard Dawson Integrative Program in Molecular and Biomedical Sciences Advisor: Jason Heaney, Ph.D.-Department of Molecular & Human Genetics Testicular germ cell tumors (TGCTs) are the most frequent tumor type diagnosed in young men. TGCTs arise from fetal germ cells whose pluripotent capacities facilitate tumor differentiation into a variety of somatic cell types. In the 129 inbred mouse model of human TGCTs, tumors initiate between embryonic days (E)13.5 to 15.5, which coincides with the mitotic:meiotic switch: germ cells of both sexes lose pluripotent capacity, female germ cells (oogonia) commit to meiosis, and male germ cells (gonocytes) enter mitotic arrest. We previously demonstrated that gonocyte proliferation, retention of pluripotency, and aberrant expression of genes associated with pre-meiotic oogonia and adult spermatogonia, including cyclin D1 (Ccnd1) and stimulated by retinoic acid 8 (Stra8), at E15.5 were directly related with increased tumor risk. Based on the known oncogenic potential of Ccnd1 overexpression, we hypothesized that aberrant expression of Ccnd1 significantly contributes to TGCT initiation by inducing a breakdown in the mitotic:meiotic switch, which causes gonocytes to continue proliferating and retain pluripotency. Using a Ccnd1 knockout on the high TGCT risk 129-Chr19MOLF/Ei (M19) background, we found that Ccnd1 deficiency significantly reduced TGCT incidence by 62% (p<0.001) compared to wild-type and heterozygous animals. Immunohistochemistry experiments suggest that Ccnd1 deficiency reduces the number of proliferating (KI67-positive) and pluripotent (NANOGpositive) gonocytes. Ccnd1 expression in post-migratory embryonic germ cells is normally restricted to oogonia and is transiently expressed from E12.5 to E15.5, prior to their entry into meiosis. Expression of Ccnd1 from E12.5 to E15.5 in TGCT-susceptible gonocytes suggests that a signal normally restricted to the developing ovary is aberrantly active in the TGCT-susceptible testis. Retinoic acid (RA) signaling normally precedes Ccnd1 expression in embryonic oogonia, while RA is normally catabolized by CYP26B1 in the embryonic testis. Curiously, we find an altered expression pattern of embryonic germ cell sex-specific genes (e.g. Stra8, Ccnd1, Nanos2, and Nodal) in TGCT-susceptible gonocytes. Therefore, we tested whether gonocyte expression of Ccnd1 is induced by an ectopic RA signal. Culturing embryonic testes from TGCTresistant or TGCT-susceptible mice in medium containing RA induced CCND1 expression in gonocytes. Furthermore, induction of CCND1 in oogonia and TGCTsusceptible gonocytes deficient for Stra8 demonstrates that RA-induced expression is independent of Stra8 function. We hypothesize that TGCT-susceptible gonocytes respond to both an abnormal RA signal and normal gonocyte developmental signals, which disrupt gonocyte entry into mitotic arrest during the mitotic:meiotic switch and lead to TGCT initiation. Contributors: Dawson, Emily; Lanza, Denise; Benton, Susan; Heaney, Jason. 60 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF THE ESSENTIAL PROTEIN RBM17 AND ITS IMPLICATION IN SPINOCEREBELLAR ATAXIA TYPE1 (SCA1) PATHOGENESIS Antonia De Maio Program in Developmental Biology Advisor: Huda Zoghbi, M.D.-Department of Pediatrics Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disorder characterized by death of Purkinje cell and brain stem neurons. SCA1 is caused by expansion of CAG repeats within the coding region of the Ataxin1 gene (Atxn1), leading to the production of a mutant variant of the cognate protein harboring an expanded glutamine tract (PolyQ). Previous work in our lab defined the spliceosomal RNA binding protein, RBM17, as an ATXN1 interactor that preferentially binds upon expansion of the polyQ tract and requires phosphorylation of the ATXN1 residue S776. These two aspects of ATXN1 are both critical in the SCA1 pathogenesis suggesting that RBM17 has a key role in the disease mechanism. To investigate in depth the function of Rbm17 we generated an Rbm17 constitutive knockout and found this protein to have an essential developmental role, as its constitutive ablation causes early embryonic lethality. To evaluate the effects of Rbm17 loss-of-function we looked at conditional deletion models lacking Rbm17 exclusively from developing cerebellum or adult Purkinje cells. These mice revealed that absence of Rbm17 severely impairs development of cerebellum while Purkinje cells devoid of the protein undergo progressive and fast degeneration. Having established the critical role of Rbm17 for development and cell viability we propose that this factor is a key component of the spliceosomal complex and that its ablation causes aberrant splicing of still unknown RNA targets whose tight regulation is essential for successful accomplishment of both processes. To test our hypothesis we will use Immunoprecipitation/mass spectrometry (IP/MS) and Crosslinking and immunoprecipitation (CLIP) methods to, respectively, identify Rbm17 protein interactors and its RNA binding pattern throughout the transcriptome. Using MS we analyzed the composition of Rbm17 immunoprecipitation (IP) samples from wild-type mouse cerebellum. Our preliminary results confirmed some previously known interactions and pointed out several putative Rbm17 partners within and beyond the spliceosomal complex, including splicing factors and RNA metabolism modulators. Currently, we are optimizing the CLIP protocol for Rbm17 in order to pull down the protein and its associated RNA targets. RNA-sequencing will eventually reveal the identity of these targets, shedding light on the essential cellular network regulated by Rbm17. The integration of these data will permit us to uncover the molecular mechanism underlying the requirement of Rbm17 for cell survival and will help us establish if and how Rbm17 contributes to SCA1 pathogenesis. Contributors: De Maio, Antonia; Park, Jeehye; Zoghbi, Huda Y. 61 BAYLOR COLLEGE OF MEDICINE THE ROLE OF INTERNAL SIGNALS IN STRUCTURING V1 POPULATION ACTIVITY George Hilton Denfield Department of Neuroscience/M.D.-Ph.D. Program Advisor: Andreas Tolias, Ph.D.-Department of Neuroscience Neuronal responses to identical visual stimuli are variable, but the source of this variability is unknown. It is widely accepted that this variability results from neuronal noise. However, we offer an alternative explanation: this variability is not noise but reflects, and is due to, computations internal to the brain. Internal signals such as cortical state or attention interact with sensory information processing in early sensory areas. However, little research has examined the effect of fluctuations in these signals on neuronal responses, leaving a number of uncontrolled parameters that may contribute to neuronal variability. One such variable is attention. We hypothesize that fluctuations in attentional signals contribute to neuronal response variability and that controlling for such fluctuations will reduce this variability. We predict attentional effects on neuronal variability that differ from those predicted by the standard model of attention, in which attention reduces cortical noise to improve sensory processing, producing an inverse relationship between degree of attention and neuronal variability. Our model considers the effects of fluctuations in the attentional signal on recipient neurons and predicts the greatest degree of neuronal variability at an intermediate level of attention, corresponding to a state where the focus of attention is most variable. We control for fluctuations in the attentional signal in a change-detection task, varying across blocks of trials the degree to which subjects must attend to one of two stimuli presented in the visual field, while recording macaque primary visual cortex. Contributors: Denfield, George; Ecker, Alexander; Tolias, Andreas 62 2014 GRADUATE STUDENT SYMPOSIUM MODELING FOXF1 DEFICIENCY AND OVEREXPRESSION IN MICE Avinash Vijay Dharmadhikari Program in Translational Biology & Molecular Medicine Advisor: Pawel Stankiewicz, M.D./Ph.D.-Department of Molecular & Human Genetics Ignatia Van Den Veyver, M.D.-Department of Obstetrics & Gynecology Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a rare neonatally-lethal diffuse developmental disorder of the lungs caused by haploinsufficiency of FOXF1. All affected newborns die in the first month of life due to severe respiratory distress and pulmonary hypertension. Foxf1 null mice die by midgestation as a result of defects in mesodermal differentiation and cell adhesion. Foxf1 heterozygous mice exhibit up to 90% neonatal mortality, depending on genetic background. For the current study, Foxf1 knockout mice with a deletion of the forkhead binding domain were generated and are congenic on the C57BL/6J background. Analysis of RNA from postnatal day 0.5 Foxf1+/- and wildtype lungs using Illumina mouse WG-6 v2.0 expression bead chip microarray revealed statistically significant deregulation (p<0.05, fdr<0.05) of several genes, including those involved in pulmonary vascular development (Sema3C, Dll4, and Ednrb), lung branching morphogenesis (Fgf10 and Lama1), and the blood pressure regulating renin-angiotensin system (Ren1, Cma1, and Cpa3). To study the effects of Foxf1 overexpression, we knocked a Creinducible Foxf1 allele into the ROSA26 locus. ROSA26-LSL-Foxf1 mice were mated to CMV-cre mice to overexpress Foxf1 in all tissues and were mated to Tie2-cre mice to overexpress Foxf1 specifically in vascular endothelial cells. ROSA26Foxf1; CMV-cre mice exhibit early embryonic lethality around e12.5 while ROSA26Foxf1; Tie2-cre mice exhibit perinatal lethality around birth. Transcriptional changes in ROSA26Foxf1; Tie2cre e18.5 lungs (n=3) were compared to e18.5 ROSA26-LSL-Foxf1 (n=3) control lungs using the Illumina WG-6 v2.0 expression BeadChip kit microarrays. Pathways associated with granulocyte adhesion and diapedesis were found to be deregulated. Future studies will be directed towards characterizing lung vascular defects in the ROSA26Foxf1; Tie2-cre embryos. We are currently mating ROSA26Foxf1; Tie2-cre mice to Foxf1+/- mice, which we hypothesize, will rescue early postnatal mortality. This could inform future gene therapy studies in patients with ACDMPV. Contributors: Dharmadhikari, Avinash; Carofino, Brandi; Zabielska, Joanna; Wronowska, Weronika; Majewski, Tadeusz; Gambin, Anna; Szafranski, Przemyslaw; Justice, Monica J, Stankiewicz P. 63 BAYLOR COLLEGE OF MEDICINE A HIGH-THROUGHPUT FUNCTIONAL GENOMICS PLATFORM FOR DISCOVERY OF SOMATIC ABERRATIONS OF CANCER Turgut Dogruluk Department of Molecular & Human Genetics Advisor: Kenneth Scott, Ph.D.-Department of Molecular & Human Genetics Tumor sequencing projects such as The Cancer Genome Atlas (TCGA) have revealed the high complexity of cancer genomes that are comprised of both pathogenic “driver” aberrations and neutral “passenger” events. A major effort in the cancer research community involves discriminating drivers from passengers with the goal of prioritizing new therapeutic targets. To expedite driver discovery, I established a screening platform involving novel high-throughput mutagenesis and molecular barcoding (HiTMMoB) technology allowing our laboratory to engineer somatic mutations identified by TCGA into our collection of >32,000 human open reading frames (ORFs). Wild-type and mutant barcoded ORFs are subsequently entered into pooled in vitro/vivo genetic screens to identify those that can drive cancer phenotypes. Since developing HiTMMoB, I have continued using this technology for my own screens that include in vitro/vivo driver and pharmacological screens to assess differing effects of numerous mutations in the PIK3CA oncogene. PIK3CA is frequently mutated in breast cancer among other tissue types and regulates cancer’s most mutated pathway, PI3K. However, much of the focus of PIK3CA research is on the hot spot mutations of the PIK3CA gene and neglects infrequent ‘tail’ mutations. I have created 24 rare mutations of PIK3CA by utilizing HiTMMoB and entered them into few in vitro and in vivo screens to assess the effects of the mutations on several cancer phenotypes i.e. tumor formation, soft agar colony formation, growth-factor-independent growth and drug resistance/sensitivity. Even though there seems to be a general correlation between PIK3CA tail mutation frequency and ‘phenotype strength’ as expected, these experiments interestingly revealed that (1) PIK3CA mutations can be classified into two separate groups in terms of pathway activations, (2) some of the rarest PIK3CA mutations are still significantly oncogenic. All these data emphasized the need for ‘personalized and mutation specific’ cancer treatment approaches for PIK3CA-positive patients. In summary, this discovery pipeline revealed that not all the PIK3CA mutations are the same and thus should be treated differently requiring patient specific cancer therapies. The HiTTMoB technology together with downstream screening platforms promise to provide the cancer research community the functional annotation on the most promising cancer aberrations for drug development and targeting. Contributors: Dogruluk, Turgut; Dogruluk, Armel; Mills, Gordon B.; Scott, Kenneth L. 64 2014 GRADUATE STUDENT SYMPOSIUM DISCOVERY OF NOVEL REGULATORS OF SENSITIVITY AND RESISTANCE TO HER-FAMILY TARGETED THERAPIES Rocio Dominguez-Vidana Integrative Program in Molecular and Biomedical Sciences Advisor: Thomas Westbrook, Ph.D.-Department of Biochemistry & Molecular Biology Background: Targeted therapies exploit the fact that many tumors have genetic drivers that control tumor progression. Tumors rely on these drivers for their continued survival; therefore, inhibition of these genes strongly impairs tumor growth with minimal side effects to normal cells. However, resistance can arise when these drugs are used as single agents. Clinical and experimental data suggest that there is substantial heterogeneity in drug-response between patients and even between cells of the same tumor. However, the mechanisms driving this heterogeneity are poorly understood. The goal of this project is to identify the genetic networks that govern response to the anticancer drug lapatinib in Her2+ breast cancers. Experimental design and methods: We performed pooled RNA interferencebased genetic screens in a panel of Her2+ breast cancer cell lines using a short hairpin RNA (shRNA) library with ~9k unique hairpins targeting ~2k genes. We then assessed the effects of these shRNAs in untreated cells and in lapatinib-treated cells. We identified ~300 Lapatinib Sensitivity Regulators (LaSRs); from which we validated ~100 using small interference RNA (siRNA) based assays, and organized in protein networks using public protein-protein interaction (PPI) databases. Results: We focused on 12 LaSRs that show a combinatorial effect with lapatinib in multiple Her2+ models, and we systematically tested whether they shared a PPI and genetic coregulation. We have identified a potential mechanism in which our LaSRs WEE1, CSNK2A1, and CDC14B deregulate mitotic entry in conjunction with lapatinib, causing cell death. Conclusion: We developed an unbiased functional screen that identified genes governing sensitivity to lapatinib, and a validated these results using orthogonal methods. We identified WEE1, CSNK2A1, and CDC14B as a network governing lapatinib sensitivity from a systematic analysis of genetic coregulation and PPIs. Contributors: Ronald J Bernardi*1, Rocio Dominguez-Vidana*2,3, Christopher S Bland*2, Mitchell Rao4, Siddhartha Tyagi2, Kathleen A Scorsone2, Earlene M Schmitt2, Martin J Shea5, Tamika Mitchell5, Sarmistha Nanda5, Susan G Hilsenbeck5, Joe W Gray6, Carlos L Arteaga7, Brent N Rexer7, C Kent Osborne5, Chad A Shaw4, Rachel Schiff5, Thomas F Westbrook1,2,3,4,5 65 BAYLOR COLLEGE OF MEDICINE TRPV STRUCTURE AND FUNCTION INVESTIGATED WITH CRYO-ELECTRON MICROSCOPY AND NOVEL ACTIVITY ASSAYS Timothy Lloyd Dosey Integrative Program in Molecular and Biomedical Sciences Advisor: Theodore Wensel, Ph.D.-Department of Biochemistry & Molecular Biology Transient Receptor Potential (TRP) channels are a superfamily of non-selective cation channels conserved in eukaryotes. In mammals, the Vanilloid (TRPV) sub-family is involved in sensory processes including nociception and temperature sensing. However, our understanding of how TRP channel activity is regulated by these diverse stimuli has been impeded by the lack of structure-function information. Here we report that TRPV2 as well as a TRPV4 functional fragment can be purified from a yeast heterologous expression system and we demonstrate that they remain mono-dispersed in detergent-free buffer when stabilized with amphipoles. Further, we have developed cell-based and vesicle-based activity assays to investigate TRPV2 and TRPV4 function and we are utilizing recent advancements in cryo-electron microscopy to achieve high resolution structures without crystallization. With these structure and function techniques, we will be able to elucidate the gating mechanisms of these TRPV channels and gain insights into how other TRP channel families respond to a wide range of stimuli. Contributors: Dosey, Timothy; Wang, Zhao; Fan, Guizhen; Zhang, Zhixian; Serysheva, Irina; Chiu, Wah; Wensel, Theodore 66 2014 GRADUATE STUDENT SYMPOSIUM SHOHAT TYPE SPONDYLOEPIMETAPHYSEAL DYSPLASIA (SEMD) IS CAUSED BY MUTATIONS IN DDRGK1, ENCODING AN ENDOPLASMIC RETICULUM PROTEIN Adetutu Taiwo Egunsola Department of Molecular & Human Genetics Advisor: Brendan Lee, M.D./Ph.D.-Department of Molecular & Human Genetics Shohat type spondyloepimetaphyseal dysplasia (SEMD) is part of a heterogeneous group of disorders characterized by a combination of vertebral, epiphyseal and metaphyseal anomalies. Patients with Shohat type SEMD present with disproportionate short stature, a short neck, a small chest, abdominal distension with hepatosplenomegaly, lordosis, short limbs, genu varum and joint laxity. Radiographically, these patients also have a delayed bone age, platyspondyly with central notches in the vertebral end-plates, radiolucency of the femoral metaphyses as well as fibular overgrowth. The genetic basis of Shohat type SEMD is unknown. By exome sequencing of three individuals from two families, we identified a homozygous donor splice-site mutation in DDRGK1 in all affected individuals, and confirmed it in a separate family by Sanger sequencing. Patients with this splice-site mutation, a predicted frameshift mutation, have significantly reduced levels of DDRGK1 compared to controls. DDRGK1 is an ufmylated ER protein that has a conserved PCI domain, a domain predicted to participate in protein-protein interactions. To understand the role of DDRGK1 in skeletal development we knockdown DDRGK1 in zebrafish using morpholinos. DDRGK1 morphants have abnormal craniofacial features, such as disfigured cartilages and reduced number of branchial arches. Furthermore, there is a decreased level of SOX9, a type II collagen transcription activator, when DDRGK1 is knockdown in differentiated ATDC5, a chondrocyte cell line. Through Co-IP, we found that DDRGK1 interacts with SOX9. We then performed rescue experiments to determine whether SOX9 is a downstream target of DDRGK1 and found that overexpression of SOX9 rescues the craniofacial phenotype seen in DDRGK1 morphants. Therefore, we hypothesize that DDRGK1 regulates the stability of SOX9 and a deficiency in DDRGK1 decreases levels of SOX9. We are currently assessing whether DDRGK1 regulates the stability of SOX9 by affecting the ubiquitination state of SOX9. In addition, we are generating Ddrgk1 knockout mice using CRISPR/Cas9 system to future characterize the role of DDRGK1 in skeletal development. Contributors: Bae, Yangjin; Liu, David; Campeau, Philippe; Lu, James; Cohn, Daniel; Lachman, Ralph; Swindell, Eric; Shohat, Mordechai; Gibbs, Richard; Lee, Brendan 67 BAYLOR COLLEGE OF MEDICINE THE EFFECTS OF EXERCISE ON CRAVING AND USE OF CIGARETTES BY PARTICIPANTS DIAGNOSED WITH CONCURRENT TOBACCO- AND COCAINEUSE DISORDERS Joel David Eisenhofer Department of Neuroscience Advisor: Richard De La Garza, Ph.D.-Department of Psychiatry & Behavioral Sciences Individual exercise sessions (usually 10-30 min duration) have been shown to attenuate cravings to smoke and withdrawal symptoms in humans, though trials lasting several weeks have not consistently shown reductions in craving or improvements in abstinence. Key limitations of those trials include lack of sufficiently intense exercise interventions, lack of exercise adherence measurement, and change in physical activity. In this study, we evaluate the effects of treadmill exercise on basic fitness measures as well as objective and subjective measures of nicotine use and craving in individuals with concurrent cocaine and tobacco-use disorder. Cocaine-dependent cigarette smokers (N=24) were randomized to running or walking (30 min per session, 3 times per week) or sitting (placebo condition; same amount of time each day) for 4 consecutive weeks. The intensity of daily exercise for Runners and Walkers was calculated from each participant’s maximum heart rate (HR) during a treadmill test conducted while screening. Computerized cognitive behavioral therapy and contingency management were also given to all participants. Fitness measures included changes in body weight and resting HR. Objective measures for nicotine use included breath carbon monoxide (CO) and salivary cotinine. Subjective measures included changes in craving and motivation to quit smoking. Changes in cocaine use and craving are presented elsewhere (ACNP 2014). There were no differences in demographic or drug use variables among Runners (N=10), Walkers (N=7) and Sitters (N=7). On average, participants were Black (71%), male (80%), 44.7±1.1 (Mean ± S.E.M) years of age, and reported using cocaine for ~20 years. Participants smoked 12.4±1.8 cigarettes per day (CPD) for 26.4±2.1 years and had FTND scores of 4.5±0.6. At baseline, participants exhibited moderate scores (scale of 100) for Motivation to Quit smoking (56.5±6.8) and Confidence to Quit smoking (44.8±5.8). Across the 4-week study, exercise improved fitness measures including reducing body weight (-11.3±8.5, -4.0 ±2.9, +2.7±2.3 pounds; p=0.28) and decreasing resting HR (-3.3±4.1, +5.2±2.6, +9.1±3.0 bpm; p=0.05). Though not statistically significant, exercise reduced Questionnaire of Smoking Urges scores (-5.2±3.4, -18.4±5.5, -6.1±2.9, p=0.06), current craving for nicotine (VAS scale of 100)(-11.3±9.5, -20.2±9.9, +1.4±5.5, p=0.25) and CPD (-2.0±2.1, 1.6±2.1, -0.5±1.8, p=0.82). Unexpectedly, exercise reduced Motivation to Quit smoking (33.3±7.0, -3.6±16.9, +7.6±5.1, p=0.03) and reduced CO in Controls to a larger extent than Runners and Walkers (-1.7±1.5, +2.7±1.8, -5.9±1.9 ppm, p=0.01). The data show significant improvements in basic fitness measures though significant reductions in nicotine use and craving were not observed. It is established that cocaine users are 3-4 times more likely than their non-drug-abusing counterparts to smoke cigarettes, and smoking cigarettes appears to enhance craving and subsequent consumption of cocaine, making this a particularly challenging group to treat. Larger sample sizes and distinct exercise regimens should be evaluated for their potential to reduce smoking in individuals with concurrent cocaine and tobacco-use disorder. Contributors: Eisenhofer, Joel D.; Thompson-Lake, Daisy G.Y.; Debrule, Daniel S.; Makanjuola, Titi; Rodgman, Chris; Newton, Thomas F.; Yoon, Jin H.; De La Garza II, R. 68 2014 GRADUATE STUDENT SYMPOSIUM ROLE OF DNA END-BINDING IN KU HETERODIMER FUNCTION Charlene H Emerson Department of Molecular & Human Genetics Advisor: Alison Bertuch, M.D./Ph.D.-Department of Pediatrics The evolutionarily conserved Ku heterodimer is a DNA end-binding (DEB) complex involved in both telomere maintenance and non-homologous end-joining (NHEJ). In Saccharomyces cerevisiae, the Yku70/Yku80 subunits bind DNA ends via a channel structure. We previously generated DEB defective Ku mutants by mutating residues in the DNA-binding channel, revealing that DEB is re uired for Ku’s telomeric functions. Paradoxically, we also found that these mutants demonstrated increased colony survival in an assay that probes for imprecise NHEJ (Lopez et al. 2011, PLoS Genet). A subsequent study also generated DEB defective mutants of Ku by deleting residues to constrict the DNA binding channel (Pfingsten et al. 2013, Cell). These mutants similarly demonstrated a re uirement for Ku’s DEB activity at telomeres. However, these mutants were defective for imprecise NHEJ. To reconcile the differences between the two studies, we generated additional DEB defective Ku heterodimers and found that they too had increased imprecise NHEJ and defective telomere function. We also identified a novel region in Yku70 that is required solely for NHEJ and is predicted to associate with residues deleted in the DEB mutants reported by Pfingsten et al. We propose that these results reconcile the imprecise NHEJ differences observed and support our hypothesis that Ku heterodimers solely defective for DEB can modulate NHEJ off DNA ends. Currently, we are assaying whether the DEB mutants bind to double strand breaks (DSBs) in vivo. We expect that DEB mutants will be unable to bind DSBs, suggesting a previously unknown role for Ku in imprecise NHEJ. Contributors: Emerson, Charlene; Lopez, Christopher; Ribes-Zamora, Albert; Bertuch, Alison 69 BAYLOR COLLEGE OF MEDICINE POST-TRANSCRIPTIONAL REGULATION OF KRAS DURING HUMAN T CELL ACTIVATION Joseph Mario Fachini Department of Molecular Physiology & Biophysics Advisor: Joel Neilson, Ph.D.-Department of Molecular Physiology & Biophysics In our studies of post-transcriptional gene regulation in human T cells, we observed a significant upregulation of KRAS protein expression, independent of mRNA levels, upon antigen receptor stimulation in both primary human T cells and the Jurkat E6.1 cell line. Through the utilization of polysome fractionation facilitated assays, we show that KRAS is regulated at the level of translation. Additionally, we have systematically investigated putative regulatory elements and mechanisms of KRAS post-transcriptional regulation by the use of luciferase-based KRAS 3’ and 5’ untranslated region (UTR) reporter assays and poly(A) tail length assays. Through these approaches, we report that the induction of KRAS protein expression upon T cell activation is 3’ UTR mediated. By 3' UTR mapping via luciferase reporter assays, we have narrowed the region containing the cis-elements responsible for KRAS posttranscriptional regulation to approximately 1 kb. While several microRNAs have been implicated in the post-transcriptional regulation of KRAS, we show that the regulation of KRAS in our system is independent of both let-7 family microRNAs and miR-143. Through computational analysis of the mapped region, we identified HuR as a regulator of KRAS. By employing RNA-IP, luciferase reporter knockdown assays as well as deletional assays, we now demonstrate that HuR both binds to and induces translational upregulation of KRAS in human T cells post-T cell receptor mediated cellular activation. Contributors: Fachini, Joseph; Chaudhury, Arindam; Neilson, Joel 70 2014 GRADUATE STUDENT SYMPOSIUM RENAL ENDOTHELIAL CELL TNF SUPERFAMILY MEMBERS IN IMMUNE COMPLEX DISEASE River Uru Faille Department of Pathology & Immunology Advisor: Scott Wenderfer, M.D.-Department of Pediatrics Circulating immune complexes (IC)s deposit in the glomerulus of the kidney in many autoimmune diseases, including systemic lupus erythematosus (SLE), leading to proliferative glomerulonephritis and inflammation. IC binding can promote proliferation or induce apoptosis in human macrovascular EnCs, but the effects on microvascular EnCs such as those in the glomeruli are unknown. Tumor Necrosis Family (TNF) members regulate proliferation and apoptosis and are found in endothelial cells. TNF superfamily member 10 (Tnfsf10/TRAIL) is up-regulated in the kidneys of patients with IC disease. Membrane bound and soluble forms of TRAIL bind to TRAIL-R2/DR5 to induce apoptosis in some EnCs. TNF associated protein 3 (Tnfaip3/A20), is antiapoptotic, negatively regulating TRAIL/DR5. Polymorphisms in Tnfaip3 have been positively associated with disease in SLE. We hypothesize that TRAIL induces apoptosis in surrounding infiltrating leukocytes and that A20 blocks apoptosis in REnCs. Using qRT-PCR, we find that mRNA levels of these two TNF family members are upregulated in renal endothelial cells in vitro in the presence of IC binding and in vivo using a murine aCIC model . Using a Cellular ELISA for Membrane TRAIL, we show a stepwise increase in expression in REnCs upon treatment with ICs in a dose dependent manner. Using XTT and CFDA cell proliferation assays, findings suggest increased proliferation of cells treated with ICs. Future directions include using semi-quantitative western blots to measure A20 and soluble TRAIL expression levels in REnCs before and after exposure to IC. We will also study the role of IC binding to REnCs in regulation of DR5 signaling, caspase activity, and apoptosis, as well as the role of TNF superfamily members in proliferation and apoptosis. Finally, using co-culture systems, we will assess the importance of IC induced Trail production by REnCs on the proliferation or apoptosis of the leukocytes commonly found in inflammatory glomerulonephritis. Contributors: Suwanichkul, Adisak; Wenderfer, Scott E. 71 BAYLOR COLLEGE OF MEDICINE THE FUNCTION OF EXTRACELLULAR TRAP ASSOCIATED PROTEINS IN DICTYOSTELIUM DISCOIDEUM SENTINEL CELLS Timothy Farinholt Department of Biochemistry & Molecular Biology Advisor: Adam Kuspa, Ph.D.-Department of Biochemistry & Molecular Biology The social amoeba Dictyostelium discoideum is a model for studying bacteriaamoebae interactions that shape bacterial virulence. These initially solitary amoebae feed by consuming bacteria, but when starved, they lose their ability to consume bacteria and enter a 24-hour developmental cycle culminating in a fruiting body. During the migratory slug stage of development we have discovered a specialized population of Sentinel (S) cells that release structures similar to neutrophil extracellular traps (ETs) that ensnare and kill bacteria through an unknown mechanism (Chen et al., Science 317:678-81). ETs are comprised of mitochondrial DNA coated with antimicrobial proteins. We purified ETs and analyzed them by mass spectrometry, identifying several proteins, including CadA. Using available polyclonal and monoclonal anti-CadA antibodies we confirmed the presence of CadA on ETs. Amoeba lacking CadA protein (cadA-null) displayed variable growth on Klebsiella pneumoniae, which suggests that CadA is necessary for the killing of bacteria during growth and developmental phases of the Dictyostelium life cycle. Single wild type amoebae grown on a lawn of bacteria clear the bacteria in uniformly sized plaques. Single cadA-null amoeba cells form plaques of widely varying sizes that suggests a deficiency in the first few cell divisions of colony formation. Our lab’s data suggests Dictyostelium kills bacteria extracellularly before phagocytosis. Initial data suggest cadA-null amoebae are deficient in extracellular killing of bacteria. The crystal structure of CadA reveals an immunoglobulin-like and a gammacrystallin-like domains connected by a short linker sequence (Lin, Nat. Struct. Mol. Bio. 13(11):1016-22). Gamma-crystallin domains are found in plant antimicrobial proteins as well as yeast killer toxin (Huchinson, Protein Eng. 6(3):233-45). I am currently testing the potential role of CadA in the killing of bacteria by Dictyostelium discoideum. I will add exogenously purified CadA to bacteria to directly test its antimicrobial function. Purified CadA will be added to our extracellular killing assay to observe if it rescues the killing of bacteria in cadA-null amoebae. We hope to garner new insights into the ET mechanism for killing bacteria in eukaryotes. Contributors: Farinholt, Timothy; Zhuchenko, Olga 72 2014 GRADUATE STUDENT SYMPOSIUM TRPV3 AGONIST CARVACROL INDUCES HYPOTHERMIA IN CONSCIOUS MICE Viktor Feketa Program in Cardiovascular Sciences Advisor: Sean Marrelli, Ph.D.-Department of Anesthesiology Background: Therapeutic hypothermia is an effective and rapidly developing method of neuroprotection from ischemia. It is being increasingly used for the treatment of cardiac arrest and explored in stroke and traumatic brain injury. However, current methods of inducing hypothermia based on physical cooling have serious limitations, such as shivering, impaired consciousness, and respiratory depression. Novel approaches to lowering temperature in conscious patients are highly needed. One of such approaches involves pharmacological manipulation of the molecular thermosensors, which allows modulating the activity of the thermoregulatory system and may result in a regulated change in body temperature. In particular, agonists of the TRPV1 ion channel, also known as the heat and capsaicin receptor, are well known to induce hypothermia in numerous mammalian species. We aimed to explore if TRPV3 channel, which is closely related to TRPV1, may be targeted pharmacologically to achieve hypothermia. Similarly to TRPV1, TRPV3 is activated by above-neutral temperatures, expressed in peripheral tissues and was shown to be involved in transmitting temperature information to the cortex. These properties suggest that TRPV3 may also provide afferent input to the thermoregulatory system. However, this has not been yet conclusively established. We hypothesized that TRPV3-expressing sensory neurons functionally belong to the skin-warming afferent pathway, and that their activation by TRPV3 agonists provides spurious skin-warming signal to the thermoregulatory system, shifts it to less heat-generating and more heat-dissipating state, and leads to hypothermia in conscious subjects. Methods: We have determined the effects of systemic administration of TRPV3 agonist carvacrol in conscious mice on core temperature, measured by implanted telemetric transmitters, and on energy expenditure, measured by indirect calorimetry using the Comprehensive Laboratory Animal Monitoring System. Results: Intraperitoneal injection of carvacrol at 31.6 mg/kg b.w. led to ~2.5°C greater drop in core temperature after 30 min, compared to vehicle injection. This hypothermic effect was associated with a ~1.6-fold decrease in energy expenditure. Conclusions: Our findings support the working model, wherein TRPV3expressing neurons provide temperature input to the thermoregulatory system, and when activated by TRPV3 agonists, modulate the thermoeffector processes, lower whole-body heat generation, and lead to a decrease in core temperature in conscious mice. If further confirmed, this model suggests that TRPV3 agonists may be used as novel agents for induction of therapeutic hypothermia. Contributors: Marrelli, Sean. 73 BAYLOR COLLEGE OF MEDICINE A SIMPLIFIED SEQUENCE/STRUCTURE ALPHABET FOR MULTIPLE TRANSMEMBRANE HELICES ASSEMBLIES Xiang Feng Department of Pharmacology Advisor: Patrick Barth, Ph.D.-Department of Pharmacology TransMembrane Helical (TMH) domains of integral membrane proteins play critical roles in diverse physiological functions, such as the signal transduction, bioenergetics and ion transport, and are thus important drug targets. The lack of structural information on these proteins, however, hinders our understanding of their functional regulation and prevents the rational design of selective therapeutics. Computational modeling techniques represent important alternative approaches but currently lack the efficiency and accuracy required to consistently predict membrane protein structures at atomic resolution. Understanding the sequence/structure determinants controlling the packing of TMHs is a major step in accurately modeling and designing TMH proteins but such determinants has not yet been characterized for multiple TMH assemblies. We conducted a bioinformatics analysis of the sequence/structure relationships defining the packing of three TM helices. We generated a library of more than 800 closely packed TMH trimer structures from X-ray TM structures. Based on structure similarity, we found that more than half of the trimers can be classified into only 6 major clusters with distinct geometrical/topological features. We identified statistically enriched sequence motifs that correlate with the geometric features of each cluster. The motifs were used to train an SVM-based predictor of TMH trimer topologies from sequence that achieved up to 4-fold improvement over random selection, highlighting the significance and specificity of the motifs. Furthermore, enriched sequence/structure motifs share similar interaction patterns in diverse protein families, suggesting the existence of convergent determinants in TMH trimer packing. Analysis of residue conservation or co-evolution and in silico alanine scanning indicates that most motifs create energetically important contacts that are under evolutionary pressure. Structural analysis of atomic contacts mediated by these motifs uncovered novel consensus physical interactions that are unique to trimer and not found at dimer TMH interfaces. Our results indicate that a limited number of local sequence/structure motifs can recapitulate a large fraction of TMH trimer structures. These findings provide important novel insights into the sequence/structure determinants governing the packing of multiple TM helices and may guide the prediction and design of complex TMH structures. Contributors: Feng, Xiang; Barth, Patrick 74 2014 GRADUATE STUDENT SYMPOSIUM FURROW INGRESSION DRIVES DISASSEMBLY OF REMOTE MICROVILLAR FACTIN TO LIBERATE MEMBRANE FOR CELL SURFACE GROWTH Lauren Renee Figard Integrative Program in Molecular and Biomedical Sciences Advisor: Anna Sokac, Ph.D.-Department of Biochemistry & Molecular Biology Morphogenesis is driven by cell shape change, which often requires cell surface growth. Key aspects of this remodeling are still mysterious: What is the source of membrane for cell surface growth? How is membrane transferred to the site of growth? How is membrane transfer regulated? To address these questions, we study Drosophila cellularization, a dramatic tissue-building event that expands the embryo’s surface area by ~25 fold. We previously showed that microvilli unfold to provide membrane for cleavage furrow ingression during cellularization. We showed that microvillar disassembly is controlled by furrow ingression, and that microvillar membrane slides along the plane of the cell surface into ingressing furrows (Figard et al., 2013). These results provide the first direct evidence for plasma membrane unfolding, which has long been suspected as a broadly conserved mechanism of cell surface growth. But how does furrow ingression drive unfolding and the necessary disassembly of microvillar Factin cores? We propose that furrow pulling increases plasma membrane tension, antagonizing F-actin polymerization in microvilli and ultimately causing them to unfold. To validate this proposal, first, we used fluorescence recovery after photobleaching (FRAP) and drug studies to show that microvillar F-actin is constantly polymerizing and depolymerizing. Second, we used 3D time-lapse imaging to show that microvillar F-actin is depleted in sync with furrow ingression. Third, we analyzed a genetic mutant for furrow ingression and found that furrow ingression controls the depletion of microvillar F-actin. Fourth, we used physical force assays to show that furrow ingression exerts a pulling force that is transmitted to far-away microvillar F-actin. In all cells, the surface folds holding the largest membrane stores will likely be supported by F-actin. Our results suggest that plasma membrane tension is a critical regulator of unfolding, acting on F-actin to control the liberation and transfer of membrane during cell surface growth. Contributors: Figard, Lauren Renee; Sokac, Anna Marie 75 BAYLOR COLLEGE OF MEDICINE ROLE OF EXPERIENCE ON PLASTICITY OUTCOMES OF SPATIALLY SEPARATE SYNAPTIC PATHWAYS ONTO INDIVIDUAL NEURONS IN MOUSE VISUAL CORTEX Olivia Michelle Fitch Department of Neuroscience Advisor: Michael Friedlander, Ph.D.-Department of Neuroscience Mauro Costa-Mattioli, Ph.D.-Department of Neuroscience In the neocortex, individual cells of like type can undergo heterogeneous plasticity responses from depression (LTD) to potentiation (LTP), or no change (NC) in response to a common fixed time delay synaptic conditioning protocol. However it is not known whether all synapses onto a common cell have the same plasticity outcome. Nor is it known what role visual experience plays in shaping the distribution of differential plasticity outcomes. Thus, we evaluated the synaptic plasticity responses of separable sets of synaptic inputs onto common postsynaptic neurons in primary visual cortex in response to simultaneous stimulation of distinct sets of afferents in acute brain slices from visually intact or binocularly deprived mice (deprived from before the natural time of eye-opening). The two stimulation sites were isolated by occlusion testing followed by alternative activation of each pathway to evoke a postsynaptic potential (PSP) every 10 seconds in an interleaved fashion. After a stable ten minute baseline period, the activation of both pathways was simultaneously paired with direct postsynaptic activation that preceded the synaptic stimulation by 10 milliseconds resulting in 3-7 postsynaptic spikes at 0.1 Hz over a 10 min period followed by reversion to the interleaved stimulation protocol for an additional 30 minutes. The ratio of the average amplitude of the evoked PSP post/pre conditioning was calculated for each pathway taking the 5 minutes average peak amplitude over 25-30 minutes postconditioning compared to 5 minutes just before or pre-conditioning. Our results from 132 pathways inputs validate in the mouse cortex our previous findings from other species demonstrating heterogeneous plasticity outcomes ranging from LTD to LTP for individual cells with a median post/pre ratio of 0.84 in visually intact mice (both naïve and anesthesia controls). Visually deprived mice did not show a significant difference in the change of individual pathways with a mean 0.89 post/pre ratio for 63 pathways (Rank Sum U-statistic=2027, p=0.424). However, there was relationship between two pathways onto a common cell differed depending on the level of visual experience. There was a weak correlation between two pathways in visually deprived mice (median R=0.49, p=0.005, n=30 cells) and none in binocularly deprived mice (median R = 0.023, p = 0.9, n=31 cells) with a significant difference between correlation coefficients (t-test, p=0.05) as measured by a linear fit to 10,000 permutations of ordinate and abscissa designation of post/pre ratios of two pathways onto a common cell for each treatment group created by Monte Carlo Permutation analysis. Supported by NIH grant EY-12782 to MJF. Contributors: Fitch, Olivia M; Friedlander, Michael J 76 2014 GRADUATE STUDENT SYMPOSIUM SRC-2 Dynamically Regulates Gluconeogenesis and Glycolysis via Glucose-6phosphatase and Glucokinase Tiffany C Fleet Program in Translational Biology & Molecular Medicine/M.D.-Ph.D. Program Advisor: Bert O'Malley, M.D.-Department of Molecular & Cellular Biology Clifford Dacso, M.D./M.P.H.-Department of Molecular & Cellular Biology Steroid Receptor Coactivator 2 (SRC-2) is a well-characterized transcriptional coregulator of systems-wide energy homeostasis. Ablation of SRC-2 in mice protects against diet-induced obesity, impairs glucose home¬ostasis, and disrupts efficient dietary absorption of lipids. Our laboratory has defined SRC-2 as a transcriptional coactivator for RORα on the Glucose-6-phosphatase (G6pc) promoter, a rate-limiting enzyme in gluconeogenesis. Loss of SRC-2 results in decreased G6pc expression, accumulation of liver glycogen, and fasting hypoglycemia. In addition to the fasting hypoglycemia, SRC-2-/- mice have prandial hyperglycemia after a 24 hour fast. We determined that expression of Glucokinase (Gck), a rate-limiting step in glycolysis, is also decreased in SRC-2-/- mice. Therefore, we hypothesize that SRC-2 regulates the switch between gluconeogenesis and glycolysis by regulating expression of G6pc during fasting conditions and Gck during feeding conditions. To better understand the role of SRC-2 in regulating glucose metabolism, we surveyed SRC-2 cistrome occupancy on the G6pc and Gck promoters. To determine the coregulator complex that assist in this dynamic regulation, we next made biotinylated PCR fragments of both G6pc and Gck promoters for a DNA pull-down assay. The biotinylated promoters were incubated with hepatic nuclear extract containing transcriptional machinery from WT and SRC-2-/- mice that were either fasted for 24 hours or fasted and refed for 3 hours. To assess the dynamics of the coregulator complexes formed on the G6pc and Gck promoter regions, we performed proteomic analysis of the DNA pull-down via mass spectrometry and immunoblotted selected targets. We found that SRC-2 is involved in differential recruitment of coregulator complexes in a metabolic and promoter sequence specific manner. Contributors: Fleet, Tiffany; Zhang , Bin, Stashi , Erin; Jung , Sung Yun; Rajapakshe, Kimal; Dean, Adam; Gonzales, Naomi; Foulds, Charles; Coarfaor, Christian; Qin, Jun; York, Brian; O’Malley, Bert. 77 BAYLOR COLLEGE OF MEDICINE THE ROLE OF SRC-2 AND SRC-3 IN CASTRATION-RESISTANT PROSTATE CANCER Christopher James Foley Department of Molecular & Cellular Biology Advisor: Nicholas Mitsiades, M.D./Ph.D.-Department of Medicine The focus of prostate cancer research and treatment has historically been on targeting the androgen receptor (AR) signaling axis, as the anticancer activity of AR inhibition (via chemical castration or anti-androgens) has been well established. Despite this, if given enough time, prostate cancer will almost always recur as a castrationresistant prostate cancer (CRPC), which can metastasize and grow despite anti-AR treatments. Although several mechanisms have been proposed to explain this transition to CRPC, it is clear that targeting only the AR signaling axis is insufficient. The steroid receptor coactivators (SRCs) are a family of coregulators that play key roles in transcription. Overexpression of the SRCs has been implicated in numerous cancer types – including prostate. To understand the role SRCs play in CRPC, we performed Chromatin Immunoprecipitation-sequencing (ChIP-Seq) for AR, SRC-1, SRC-2 and SRC-3 in an androgen-dependent prostate cancer cell line and its androgenindependent (CRPC) subclone. We observed a substantial increase in total cellular protein levels and in the number of chromatin binding sites for both SRC-2 and SRC-3, as well as a transition away from co-localization with AR chromatin binding, in our CRPC model compared to its androgen-dependent parental cells. We integrated these results with gene expression profiles of cells treated with siRNA against SRC-2 and SRC-3, and identified a set of genes that is regulated by SRCs specifically in our CRPC model. These SRC-dependent, CRPC-specific genes included the E3 ubiquitin ligase Sphase kinase-associated protein 2 (SKP2), a known oncogene that is overexpressed in CRPC patient samples. Silencing of SRC-2 or SRC-3 or SKP2 via siRNA exerted greater anticancer activity against our CRPC model than its androgen-dependent parental cell line. In agreement with this increased dependency of CRPC cells on SRC2 and SRC-3, we found that a small molecule inhibitor (SMI) of the SRCs, which was identified via screening of a library of ~360,000 compounds, exerted greater anticancer activity against CRPC than androgen-dependent parental cells. Both SRC-3 siRNA and the SRC SMI suppressed SKP2 expression specifically in CRPC cells. Furthermore, recruitment of SRC-3 to the SKP2 promoter was suppressed by the SRC SMI. Our data suggest that SRC-2 and SRC-3 contribute to prostate cancer progression to CRPC. Targeting the SRCs with SMIs is feasible and promises an innovative therapeutic approach for patients with CRPC. Contributors: Foley, Christopher; Lanz, Rainer; Fiskus, Warren; Geng, Chuandong; Shou, John; He, Bin; Shah, Shrijal; Chew, Sue Ann; Coarfa, Cristian; O’Malley, Bert W.; Mitsiades, Nicholas 78 2014 GRADUATE STUDENT SYMPOSIUM LYSOSOMAL SIGNALING MOLECULES REGULATE LONGEVITY IN CAENORHABDITIS ELEGANS Andrew Kenji Folick Program in Developmental Biology/M.D.-Ph.D. Program Advisor: Meng Wang, Ph.D.-Department of Molecular & Human Genetics Lipids are known for their roles in energy storage and cellular architecture, but they also act as signaling molecules involved in the regulation of gene expression and signal transduction. Although fat storage and metabolism have been associated with metabolic health and aging, the role that lipid signaling plays in the regulation of longevity has not been elucidated. Recent work has implicated autophagy and lysosomal function as involved in the regulation of longevity. Here, we explore the relationship between lysosomal function and lipid signaling in aging by characterizing the role of lipl-4, a homolog of human lysosomal acid lipase, as a novel regulator of longevity in Caenorhabditis elegans, and show that constitutive expression of lipl-4 in intestinal fat storage tissue decreases fat storage and increases both mean and maximum lifespan. Increased expression of lipl-4 promotes the expression and lysosome-to-nuclear translocalization of the lipid chaperone LBP-8. LBP-8 promotes the activation of transcription by a nuclear hormone receptor complex of NHR-49 and NHR80, and over-expression of LBP-8 is sufficient to promote longevity, dependent on NHR49 and NHR-80. Using high-throughput metabolomic analysis, we identified several potential lipid messengers with increased abundance in animals over-expressing lipl-4. Among these, oleoylethanolamide bound to both LBP-8 and NHR-80 in vitro, and feeding oleoylethanolamide to worms was sufficient to promote NHR-49/NHR-80 dependent transcription. Furthermore, feeding oleoyethanolamide was sufficient to increase mean lifespan, dependent on NHR-80. In summary, we have identified a new pathway by which lipid catabolism in the lysosome signals to the nucleus to regulate lifespan, and identified a potential longevity-promoting lipid metabolite. Contributors: Folick, Andrew; Oakley, Holly; Yu, Yong; Armstrong, Eric H; Kumari, Manju; Sanor, Lucas; Moore, David D.; Zechner, Rudolf; Ortlund, Eric A.; Wang, Meng C. 79 BAYLOR COLLEGE OF MEDICINE THE ROLE OF MAGEL2 AND ITS INTERACTING PARTNERS IN NEUROPSYCHIATRIC PHENOTYPES Michael David Fountain Program in Translational Biology & Molecular Medicine Advisor: Christian Schaaf, M.D./Ph.D.-Department of Molecular & Human Genetics Daryl Scott, M.D./Ph.D.-Department of Molecular & Human Genetics The Prader-Willi syndrome (PWS) critical domain is a maternally imprinted, paternally expressed locus, which contains five protein coding genes and a family of six small nucleolar RNA (snoRNA) genes or clusters. However, the roles of each of these genes and snoRNAs in relation to PWS associated phenotypes remain largely unknown. Most recently, four individuals with major features of PWS were reported to carry truncating mutations in their paternal copy of MAGEL2, a protein-coding gene in the PWS critical domain. All of these individuals had previously been diagnosed with autism spectrum disorder (ASD). We hypothesize that loss of functional MAGEL2 predisposes to ASD, and that mutations in proteins interacting with MAGEL2 may also lead to neurocognitive and neuropsychiatric phenotypes. Specific Aim 1: To investigate whether Magel2 null mice display autism-like behaviors. Previously, Magel2 null mice were generated and evaluated for key behaviors and characteristics of PWS. However, they have not yet been evaluated for autism-like behaviors. We propose to assess male and female Magel2 null mice for autism-like behaviors, using a battery of validated, well-established neurobehavioral tests. Test animals will be compared to age- and sex-matched wild-type littermates, analyzed in parallel. The behavioral assessment will include evaluations for communication deficits, social recognition, social interaction, social dominance, repetitive behaviors and stereotypies, as well as learning and memory. Specific Aim 2: To identify and investigate the pathogenic significance of mutations in MAGEL2 interacting proteins. Recently, MAGEL2 was shown to form a functional complex with TRIM27 and USP7, which then activates the WASH complex (WASH, FAM21, CCDC53, SWIP, STRUM) in a ubiquitin-dependent fashion. The activation of the WASH complex plays a critical role in endosomal actin nucleation and protein trafficking. Due to their close physical and functional relationship, we propose that mutations in MAGEL2 interacting proteins may have similar phenotypic consequences as MAGEL2 loss-of-function itself. We queried databases of >100,000 individuals who had clinical array comparative genomic hybridization (aCGH) testing, and databases of >5,000 individuals who underwent whole exome sequencing at Baylor College of Medicine, and successfully identified a first set of individuals with loss-of-function mutations in MAGEL2 associated genes. Primary phenotypes from this query were intellectual disability, developmental delay, aggressive behavior, seizures, and autism. To investigate the significance of the respective mutations, lymphoblast and fibroblast cell lines will be established from the affected patients, which will allow us to assess the functional significance of these mutations on the MAGEL2-TRIM27-USP7 complex and the WASH complex. Contributors: Fountain, Michael; Schaaf MD PhD, Christian 80 2014 GRADUATE STUDENT SYMPOSIUM A COMPREHENSIVE INVESTIGATION OF SIGNALING AXES UPON ANTIGEN SPECIFIC ACTIVATION OF CHIMERIC ANTIGEN RECEPTOR (CAR) T CELLS Kristen Nicole Fousek Program in Translational Biology & Molecular Medicine Advisor: Nabil Ahmed, M.D.-Department of Pediatrics Helen Heslop, M.D.-Department of Pediatrics CAR T cells have shown promising results for several cancers in pre-clinical models as well as in early phase clinical trials. Throughout these studies it has been observed that first generation CAR T cells demonstrate successful killing of tumor cells but are unable to proliferate sufficiently while second generation CAR T cells are able to kill tumor cells as well as proliferate and exhibit improved survival in vitro. Furthermore, third generation CAR T cells display enhancements in cytotoxicity, proliferation, and cell survival, however, there has not yet been a comprehensive investigation that interrogates the correlation of signaling patterns with CAR endodomains and functionality thereof. Substantial knowledge on TCR signaling exists, and the pathways downstream of the TCR/CD3 zeta chain as well as those of the CD28, 4-1BB, and OX40 co-stimulatory molecules on canonical T cells are well understood. In this project I propose to build upon this T cell signaling knowledge and investigate why distinct CAR moieties confer enhanced functionalities to T cells by analyzing the activation of phosphoproteins involved in key signaling pathways on a molecular level. I plan to do this through conducting the following specific aims: Aim 1: To construct and validate the phenotype and function of Jurkat and primary human T cells expressing CARs comprised of permutations of the ζ, CD28, 41BB, and OX40 signaling moieties. Aim 2: To investigate trends in activation of surrogate nodes involved in distinct signaling pathways downstream of the co-stimulatory domains in first-, second-, and third-generation CARs. I hypothesize that encounter and binding of specific target antigens will activate CAR signaling molecules in a dose-dependent fashion and generate distinct quantifiable differences in the activation of surrogate nodes within the signaling pathways, culminating in correlative changes in the effector functions of CAR T cells. Contributors: Fousek, Kristen, Byrd, Tiara, Heczey, Andras, Varadarajan, Navin, Heslop, Helen, Gottschalk, Stephen, Dotti, Gianpietro, Hegde, Meenakshi, Ahmed, Nabil 81 BAYLOR COLLEGE OF MEDICINE THE ROLE OF ORPHAN NUCLEAR RECEPTORS NR4A1 AND NR4A3 IN HEMATOPOIESIS Pablo Riera Freire Department of Molecular & Cellular Biology Advisor: Orla Conneely, Ph.D.-Department of Molecular & Cellular Biology Hematopoiesis is a dynamic biological process that requires tight coordination between the differentiation and proliferation of hematopoietic stem cells (HSC) and progenitors. At the cellular level, these processes are controlled by transcriptional and epigenetic mechanisms that regulate the correct gene expression program for each blood cell maturation stage. Our group has previously shown that the members of the NR4A nuclear receptor family act as tumor suppressors in the hematopoietic system, and germline deletion of Nr4a1 and Nr4a3 in mice causes death by acute myeloid leukemia (AML) within 2-4 weeks after birth. However, the cellular and molecular mechanisms by which NR4As regulate hematopoietic cell development remain poorly understood. To disclose these mechanisms, we have used a tamoxifen-inducible Rosa26-Cre-ERT2; Nr4a1fl/fl; Nr4a3-/- mouse model to examine the cellular and molecular consequences of acute codepletion of Nr4a1 and Nr4a3 on hematopoietic stem (HSC) and progenitor cell homeostasis in adult mice prior to the development of AML. We have previously confirmed that temporally restricted depletion of both genes using this approach leads to development of AML in adult mice within 12 to 15 weeks after Nr4a1/3 ablation. Expression profiling of NR4A1/3 revealed highest levels of expression of both genes in the most primitive HSC population with lower levels in the myeloid progenitors. Consistent with these observations, we find that acute deletion of NR4A1/3 leads to a rapid increase (within four days) in the frequency of the most primitive HSC and multipotent progenitor (MPP) populations. Proliferation analysis revealed that accumulation of HSCs and MMPs was associated with abnormal activation of cell cycle in both populations. The same analyses were repeated four weeks after tamoxifen treatment, and we observed a depletion of the HSC compartment and an accumulation of the granulocyte-monocyte progenitor population. The cell cycle remained abnormally activated in the HSC and MPP, indicating that the excessive proliferation caused a reduction of the stem cell pool. We are currently utilizing a systems approach to identify global NR4A1/3 transcriptional targets by integrating ChIPSeq and RNA-Seq analyses in the rare long-term HSC. We expect these analyses to uncover new mechanisms controlling normal adult hematopoiesis and neoplasia. Contributors: Freire, Pablo; Conneely, Orla 82 2014 GRADUATE STUDENT SYMPOSIUM POPULATION CODE IN MOUSE V1 FACILITATES READOUT OF NATURAL SCENES THROUGH INCREASED SPARSENESS Emmanouil Froudarakis Department of Neuroscience Advisor: Andreas Tolias, Ph.D.-Department of Neuroscience Neural codes are believed to have adapted to the statistical properties of the natural environment. However, the principles that govern the organization of ensemble activity in the visual cortex during natural visual input are unknown. We recorded populations of up to 500 neurons in the mouse primary visual cortex and characterized the structure of their activity, comparing responses to natural movies with those to control stimuli. We found that higher order correlations in natural scenes induced a sparser code, in which information is encoded by reliable activation of a smaller set of neurons and can be read out more easily. This computationally advantageous encoding for natural scenes was state-dependent and apparent only in anesthetized and active awake animals, but not during quiet wakefulness. Our results argue for a functional benefit of sparsification that could be a general principle governing the structure of the population activity throughout cortical microcircuits. Contributors: Froudarakis ,Emmanouil; Berrens, Philipp; Cotton, R. James; Ecker, Alexander; Sinz, Fabian H.; Yatsenko, Dimitri; Saggau, Peter; Bethge, Matthias; Tolias, Andreas S. 83 BAYLOR COLLEGE OF MEDICINE METASTASIS-ASSOCIATED ALTERATION OF GATA6 AND TG2 EXPRESSION IN OSTEOSARCOMA Daniel G Fuja Integrative Program in Molecular and Biomedical Sciences/M.D.-Ph.D. Program Advisor: Jason Yustein, M.D./Ph.D.-Department of Pediatrics Osteosarcoma (OS) is the most common form of primary bone cancer. Age of OS incidence follows a bimodal distribution with a higher incidence in pediatric and adolescent patients. It is found primarily in long bones such as the femur and humerus, with metastases most often arising in the lung or other bony sites. Metastatic spread of OS from the primary tumor to distant metastatic sites drastically decreases survival rate. Current treatment of metastatic OS still largely parallels that of the non-metastatic disease, usually consisting of surgical resection of the primary tumor and adjuvant chemotherapy. Treatment of metastatic OS would be facilitated by a better understanding of factors driving osteosarcoma metastasis. Metastatic progression is often enabled through altered expression of genes affecting motility, invasive potential, immune evasion, and other significant functions. We hypothesize that specific genetic and molecular changes are required for OS cells to metastasize from the primary tumor and colonize a distant, physiologically distinct site. Because chemotherapy is begun immediately following diagnosis of OS, there is limited access to samples of lung metastases which have not already been exposed to chemotherapeutic regimen and, thus, some selection. In order to investigate untreated lung metastasis, our lab has developed genetically engineered mouse models (GEMMs) which phenocopy either metastatic or non-metastatic osteosarcoma. Using highthroughput genome-wide microarray, we screened RNA isolated from GEMM-derived primary tumors and their corresponding metastatic lesions for genes significantly altered. These microarrays were subsequently repeated with cell lines derived from corresponding primary and metastatic lesions. Several factors exhibited higher mRNA levels in lung metastases than their corresponding primary tumor, both in tissue and cell line mRNA. Statistical analysis and stringent filtering yielded high-probability candidates which were subsequently corroborated through multiple-sample quantitative PCR (qPCR). Some cell line protein levels were also checked through Western Blot to confirm upregulation. Corresponding data in human-derived samples were analyzed and confirmed upregulation of Gata6 and TG2. Analysis via Ingenuity Pathway Analysis (IPA) showed important connections to immune evasion, cell motility, and other prometastatic survival functions for two of the consistently-upregulated factors: Gata6 and Transglutaminase 2 (TG2). These pro-metastatic mechanisms may be required for OS dissemination, colonization and metastatic tumor growth. Functional studies indicate that alterations in Gata6 and TG2 affect cellular phenotypes which might facilitate metastatic disease through multiple functions. Anticipated in vivo experiments will demonstrate the extent of metastatic enhancement. Contributors: Fuja, Daniel; Kurenbekova, Lyazat; Roos, Alison; Gao, Yang; Donehower, Lawrence A.; Yustein, Jason T. 84 2014 GRADUATE STUDENT SYMPOSIUM THE INTERACTION OF ONCOGENIC PHOSPHATASE WIP1 AND p27KIP1 IN DNA DAMAGE RESPONSE Kenichiro Fujiwara Integrative Program in Molecular and Biomedical Sciences Advisor: Lawrence Donehower, Ph.D.-Department of Molecular Virology & Microbiology A mammalian cell undergoes the DNA Damage Response (DDR) after encountering various types of genotoxic stresses, including ionizing radiation, which results in single or double-stranded breaks in the DNA. The sensor kinases ATM and ATR detect this damage and activate downstream effectors through phosphorylation cascades to promote various cellular responses, including cell cycle arrest. Once DNA repair is complete, the expression of Wild-type p53-induced phosphatase 1 (WIP1) is induced to return the cell to a homeostatic state. WIP1 has been shown to dephosphorylate and downregulate various DDR mediators and effectors, including p53, CHK1, CHK2, ATM/ATR, MDM2, and H2AX. WIP1 is amplified and overexpressed in numerous human cancers. WIP1 overexpression in cancer cells expressing wild-type p53 leads to suppression of p53 activity. WIP1 inhibition, either chemically or through the overexpression of an inhibitory miRNA, has been shown to suppress tumor growth via induction of cellular senescence or apoptosis. Thus, understanding the influence of WIP1 on various cellular pathways involved in cancer is critical, since WIP1 has been suggested to be a potential therapeutic target. Currently, our knowledge of the function of WIP1 is mainly limited to its role in the DDR pathway. Preliminary data from our lab suggests that WIP1 may play a role in cell cycle regulation. Our findings suggest that WIP1 may target p27Kip1 (also known as CDKN1B), a known inhibitor of CDK2 and associated cyclins that directly regulate the cell cycle. Thus, we believe that WIP1 may play a more direct role in controlling the cell cycle than has previously been suggested. We hypothesize that WIP1 dephosphorylates p27Kip1 after completion of DDR to promote cell cycle progression. To investigate the role of p27Kip1 in DDR, we are examining the ability of WIP1 to target p27Kip1 S140, a site implicated in DDR signaling by ATM, and testing the ability of WIP1 to promote cell cycle progression. Previous results indicate that WIP1 dephosphorylates p27Kip1 S140 in vitro, and western blot analysis suggests that this same site is phosphorylated by ATM in 293 cells. Studies are currently underway that are designed to observe p27Kip1 S140 dephosphorylation by WIP1 in cultured cells and to elucidate the mechanism for cell cycle regulation through WIP1 modulation of p27Kip1. The significance of this study is the potential discovery of additional cell cycle targets for cancer treatments that involve WIP1 modulation. Contributors: Fujiwara, Kenichiro; Donehower, Larry 85 BAYLOR COLLEGE OF MEDICINE SIGNALING THROUGH SMAD4 IN PANCREATIC CANCER Paul Thomas Fullerton Department of Molecular & Human Genetics Advisor: Martin Matzuk, M.D./Ph.D.-Department of Pathology & Immunology Pancreatic cancer is fourth-leading cause of cancer death in the United States with approximately 43,000 cases diagnosed each year. For patients who present with locally advanced or metastatic disease, average survival times are less than one year and there are no curative treatments. The transforming growth factor β (TGFβ) signaling protein SMAD4 is lost in 75% of metastatic pancreatic cancers and has been associated with poorer prognosis, suggesting that SMAD4 loss is an important event in the development of aggressive, metastatic disease. However, the mechanisms by which SMAD4 loss promotes the development of a highly aggressive disease are not well understood. In our preliminary studies, we discovered that expressing SMAD4 in a SMAD4-null human pancreatic cancer cell line inhibits proliferation and induces senescence, suggesting that targeting the TGFβ/SMAD4 signaling pathway could be a promising therapeutic strategy. We hypothesized that understanding the mechanisms underlying the anti-proliferative effect of SMAD4 expression would enable the development of new therapies. To facilitate a more detailed study, we generated an inducible system for SMAD4 expression in the SMAD4-null human pancreatic cancer cell line BxPC3. By comparing the expression profiles of this cell line with and without SMAD4 at a variety of time points after induction of SMAD4 expression, we identified candidate regulatory partners and targets of SMAD4 in pancreatic cancer. Our future studies will validate these candidate genes and characterize their connection to the tumor suppressive role of SMAD4, laying the groundwork for the development of therapies targeting the TGFβ/SMAD4 signaling pathway. Contributors: Fullerton, Paul 86 2014 GRADUATE STUDENT SYMPOSIUM MICROBIAL DERIVED HISTAMINE: EFFECTS ON TNF PRODUCTION BY INTESTINAL MACROPHAGES Robert Steven Fultz Integrative Program in Molecular and Biomedical Sciences Advisor: James Versalovic, M.D./Ph.D.-Department of Pathology & Immunology Chronic intestinal inflammation reduces quality of life and is deleterious to intestinal tissues. Due to the intimate relationship between the intestinal microbiota and host intestinal tissues, probiotic therapies are an attractive approach for treating chronic colitis, yet no such therapy has been proven effective in inducing remission and maintenance of chronic intestinal inflammation. Currently, therapies with the TNF-α directed monoclonal antibodies are indicated and effective for IBD refractory to corticosteroids, but are prohibitively expensive for some patients and require long-term administration [1]. In vitro, administration of the versatile, biogenic amine, histamine reduces expression and production of TNF-α in human monocyte derived macrophages [2]. The probiotic, lactic acid bacterium, Lactobacillus reuteri is a native inhabitant of the mammalian gut and produces and secretes histamine. Administration of L. reuteri ATCC 6475 to mice with TNBS-induced colitis ameliorates local and systemic markers of inflammation in a histamine-dependent manner [†]. Production and secretion of histamine by L. reuteri ATCC 6475 in the gut is, therefore, hypothesized to reduce colitis by inhibiting TNF production from intestinal macrophages; however the mechanism by which this occurs in not fully elucidated. A cell model for studying the effects of L. reuteri ATCC 6475 on macrophages has been characterized. The mechanism by which histamine signaling reduces TNF transcription in intestinal macrophages will next be determined by assaying phosphorylation states of MAP kinase pathway proteins in macrophages exposed to histamine in vitro, or L. reuteri ATCC 6475 in vivo. Elucidation of this mechanism is essential for understanding the probiotic functions of microbial-derived histamine and its clinical applications and limitations. 1. Blonski W, et al. (2011) Inflammatory bowel disease therapy: current state-ofthe-art. Current Opinion in Gastroenterology, 27:346-357. 2. Thomas C, et al. (2012) Histamine Derived from Probiotic Lactobacillus reuteri Suppresses TNF via Modulation of PKA and ERK Signaling. PLoS ONE, 7(2):e31951. †. Unpublished data. Contributors: Fultz, Robert 87 BAYLOR COLLEGE OF MEDICINE A VOLTAGE GATED CALCIUM CHANNEL REGULATES LYSOSOMAL FUSION WITH ENDOSOMES AND AUTOPHAGOSOMES AND IS REQUIRED FOR NEURONAL HOMEOSTASIS Upasana Gala Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics Autophagy helps deliver sequestered intracellular cargo to lysosomes for proteolytic degradation and thereby maintains cellular homeostasis by preventing accumulation of toxic substances in cells. In a forward mosaic screen in Drosophila designed to identify genes required for neuronal function and maintenance, we identified multiple cacophony (cac) mutant alleles. They exhibit an age dependent accumulation of autophagic vacuoles (AVs) in photoreceptor terminals and eventually a degeneration of the terminals and surrounding glia. cac encodes an α1 subunit of a Drosophila voltage gated calcium channel (VGCC) that is required for synaptic vesicle fusion with the plasma membrane and neurotransmitter release. Here, we show that cac mutant photoreceptor terminals accumulate AV-lysosomal fusion intermediates suggesting that Cac is necessary for the fusion of AVs with lysosomes, a poorly defined process. Loss of another subunit of the VGCC, α2δ or straightjacket (stj), causes very similar phenotypes as the loss of cac, indicating that the VGCC is required for AVlysosomal fusion. The role of VGCC in AV-lysosomal fusion is evolutionarily conserved as the loss of the mouse homologues, Cacna1a and Cacna2d2, also leads to autophagic defects in mice. Moreover, we find that CACNA1A is localized to the lysosomes and that loss of lysosomal Cacna1a in cerebellar cultured neurons leads to a failure of lysosomes to fuse with endosomes and autophagosomes. Finally, increasing cytosolic calcium levels rescues lysosomal fusion defects in Cacna1a mutant neurons. We present a model in which the VGCC plays a role in autophagy by regulating the fusion of AVs with lysosomes through its calcium channel activity and hence functions in maintaining neuronal homeostasis. Contributors: Gala, Upasana; Tong, Chao; Tian, Xieujen; Nagarkar, Sonal; Yamamoto, Shinya; Jaiswal, Manish; Sillitoe, Roy; Bellen, Hugo 88 2014 GRADUATE STUDENT SYMPOSIUM A NEW PATHWAY FOR CALCIUM MOBILIZATION BY BETA-2 ADRENERGIC RECEPTOR Monica Laura Galaz-Montoya Department of Biochemistry & Molecular Biology Advisor: Theodore Wensel, Ph.D.-Department of Biochemistry & Molecular Biology Beta adrenergic receptors are important for cardiovascular regulation and for physiological responses to the hormones/neurotransmitters adrenaline and noradrenaline. They are found in the nervous system and throughout the body and are the targets of numerous widely used drugs. Their canonical signaling pathway involves activation of adenylate cyclase (AC) and a rise in cyclic AMP (cAMP) levels which leads to activation of cAMP-dependent protein kinase (PKA). Beta adrenergic receptors have long been thought to activate distinct signaling pathways from those downstream of Gprotein coupled receptors which activate phospholipase C (PLC) and elevate intracellular Ca2+. By monitoring of intracellular Ca2+ levels in real time using a fluorescent indicator dye we found that an endogenous receptor in HEK-293 cells responds to the adrenergic agonist norepinephrine, by a delayed rise in intracellular [Ca2+]. The response is blocked by ICI 118,551, a selective antagonist for beta-2 adrenergic receptor (β2-AR), and the relative potency of agonists is isoproterenol > epinephrine > norepinephrine, consistent with the pharmacological profile of β2-AR. Treatment with thapsigargin (an inhibitor of the SERCA Ca2+ pump of the endoplasmic reticulum) and chelation of extracellular Ca2+ revealed that the Ca2+ is released from intracellular stores. The release is sensitive to inhibition of PLC with U73122 and of InsP3 receptors with 2-APB. Treatment with cholera toxin, a drug that activates Gαs, indicated that direct activation of this G protein is not sufficient for Ca2+ release. Increasing intracellular cyclic AMP levels by treatment with phosphodiesterase inhibitors IBMX and rolipram, does not mimic the response or potentiate the response of adrenergic agonists. Additionally, treatment with PKA inhibitors H-89 and KT5720 had no effect on the Ca2+ signal, and treatment with the cAMP analogue 8-bromo-cAMP, which selectively activates PKA, did not trigger a Ca2+ response. These results strongly suggest that AC, cAMP and the cAMP effector PKA are not involved in this signaling pathway. Together these findings indicate that activation of β2-AR leads to an increase in cytoplasmic [Ca2+] by a previously unrecognized signaling pathway. The discovery of a new signaling pathway for β2-AR could have serious implications in the use of beta agonists and blockers for the treatment of disease. Contributors: Galaz-Montoya, Monica; Rodriguez, Gustavo; Lichtarge, Olivier and Wensel, Theodore 89 BAYLOR COLLEGE OF MEDICINE LACTOBACILLUS REUTERI EXERTS ANTI-INFLAMMATORY EFFECTS VIA CONVERSION OF L-HISTIDINE TO HISTAMINE AND ACTIVATION OF HISTAMINE RECEPTOR 2 Chunxu Gao Department of Molecular Virology & Microbiology Advisor: James Versalovic, M.D./Ph.D.-Department of Pathology & Immunology Supplementation with probiotic Lactobacillus reuteri strains that naturally colonize the gut of mammals has been effective at ameliorating intestinal inflammation in rodent colitis models, but the underlying mechanisms are unknown. Recent pangenomic studies showed that L. reuteri strains with anti-inflammatory properties contain a complete hdc gene cluster which is responsible for synthesis and secretion of histamine, indicating a potential role for histamine in alleviation of inflammation. L. reuteri strain ATCC PTA 6475 which contains an intact hdc gene cluster was found to suppress TNF production in activated THP-1 cells through the production of histamine and activation of histamine receptor 2 (H2R). Targeted mutagenesis of the hdc genes resulted in diminished anti-TNF activity and loss of histamine production, indicating the anti-TNF activity of histamine in vitro. Based on these studies, we hypothesize that hdc+ L. reuteri attenuate inflammation in vivo via histamine production and activation of H2R. Using a trinitrobenzene sulfonic acid (TNBS)-induced mouse model of colitis, we showed that L. reuteri 6475 administration with a dose of 5×109 CFU once per day for seven days protected eight-week female BALB/c mice against colitis, as indicated by significantly decreased weight loss, colonic injury graded by the Wallace score and serum amyloid A protein concentrations compared to the mice receiving media control. Positron emission tomography (PET) imaging also showed that L. reuteri 6475 significantly reduced the uptake of [18F]fluorodeoxyglucose ([18F]FDG) in the colon, indicating attenuation of colonic inflammation by L. reuteri. RT-qPCR experiments revealed that TNBS instillation induced IL-6 and IL-1β gene expression in the colon of colitic mice compared to healthy mice while L. reuteri treatment decreased the gene expression of these proinflammatory cytokines. Further experiments found that the hdcA mutant of L. reuteri 6475 which failed to produce histamine showed diminished ability to attenuate colitis compared to the wild type strain. Moreover, H2R was detected in the mouse colon by immunohistochemistry and blocking H2R with its specific antagonist ranitidine diminished the anti-inflammatory ability of L. reuteri 6475 while blocking H1R with its specific antagonist pyrilamine did not affect the anti-inflammatory activity of this probiotic strain. In addition, feeding mice with a histidine-free diet diminished L. reuteri’s ability to attenuate colitis compared to the regular diet which contains 0.4% of histidine. These combined investigations indicate that L. reuteri 6475 attenuates experimental colitis via histamine production, and provide important insights into understanding the molecular mechanisms underlying probiotic immunomodulation. Contributors: Gao, Chunxu; Spinler, Jennifer; Major, Angela; Jackson, Vanessa; Lugo, Monica; Versalovic, James 90 2014 GRADUATE STUDENT SYMPOSIUM VESTIBULAR BEHAVIORAL THRESHOLDS FOR YAW ROTATION DISCRIMINATION IN RHESUS MACAQUES Courtney Dawn Garcia Department of Neuroscience Advisor: Dora Angelaki, Ph.D.-Department of Neuroscience The vestibular system provides for perception of movement, orientation in space, and motion related behaviors. In relation to self-motion perception, human yaw rotation velocity thresholds have been previously measured to be 0.7 deg s-1 at 0.5 Hz (Merfeld et al. 2008). However, the available data on neuronal direction detection thresholds for semicircular canal afferents in macaques have been quite disparate. For example, average neuronal direction detection thresholds for semicircular canal afferents were reported to be ~3.75 deg s-1 for regular afferents and ~8.25 deg s-1 for irregular afferents by Sadeghi et al. (2007). In contrast, Yu et al. found average direction detection thresholds for semicircular canal afferents to be much lower at 2.17 deg s-1 ± 1.32 (n=85) for regular afferents and 1.91 deg s-1 ± 1.49 (n=32) for irregular afferents (2013 SFN Abstract). Both studies used sinusoidal stimuli without simultaneously measuring behavioral thresholds. Furthermore, both studies compared neural thresholds in macaques with human perceptual thresholds. Thus, it is possible that macaque and human perceptual thresholds are different. Here we set out to investigate how macaque perceptual yaw rotation velocity detection thresholds compare with human thresholds. We measured macaque thresholds using transient smooth trajectories that followed a 1s Gausian velocity profile (peak velocity: 1.3-30 deg s-1). Two rhesus macaques were trained to perform a two-alternative-forced-choice task, in which they reported leftward or rightward rotation by making a saccade to one of two targets presented at motion end. Psychometric functions then determined direction detection thresholds over time. Mean yaw rotation detection thresholds were initially 8.59 deg s-1 for monkey H and 6.85 deg s-1 for monkey I. However, after continued training, perceptual thresholds in both monkeys decreased to 3.31 deg s-1 for monkey H and 1.48 deg s-1 for monkey I. In order to directly compare neuronal and perceptual detection thresholds for yaw rotation, we are currently measuring canal afferent thresholds simultaneously with perceptual detection thresholds in both animals. This work was supported by NIDCD DC04260. Contributors: Garcia, Courtney D.; Yu, Xiong-Jie; Liu, S.; Dickman, J. David; Angelaki, Dora E. 91 BAYLOR COLLEGE OF MEDICINE DEVELOPMENT OF CHAGAS CARDIAC MANIFESTATIONS AMONG TEXAS BLOOD DONORS Melissa Nolan Garcia Clinical Scientist Training Program Advisor: Kristy Murray, Ph.D.-Epidemiology Chagas disease, infection with the parasite Trypanosoma cruzi, has recently been identified as an important emerging parasitic disease in the United States. To describe the cardiac abnormalities among T. cruzi positive blood donors in Southeast Texas, we performed a pilot study of donors who had screened positive from 20072012. This one-time assessment included: 1) a questionnaire to evaluate source of infection, cardiac symptoms and health co-morbidities, 2) an electrocardiogram, 3) an echocardiogram if ECG was abnormal, and 4) measurement of a high-sensitivity troponin T biomarker. Of those with confirmed infection, 41% (7/17) had an electrocardiographic abnormality consistent with Chagas cardiomyopathy. In addition, 36% (6/17) were suspected to be locally acquired cases. Biomarker high-sensitivity troponin T serum levels increased with cardiac severity. Cardiologists should consider Chagas disease in their differential for patients who may have clinically compatible electrocardiogram changes or cardiomyopathy, even if the patient has no history of residing in a Chagas-endemic country Contributors: Garcia, Melissa; Murray, Kristy; Hotez, Peter; Rossman, Susan; Gorchakov, Rodion; Woc-Colburn, Laila; Bottazzi, Maria Elena; Rhodes, Charles; Ballantyne, Christie; Aguilar, David. 92 2014 GRADUATE STUDENT SYMPOSIUM TRANSCRIPTIONAL REGULATION OF MESENCHYMAL STEM CELL DIFFERENTIATION POTENTIAL WITH AGE Sandy Lynn Garno Integrative Program in Molecular and Biomedical Sciences Advisor: Weiwei Dang, Ph.D.-Department of Molecular & Human Genetics Human mesenchymal stem cells (MSCs) are easily isolated, pluripotent adult stem cells capable of differentiating into various lineages including (but not limited to) adipocytes, chondrocytes, and osteoblasts. These characteristics are ideal for stem cells in regenerative medicine, thus MSCs are widely investigated for their therapeutic potential. Regeneration is a critical topic when it comes to aging, as aging is defined by widespread deterioration and dysfunction of normal anatomy and physiology. MSCs have the potential to regenerate many structures lost or damaged as aging occurs, and also to treat any condition that requires the replacement of damaged cells. The problem is that MSCs isolated from older individuals have reduced differentiation potential for all lineages, and all MSCs are subject to a limited number of expandable divisions in vitro. This creates difficulty when attempting to collect MSCs for autologous use from aged individuals, as MSCs can only be safely isolated in small amounts. Thus, it is ideal to investigate alterations to differentiation potential that occur with aging of human mesenchymal stem cells in order to maximize their usefulness in regenerative medicine. It is well known that changes to transcription accompany aging in eukaryotes. We will perform RNA-Seq analysis on young and old mesenchymal stem cells in order to determine exactly how transcription is changing with age in these cells. Processes expected to show significant alteration are intragenic cryptic transcription, non-coding RNA regulation, and alternative splicing, among others. We will target these processes in old MSCs via knockdown or overexpression of critical enzymes in order to mimic the transcription profile of young MSCs. By doing this, we intend to restore the declining differentiation potential in old MSCs to the robust differentiation potential of young MSCs. Ultimately this will allow aged individuals access to the full power of mesenchymal stem cell regenerative medicine that is available to young individuals. Contributors: Garno, Sandy; McCauley, Brenna; Dang, Weiwei 93 BAYLOR COLLEGE OF MEDICINE H3K4ME3-PROMOTED HISTONE ACETYLATION IN ER -MEDIATED TRANSCRIPTION Leah Ashley Gates Department of Molecular & Cellular Biology Advisor: Bert O'Malley, M.D.-Department of Molecular & Cellular Biology Estrogen receptor alpha (ER ) directs expression of its target genes by binding particular DNA elements and recruiting different transcriptional coregulators (CoRs) in a hormone-stimulated fashion. In addition to being regulated by ligands, ER activity is also modulated through the chromatin environment by particular interactions with specific CoRs. Our lab is interested in dissecting the mechanisms of transcriptional regulation, which is essential for deepening our knowledge of basic molecular biology and may aid in the identification of potential therapeutic targets for hormone-driven cancers. Epigenetic modifications such as histone marks can promote the recruitment or dismissal of CoRs that directly influence the transcriptional outcome. H3K4 trimethylation (H3K4me3) is a histone mark found at actively transcribing gene promoters and is correlated with histone acetylation. The presence of H3K4me3 stimulates acetylation selectively at H3K9 (H3K9Ac) and at several H4 lysines, including H4K5 (H4K5Ac) on recombinant nucleosomes in vitro with ER . By performing biotinylated histone peptide pulldowns with HeLa nuclear extract followed by mass spectrometry and immunoblotting, we now report that the dual modification H3K4me3K9Ac recruits the super elongation complex (SEC) as compared to the H3K4me3 peptide alone. Additionally, H4K5Ac recruits the BET family members Brd2, Brd3, and Brd4 compared to the unmodified H4 peptide. Based on this data, we propose a model in which H3K4me3 is required for transcription initiation and promotes acetylation of H3K9 and H4 lysines through the recruitment of histone acetyltransferases. These acetylation marks can then recruit elongation machinery via Brd proteins to H4 acetyl marks and the SEC to H3K9Ac. Next, we will determine the functional relevance of H3K9Ac and H4K5Ac in ER -mediated transcription by using a combination of biochemical and cell-based functional assays. Importantly, these studies will advance our understanding of how ER and epigenetic marks can regulate transcription at the level of RNA Pol II elongation in response to hormonal stimulation. Contributors: Leah A. Gates, Qin Feng, Mark T Bedford, Sung Yun Jung, Jun Qin, Charles E. Foulds, and Bert W. O’Malley 94 2014 GRADUATE STUDENT SYMPOSIUM LRP6 CAUSES A NOVEL HUMAN DEVELOPMENTAL DISEASE Violet Gelowani Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D.-Department of Molecular & Human Genetics A patient family entered the clinic displaying a novel combination of clinical features consisting of glaucoma, short stature, microphthalmia, bracydactyly, and choroidal effusions. We were interested in finding the genetic cause for this novel syndrome. Using whole exome sequencing and pedigree segregation analysis we identified the patients to be heterozygous for a mutation in LRP6. LRP6 is known to be involved in the WNT signaling pathway. To test whether the patient mutation disrupts WNT signaling we performed in vitro luciferase reporter assay and saw that the mutation reduced WNT signaling. We performed morpholino knockdown and rescue experiments in zebrafish and found that the mutant LRP6 allele failed to rescue the skeletal defects and the microphthalmia phenotype of LRP6 knockdown zebrafish. This observation validated the mutant allele of LRP6 as the genetic cause for the mutant phenotype in patients. LRP6 heterozygouse null mice were found to have elevated ocular pressure, optic nerve malformations, and skeletal defects confirming that LRP6 is haploinsufficient. These results suggest that LRP6 is a novel human disease gene, and that our newly developed mouse model may serve as a disease model for this novel syndrome and for glaucoma. Contributors: Gelowani, Violet; Jacques Zaneveld; Xu Mingchu; Liu, Wei; Wang, Hui; Wang, Xia; Wang, Feng; Salvo, Jason; Jiang, Lichun; Li, Yumei; Guy, Hughes; Chen, Rui. 95 BAYLOR COLLEGE OF MEDICINE LOSS OF miR-148 ENHANCES OVARIAN CANCER GROWTH AND METASTASIS BY PROMOTING MTMR9 EXPRESSION Triparna Ghosh-Choudhury Program in Translational Biology & Molecular Medicine Advisor: Matthew Anderson, M.D./Ph.D.-Department of Obstetrics & Gynecology Creighton Edwards, M.D.-Department of Obstetrics & Gynecology INTRODUCTION: MicroRNAs (miRNAs) are endogenous, non-coding RNA transcripts that play a critical role in regulating diverse patterns of gene expression. Although altered miRNA expression has been previously described in ovarian cancer, the role of individual miRNAs in this disease remains poorly understood. METHODS: Level 3 data from the Cancer Genome Atlas Consortium (TCGA) were interrogated by using Kaplan-Meier statistics to test for association between levels of individual miRNAs and outcome demographics. miRNA mimics were transfected into established ovarian cancer cell lines (HeyA8, SKOV3ip1, OVCAR8) using Lipofectamine (Invitrogen). Proliferation and apoptosis were measured using MTS and Caspase Glo 3/7 assays (Promega). Western blot and quantitative real time PCR were used to validate gene expression. In vivo studies were done in nude mice injected intraperitoneally with RFP (+) OVCAR8 cells. Statistical significance was calculated using two-tailed T-tests. RESULTS: Our analyses indicate that 3 of the 10 miRNA transcripts most robustly associated with ovarian cancer outcome (miR-148a, miR-148a* and miR-148b) share a common seed sequence. Transfection of both TP53-null SKOV3ip1 and TP53-mutated OVCAR8 ovarian cancer cells with mimics for any of these three miRNAs significantly inhibited proliferation, enhanced apoptosis, inhibited migration and invasion. To identify targets, we recognized correlations between patterns of gene and miRNA expression using Lasso analyses with L1-normalization, after which potential miR-148 targets were screened using established target prediction algorithms (Targetscan, RNA22). These analyses pinpointed MTMR9, a poorly understood cofactor important for regulating phosphoinositide metabolism, as a key target for miR-148 in ovarian cancer. Overexpression of miR-148 results in decreased expression of MTMR9 and decreased catalytic activity of MTMR6 and MTMR8 contributing to apoptosis of cells. Direct knockdown of MTMR9 using siRNAs reduces proliferation, enhances apoptosis and decreases rates of migration and invasion in OVCAR8 and HeyA8 cells. Treatment with siMTMR9 liposomes in vivo reduces ovarian cancer growth and metastasis. Control mice were treated with siNT and had significant metastasis throughout abdomen and diaphragm. KI67 stainings of cross sections of dissected tumors showed a large number of control cells to be proliferative where as significantly lower amounts were seen in siMTMR9 mouse cross sections. CONCLUSION: Our findings indicate that the frequent loss of miR-148 miRNAs promotes ovarian cancer growth and metastasis by enhancing MTMR9 expression. These findings not only validate the unique approach we have adopted for screening high throughput data sets for relevant miRNA targets, but also implicate novel aspects of phosphoinositide metabolism in ovarian cancer biology. Contributors: Ghosh-Choudhury, Triparna; Wan, Ying-Wooi; Liu, Zhandong; Anderson, Matthew L 96 2014 GRADUATE STUDENT SYMPOSIUM LETHAL LOVE: BIOENGINEERING A BACTERIUM WITH ANTIMICROBIAL ACTIVITY BY EXPLOITING BACTERIAL CONJUGATION TO DELIVER TOXIC ELEMENTS Mary Elizabeth Girard Department of Molecular Virology & Microbiology Advisor: Christophe Herman, Ph.D.-Department of Molecular & Human Genetics Bacterial infections resistant to antibiotics are an alarming threat as the incidence numbers continue to rise, leaving us in need of new antimicrobial methods. We propose to engineer an antimicrobial bacterium, or rather, a bacterium able to kill other bacteria. With this novel antimicrobial approach, we highjack bacterial conjugation as a platform to deliver toxic elements to bacteria. Conjugation is the horizontal transfer of genes between bacteria via a conjugative plasmid through a sex pilus from the donor to the recipient bacterium. This process is a very efficient mode of gene transfer and is quite promiscuous, theoretically allowing us to target a variety of bacteria. Additionally, with the current methodologies of bacterial genetic engineering, we are able to construct a variety of conjugative plasmids encoding toxic genes of choice with ease. For the toxic component of our antimicrobial platform, we envision utilizing either toxins from bacterial toxin-antitoxin systems or the Cas endonuclease from bacterial CRISPR/Cas systems. Both options are highly advantageous, untapped resources for potential antimicrobials. The TA system toxins are known to be quite toxic to their bacterial hosts in absence of the cognate antitoxin, and have thus often been suggested as putative alternatives to antibiotics. Here, the engineered bacterium carrying the toxin-encoding conjugative plasmid mates with a recipient, transferring and expressing the toxin, killing the recipient bacterium. For our other choice toxic element, the CRISPR/Cas systems are also known to be lethal to the host bacterium if targeted to cleave bacterial host DNA. The CRISPR system is very exciting as a putative antimicrobial as the Cas nuclease can be targeted to cleave very specific SNPs of DNA, and this specificity allows us to target its lethal activity to bacterial pathogen SNPs. In this case, the engineered bacterium carries a conjugative plasmid encoding the Cas nuclease targeted to a desired pathogen SNP. Upon mating, the Cas nuclease specifically cleaves the pathogen DNA, thereby only killing the desired pathogen. Taken together, this antimicrobial platform combines the toxic elements of TA systems and CRISPR/Cas systems with conjugation as a delivery mechanism, transferred by a harmless bacterial carrier. We are initially testing this antimicrobial strategy by performing mating assays between our engineered strains against nonpathogenic lab strains of E. coli, scoring for recipient cell viability. Preliminary data shows this method is effective against E. coli, and next, we will be performing mating and viability assays against a variety of bacterial pathogens. In the future, this work may lead to the development of this system for application in biotherapeutics or biocontainment. Contributors: Girard, Mary; Herman, Christophe 97 BAYLOR COLLEGE OF MEDICINE THE ROLE OF PREOPERATIVE TRANSURETHRAL BIOPSY OF THE PROSTATIC URETHRA FOR EVALUATION OF THE URETHRAL MARGIN AT RADICAL CYSTOPROSTATECTOMY Guilherme Godoy Clinical Scientist Training Program Advisor: Seth Lerner, M.D.-Department of Urology Introduction and Objective: We have used the prostatic urethra biopsy as part of our preoperative management and clinical staging prior to radical cystoprostatectomy (RC). Our hypothesis is that a negative preoperative transurethral resection (TUR) biopsy of the prostatic urethra accurately predicts a final negative apical urethral margin, safely replacing an intraoperative frozen section for urethral preservation and orthotopic neobladder utilization. Methods: TUR biopsies of the prostatic urethra adjacent to the verumontanum at 5 and 7 o’clock were performed on 272 male patients that underwent RC at our institution between 1987 and 2013. Pathology results of biopsy were correlated with those of final apical urethral margin status of the RC specimen, and whether or not the surgeon had done an intraoperative frozen section of the urethral margin and performed a primary or secondary urethrectomy. Whole mount step section of the entire prostate was performed and examined microscopically in the majority of cases. Results: Of the 272 patients, 200 (74%) had a negative and 72 (26%) had a positive biopsy. The overall final positive apical urethral margin rate in the RC specimen in this series was 2.2% (6/272 ). Five of those patients underwent a primary or secondary urethrectomy. In one patient with a positive final margin, a urethrectomy was not performed as the frozen section of apical urethra had been negative despite the finding of an invasive urothelial carcinoma on the preoperative TUR biopsy. The negative predictive value of a TUR biopsy was very high (99.5%) with a very low positive predictive value (PPV) for the final pathological margin status. Conclusion: The apical urethral positive margin rate is very low in patients undergoing RC. A positive TUR biopsy has a low PPV for a positive final apical margin (7%). A negative preoperative TUR biopsy of the prostatic urethra can reliably predict a negative final margin and may obviate the need for intraoperative frozen section evaluation. This information facilitates preoperative counseling patients about the feasibility for orthotopic neobladder reconstruction of the bladder after RC. Contributors: von Rundstedt, Friedrich-Carl; Godoy, Guilherme; Shen, Steven; Lerner, Seth P. 98 2014 GRADUATE STUDENT SYMPOSIUM EFFECTS OF IN UTERO GLUCOCORTICOID (GLC) EXPOSURE ON SKELETAL MUSCLE SATELLITE CELL POPULATION IN THE FETAL RAT Ganga Gokulakrishnan Clinical Scientist Training Program Advisor: Steven Abrams, M.D.-Department of Pediatrics BACKGROUND: Antenatal GLC exposure is known to accelerate the maturation of several tissues in the fetus, but whether it is also true for the skeletal muscle is unknown. It has been shown that in utero GLC exposure results in decreased skeletal muscle mass and decreased protein content independent of its effect on maternal food intake. Satellite cells are mononuclear myogenic cells responsible for postnatal skeletal muscle growth and mass. However the effects of in utero GLC exposure on satellite cells in the perinatal period are unknown. OBJECTIVE: To test the hypothesis that the decrease in skeletal muscle mass following in utero GLC exposure is a result of a decrease in satellite cell population. The aims were to determine if fetal exposure to GLC alters 1) myofiber cross sectional area (CSA) 2) myonuclear accretion 3) Satellite cell population. DESIGN/METHODS: Three groups (n=7/group) of timed-pregnant SpragueDawley rats were studied: Control (CON): ad libitum food intake; Dexamethasone (DEX): ad libitum food intake; dexamethasone (1 mg/L drinking water ad lib from embryonic day (ED) 13 to 21). Pair-fed (PF): pair-fed to DEX group from ED 13 to 21; no DEX. Three injections of 5-bromo-2′-deoxyuridine (BrdU) were given 12 hours apart, 36 hours prior to retrieval. On ED 22 (term), fetuses were retrieved, dissected and muscles collected. Quadriceps muscles were analyzed for the following: Muscle fiber cross sectional area (CSA), myonuclear number, myonuclear proliferation and satellite cell number using immunofluorescence techniques. RESULTS: The mean CSA was different among all 3 groups with DEX group being the smallest (P < 0.01). The myonuclei/fiber ratio was 0.79, 0.55 and 0.68 in the CON, DEX and PF groups respectively with DEX significantly different from CON (P = 0.014). BrdU labeling of myonuclei was similar among all groups. Satellite cell/myofiber ratio was 0.13, 0.07, 0.16 in CON, DEX and PF groups respectively with all 3 groups being significantly different from each other (P< 0.001). The percentage of satellite cells noted within the myofiber was similar among all groups. CONCLUSIONS: In utero GLC exposure resulted in a decrease in fiber size as evidenced by cross sectional area. The decrease in myonuclear accretion noted in the DEX group can be explained by the decrease in satellite cell population secondary to impaired proliferation. No impairment in satellite cell fusion was noted. Apoptosis of myonuclei could also have contributed to decrease in myonuclear accretion. These findings suggest a possible permanent reduction in skeletal muscle mass in adulthood. Contributors: Gokulakrishnan, Ganga; Chang, Xiaoyan; Fiorotto, Marta L. 99 BAYLOR COLLEGE OF MEDICINE TELOMERE LENGTH AND TELOMERASE-ASSOCIATED GENE VARIANTS IN PEDIATRIC ACUTE MYELOID LEUKEMIA (AML) Maria Monica Gramatges Clinical Scientist Training Program Advisor: Alison Bertuch, M.D./Ph.D.-Department of Pediatrics Introduction: Constitutional mutations in telomere maintenance genes are associated with short telomeres and a spectrum of disorders including dyskeratosis congenita (DC). DC confers a 90% lifetime risk for bone marrow failure, a 200-fold risk for acute myeloid leukemia (AML), a 2500fold risk for myelodysplastic syndrome (MDS), and is associated with chemosensitivity in affected individuals. Constitutional defects in telomere maintenance genes are also enriched in patient cohorts with various hematologic malignancies, including AML. Given this association, we investigated telomere length and the incidence of constitutional telomerase variants in de novo pediatric AML cases and their role in therapy-related adverse events (AE’s). We hypothesized that variants would be (1) more frequent in AML cases compared with controls, (2) associated with characteristics of telomere biology disorders, and (3) in addition to telomere length, would further characterize AML cases with specific AE’s. Methods: We sequenced the exons and flanking intronic regions of the telomerase-related genes TERT, DKC1, hTR, and TINF2, in a local pediatric AML/MDS cohort (n=104), a Children’s Oncology Group (COG) AML cohort (n=115), and local controls racially and ethnically matched to our AML/MDS cohort (n=254). We blindly reviewed medical records in the local AML cohort for six characteristics suggestive of DC. For the COG cohort, we compared the number of variants and remission telomere content (TC), measured by qPCR, in subjects with expected blood count recovery after chemotherapy (n=62) to those with significantly prolonged recovery times (n=53). A relationship between variants, TC, and specific grade 3 or 4 AE’s was also explored. Results: In the local AML/MDS cohort, the number of novel or rare telomerase variants significantly exceeded what was observed in both population databases and local controls (p<0.0001). Retrospective medical record review demonstrated a significant association between the number of DC features present and presence of a novel or rare variant (p=0.04). Within the COG AML cohort, a significant association was observed between less TC at the end of initial chemotherapy and delays in time to blood count recovery in later cycles of chemotherapy (p=0.03). Conclusions: Novel and rare constitutional telomerase variants are enriched in pediatric AML/MDS cases compared with controls. These variants are significantly associated with clinical features of telomere biology disorders. In addition, less TC at end of AML induction is associated with significant delays in time to blood count recovery in later cycles of AML chemotherapy, suggesting that short telomeres are indicative of an underlying impairment in capacity for marrow stem cell reconstitution. Contributors: Gramatges, MM; Sasa, G; Gerbing R; Alonzo T; Gamis A; Meshinchi S; Plon, SE; Bertuch, AA. REGULATION OF E2F1 TRANSCRIPTIONAL ACTIVITY BY SUMOYLATION 100 2014 GRADUATE STUDENT SYMPOSIUM Joshua Daniel Graves Integrative Program in Molecular and Biomedical Sciences Advisor: Weei-Chin Lin, M.D./Ph.D.-Department of Medicine The E2F family of transcription factors consists of crucial proteins in higher eukaryotes with functions including cell proliferation, apoptosis, senescence, and metabolism. How a handful of proteins, and in some cases the same family member (e.g. E2F1), can carry out such opposing functions is an unresolved paradigm. Thus far, specifying and directing activity has been largely attributed to the numerous posttranslational modifications found on E2Fs. I have demonstrated that the small ubiquitinrelated modifier 2 (SUMO2) modifies E2F1-5. Due to the high frequency of perturbations in the Rb/E2F1 pathway in all cancer types, E2F1 is the main focus of further study. More than 95% of SUMO2 conjugation on E2F1 requires two lysines of E2F1. Overexpressing Ubc9, the sole E2 conjugating enzyme in the SUMO pathway, greatly induces modification dependent on these two lysines. When using an E2F1specific luciferase reporter assay, Ubc9 overexpression reduced E2F1 transcriptional activity by up to 60%. Additionally, E2F1 point mutants for sumoylated lysines significantly rescued this Ubc9-mediated transcriptional repression. This suggests that sumoylation specifically on E2F1 is responsible for the observed decrease in activity. Furthermore, reduction of transcriptional activity by Ubc9 has been recapitulated at endogenous E2F1 target genes by qRT-PCR. I hypothesize that SUMO2 modifying E2F1 serves as an additional means to inhibit specific E2F1 transcriptional activity, resulting in changes to cell cycle progression, apoptosis, senescence, or metabolism. E2Fs1-3 have essential roles in promoting cell growth, as is exemplified by the fact that a triple knockout mouse is embryonic lethal. Consequently, the pathways that inhibit their activity are highly mutated in all cancer types. By further understanding how SUMO2 addition to E2F1 inhibits its transcriptional activity, as well as how this mechanism is perturbed in cancer, new opportunities could be presented as pharmacologic targets to inhibit E2F1 dependent cell growth. Moreover, since SUMO2 has also been observed on E2Fs2-3, the findings of this and future work might yield a means to shut down all E2F dependent growth in cancer. Contributors: Graves, Joshua; Lin, Weei-Chin 101 BAYLOR COLLEGE OF MEDICINE NOVEL NANO-ANTIOXIDANT TREATMENT IN A DIETARY MODEL OF NONALCOHOLIC FATTY LIVER DISEASE Deric Maurice Griffin Program in Translational Biology & Molecular Medicine Advisor: Robia Pautler, Ph.D.-Department of Molecular Physiology & Biophysics Stephanie Abrams, M.D.-Department of Pediatrics Non-alcoholic fatty liver disease (NAFLD) is a spectrum of disorders, encompassing hepatosteatosis (fat in the liver), non-alcoholic steatohepatitis (NASH), and cirrhosis. NAFLD’s incidence has been increasing alongside obesity in the U.S. and is becoming a leading cause for liver transplant in the United States. Oxidative stress has been heavily implicated in disease pathology, but its full role along with an effective antioxidant therapy is still unclear. Here we use a methionine-choline deficient diet (MCDD) model of NAFLD, which captures the liver phenotype of the disease and leads to decreased production of the antioxidant glutathione (GSH). We used the MCDD model to define the effect of overexpressing the endogenous antioxidant, superoxide dismutase 2 (SOD2), which breaks down the reactive oxygen species (ROS) known as superoxide radical. We also sought to evaluate the efficacy of pegylated-hydrophilic carbon clusters (PEG-HCCs), a potent bioengineered nano-antioxidant, on NAFLD progression. In the context of MCDD, mice overexpressing SOD2 showed increases in oxidative stress markers and no improvement in markers of liver damage, while PEG-HCC treated mice had significant decreases in oxidative stress markers in addition to displaying trends towards less liver damage. We predict that these observed differences are partially due to an increase in hydrogen peroxide, which is a ROS by-product of SOD2 activity. We suspect that this increase in hydrogen peroxide due to increased SOD2 activity, along with the restriction of GSH production, led to increased oxidative stress in MCDD fed SOD2 overexpression mice. This effect was not observed in the PEG-HCC treated group and we believe this to be due to the PEG-HCCs ability to scavenge oxygen radicals without the producing ROS byproducts, thus highlighting their efficacy as a potential therapeutic. Contributors: Griffin, Deric M.; Criss II, Zachary K.; Tour, James; Pautler, Robia 102 2014 GRADUATE STUDENT SYMPOSIUM IDENTIFICATION OF ONCOGENIC DRIVERS IN AN IN VIVO LUNG CANCER MODEL Caitlin Grzeskowiak Department of Molecular & Human Genetics Advisor: Kenneth Scott, Ph.D.-Department of Molecular & Human Genetics Discoveries in cancer genomics have revolutionized advancements in personalized medicine. Through large-scale sequencing efforts, many critical genetic alterations have been identified to drive tumor initiation and metastasis. Several consortiums have been established to analyze biopsied patient tumors to identify potential oncogenic mutations. However, these mutations often still require extensive validation efforts as they are usually diluted in these studies with passenger mutations in late stages of disease progression. Alternatively, large scale in vitro screening efforts using cell culture assays do not faithfully recapitulate hallmarks of spontaneous tumors presented in the clinic. To combat these challenges, in vivo models are often used to validate oncogenic drivers. We have developed an in vivo screening platform to identify oncogenic drivers of tumor initiation and metastasis in a Genetically-Engineered-Mouse (GEM) lung cancer model. In this approach, GEM models provide the proper microenvironment, yet drivers are readily identified with previously generated barcoding technology developed in our lab. Cre recombinase is activated in LSL-KRASG12D mice while simultaneously expressing barcoded potential drivers. Using a pool of candidate oncogenes, we are able to readily identify the genetic drivers of metastasis and tumor initiation by sequencing for barcode enrichment. To demonstrate the power of this method, dominant negative forms of p53 were expressed with Cre recombinase in the lung. Six months after intubation, mice were sacrificed and tumor analysis showed obvious primary tumors with metastases, confirming this approach as powerful way of identifying not only drivers of primary tumors, but also metastatic determinants. Therefore, we are able to reproducibly activate oncogenic KRAS with proper genetic context in addition to expressing candidate oncogenic drivers exclusively in the lung. Once established, these models can be used directly as a preclinical model for testing therapeutics targeted to specific mutations that arise in individual patient tumors. Contributors: Wu, Ping, Scott, Kenneth 103 BAYLOR COLLEGE OF MEDICINE MAINTAINING DISTANCE: THE E3 UBIQUITIN LIGASE ARIADNE-1 CONTROLS NUCLEAR POSITIONING DURING MUSCLE DEVELOPMENT IN DROSOPHILA Nele Haelterman Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics Proper muscle physiology is central to allow an organism to move and is thus essential for every organism’s survival. Multiple fatal human diseases, such as Spinal Muscular Dystrophy, are caused by muscle dysfunction. To identify novel genes required for normal muscle development and function, our lab performed a forward genetic mutagenesis screen on the Drosophila X-chromosome. From this screen, we identified several mutations in ariadne-1 (ari-1), which encodes a conserved Ring-In Between-Ring (RBR) E3 ubiquitin ligase and hence plays a role in maintaining protein homeostasis. Under normal circumstances, nuclei are evenly spaced within the multinucleated muscle fiber and this positioning is required to maintain proper muscle function. For instance, abnormal myonuclear positioning is seen in human patients with Central Nuclear Myopathies. We found that loss of ari-1 leads to nuclear clustering within larval body wall muscles, a phenotype that is very rarely observed in wild type animals. It is well established that the cytoskeleton plays a critical role in nuclear positioning, but less is known about the proteins that connect the nucleus to the cytoskeleton. Recently, the KASH-domain containing proteins Msp-300 and Klarsicht were found to form a complex that links the outer nuclear membrane to both the microtubular and the actin cytoskeleton. Interestingly, the subcellular localization of Msp-300 is severely affected in ari-1 mutants. In addition, the microtubular network appears fragmented upon loss of ari-1, whereas the actin cytoskeleton remains intact. One way to interpret our data is that Ari-1 directly regulates microtubule dynamics, the malfunction of which affects nuclear migration. However, a similar microtubular phenotype was observed in Msp-300 mutants that lack the KASH-domain. It is therefore possible that, once properly placed, Msp-300 is required to maintain the astral microtubular network that surrounds nuclei. Ari-1 would then control the localization, and hence function, of Msp-300, possibly by mono-ubiquitinating the protein. If our hypothesis holds true, Ari-1 would be the first known protein to fine-tune Msp-300’s function in controlling nuclear positioning and sustaining muscle function. Contributors: Haelterman, Nele*; Tan, Kai Li*; Jaiswal-Nagarkar, Sonal; Lee, Pei-Tseng; Bellen, Hugo 104 2014 GRADUATE STUDENT SYMPOSIUM DISCOVERY OF CHIMERIC TRANSCRIPTS IN HEPATOBLASTOMA Katherine Haines Department of Molecular & Human Genetics Advisor: Sharon Plon, M.D./Ph.D.-Department of Pediatrics Introduction: Hepatoblastoma, a pediatric cancer of the liver, has few recurrent genomic abnormalities. Known alterations include Wnt pathway activation through somatic mutations of CTNNB1 or rare germline APC mutations, an unmapped recurrent t(1,4) translocation, and gains or losses of whole chromosomes; however, these abnormalities do not fully explain the molecular basis of this tumor. We hypothesize that gene fusion events may present a mechanism for oncogenesis in hepatoblastoma. Methods: We have used RNA sequencing (RNA-seq) to survey a cohort of 24 hepatoblastomas for chimeric transcripts. High quality RNA (RIN: 6.6-9.7) was extracted from fresh-frozen tissue and strand-specific, poly-A+ RNA-seq libraries were prepared for Illumina sequencing. Approximately 85 million paired-end reads (42.5 million fragments) of 2 x 100 bp length were generated per sample. Fusion transcripts were detected using deFuse (v.0.6.1) and/or TopHat-Fusion on FASTQ files. Candidate fusions were verified using BLAST and validated using RT-PCR. Results: A total of 143 in-frame chimeric transcripts were detected. Of all chimeric transcripts 60% represented likely read through events. These include one transcript, WDR81-SERPINF2, which was detected in 9 of 24 hepatoblastomas. This transcript results from a read through of the coding regions from two adjacent genes on chromosome 17. Additional filtering yielded 109 fusions across the tumor set that contain at least one gene partner from the COSMIC Cancer Gene Census with a known role in cancer. Conclusions: We have established an RNA-seq pipeline for identification of fusions in pediatric solid tumors. From this initial cohort of hepatoblastomas, 100% of samples displayed at least one chimeric transcript, alterations which would not have been observed with DNA sequencing or standard clinical assays. This pipeline has identified a novel read through, WDR81-SERPINF2 which is also detected in matched normal liver but not lymphoblastoid cell lines and therefore may be a liver-specific transcript of unknown significance. Contributors: Katherine Haines1, Angshumoy Roy2,3, Linghua Wang4, Pavel Sumazin2, Kyle R. Covington4, Donna M. Muzny1,4, Vijetha Kumar3, Dolores Lopez-Terrada2,3, David A. Wheeler1,4, Sharon E. Plon1,2,4 and D. Williams Parsons1,2,4 Departments of 1 Molecular and Human Genetics, 2Pediatrics, 3Pathology & Immunology, and 4Human Genome Sequencing Center, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX 77030 105 BAYLOR COLLEGE OF MEDICINE THE ROLE OF MAZ IN THE REGULATION OF GENITOURINARY DEVELOPMENT VIA MODULATION OF WNT SIGNALING Meade Haller Department of Molecular & Cellular Biology Advisor: Dolores Lamb, Ph.D.-Department of Urology Genitourinary (GU) birth defects comprise some of the most common yet least studied congenital malformations and range in severity from conditions such as undescended testes (cryptorchidism) and ventrally misplaced urethral meatus (hypospadias) to highly complex malformations such as bladder exstrophy epispadias complex (BEEC) and ambiguous genitalia. Congenital anomalies of the kidney & urinary tract (CAKUT) are also common and include phenotypes such as duplicated tract components, kidney agenesis, congenital hydronephrosis, horseshoe kidney, and cystic kidneys. Genomic aberrations such as copy number gains and losses can result in congenital malformations of the GU tract, among other organ systems. Genome wide array comparative genomic hybridization (aCGH) together with an extensive literature review allowed identification of genomic hotspots of GU development and delineated the smallest CNV regions of maximum overlap. CNV mapping revealed over 30 patients with GU defects harboring duplications or deletions in the syndromic genomic region, 16p11.2 – the most common known pathogenic gene dosage region in humans. The only gene covered collectively by all the mapped CNVs in patients with GU defects was MYC-associated zinc finger (MAZ). MAZ encodes a transcription factor with a similar consensus sequence to that of WT1, and is implicated in WNT signaling. In situ hybridization on mouse embryos and isolated mouse GU tracts defined the expression profile of Maz during development. In situ experiments showed robust staining of the GU tract including the kidneys, ureters, testes, bladder and genital tubercle. Immunohistochemistry confirmed this expression pattern at the protein level. Analysis of the lab’s cohort of genomic DNA from GU-abnormal patients and GU-normal fertile controls via CNV qPCR determined the frequency of MAZ CNVs in these groups. Validated by two independent probe sets, 6% of our GU abnormal cohort displayed CNVs in MAZ (n=258) compared to 0% in controls (n=57). As predicted based on its consensus sequence, knockdown or overexpression of MAZ in human embryonic kidney (HEK293) cells results in differential expression of several WNT pathway genes including DKK1/2, LRP5, FZD9/10, FRZB, DACT1, SFRP5, WNT2, WNT4, and WNT11. MAZ, once thought to be simply a housekeeping gene, encodes a dosage sensitive transcription factor that may play a key role in urogenital development and contribute to the congenital malformations of the 16p11.2 phenotype. Contributors: Haller, Meade; Lamb, Dolores J. 106 2014 GRADUATE STUDENT SYMPOSIUM DEVELOPMENT OF THE ARGONAUT CROSSLINKING IMMUNOPRECIPITATION (AGO-CLIP) PROTOCOL USING THE ILLUMINA TRUSEQ PLATFORM Mark Patrick Hamilton Department of Molecular & Cellular Biology/M.D.-Ph.D. Program Advisor: Sean Mcguire, M.D./Ph.D.-Department of Molecular & Cellular Biology MicroRNAs are a large class of noncoding RNA that suppress mRNA expression. MicroRNAs bind through their seven nucleotide “seed” region in complex with the Argonaute protein, typically to the 3’ untranslated region (3’UTR) of target mRNAs. A given microRNA may have hundreds of targets, and individual targets may only exist in specific cellular contexts. Due to this, the microRNA target spectrum in any given cell includes tens or hundreds of thousands of context specific interactions. Traditional methods of microRNA target interaction include analysis of conserved sequence in the 3’UTR and computation of biochemical binding properties between a microRNA’s seed and its putative target. While these methods are effective, they are often unable to distinguish non-canonical sites outside the, are only inferences of possible microRNA binding, and are not context specific. Argonaute crosslinking immunoprecipitation (AGO-CLIP) is a method to directly query global microRNA binding in the cell by sequencing the mRNA targets associated with the Argonaute-microRNA complex. AGO-CLIP technology is underutilized, with data existing in only 9 human cell lines and 2 mouse tissues due to the complexity of the AGO-CLIP method, which requires cDNA library preparation following Argonaute protein pull-down and radiolabeling of Argonaute-associated RNAs. Library preparation requires multiple gel isolations and takes several weeks to perform. Further, cDNA library preparations reported in current AGO-CLIP protocols require in-house-generated reagents inaccessible to most researchers. Finally, the RNA yield from Argonaut pulldown is low, requiring multiple rounds of PCR amplification that reduce the unique sequence composition of the AGO-CLIP library leading to inefficient sequencing results. Here we report the development of the AGO-CLIP protocol using the Illumina TruSeq Small RNA sample preparation kit. TruSeq library preparation removes the need to isolate multiple gels during library preparation saving time and preventing RNA loss. The TruSeq kit is readily accessible and requires no in-house generated reagents. Finally the TruSeq kit is rapid and contains multiplex compatible sequencing primers. We are able to rapidly generate successful AGO-CLIP libraries using the TruSeq RNA kit using less than 15 cycles of PCR amplification that exhibit improvements in percent of unique sequence and thus overall gains in sequencing efficiency. Use of the TruSeq kit promises to increase the speed and quality of AGO-CLIP library generation allowing an unprecedented view of microRNA binding in cells. Contributors: Hamilton, Mark P.; Rajapakshe, Kimal; Hartig, Sean M.; Coarfa, Cristian; McGuire, Sean E. 107 BAYLOR COLLEGE OF MEDICINE SV40 PERSISTENCE IN HUMAN LYMPHOCYTE CELL LINES Lindsay Jeanette Harrigal Integrative Program in Molecular and Biomedical Sciences Advisor: Janet Butel, Ph.D.-Department of Molecular Virology & Microbiology SV40 induces tumors, including lymphomas, in the Syrian golden hamster model and has been found in peripheral blood mononuclear cells (PBMCs) of immunocompromised monkeys. SV40 has been detected in human PBMCs, in human tonsils, and in non-Hodgkin lymphoma (NHL), suggesting that SV40 may be lymphotropic in humans. Other polyomaviruses have also been detected in lymphoid tissues and cells, including JCV, BKV, and MCV. We hypothesize that SV40 establishes a persistent infection in B cells that is rarely productive. Preliminary results using SV40-infected DG75 cells (human B cell line) suggest that viral genomes are maintained at low copy numbers and that SV40 early protein T-antigen (T-ag) is expressed. One goal is to determine the mechanism by which the SV40 genome is maintained in human B cells. We speculate cellular mitotic protein(s) could function in tethering T-ag-bound viral DNA to the chromosomes during cell division, allowing for proper segregation to each daughter cell. Genome tethering or a “piggyback” mechanism has been observed in other virus systems that establish persistent infections, including human papillomavirus (HPV) and Epstein–Barr virus (EBV). Currently, a mechanism for SV40 genome maintenance in human cell lines is unknown. T-ag binds to the origin of replication in the SV40 genome and is predicted to function in a tethering mechanism, allowing segregation of the viral genome during cell division. Transient DNA transfections were used to examine SV40 genome tethering in human B cell lines. The pEGFP-N1 vector was used to represent the SV40 genome in transient transfections with the human B cell line DG75. Cells were transfected with pEGFP-N1 and pCMV-CPC-T (encoding SV40 T-ag). The collected cells were analyzed by flow cytometry for EGFP expression. Through 72 hours post transfection, there were greater numbers of EGFP+ cells in the co-transfected (EGFP+CPC-T) DG75 cells compared to the vector control (EGFP only). The presence of SV40 T-ag led to an increase in EGFP expression detected by flow cytometry, which suggests T-ag promoted the retention of SV40-like DNA during B cell division; the observed “T-ag effect” was significant. To further investigate the genome tethering ability of T-ag, sitedirected mutagenesis was used to disrupt the binding site on T-ag for the SV40 origin of replication or binding sites on T-ag for cellular proteins that might act as linkers. These could theoretically block the tethering of SV40 DNA to the chromosome. Transient transfections will be performed with the SV40 T-ag mutants to evaluate changes in the “T-ag effect” on human B cells. Contributors: Harrigal, Lindsay J.; McNees, Adrienne L.; Butel, Janet S. 108 2014 GRADUATE STUDENT SYMPOSIUM A BIVALENT FUSION PROTEIN VACCINE FOR BROAD PROTECTION AGAINST RESPIRATORY SYNCYTIAL VIRUS Anne M Hause Program in Translational Biology & Molecular Medicine Advisor: Pedro Piedra, M.D.-Department of Molecular Virology & Microbiology Robert Atmar, M.D.-Department of Medicine Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections (LRTI) among infants and young children. The fusion (F) protein is a promising vaccine target because it is relatively conserved among RSV-A and RSV-B subgroups and induces neutralizing antibodies. Antigenic site II of the F protein is the targeted site of palivizumab, a monoclonal antibody that is used prophylactically for high-risk infants. Palivizumab is approximately 50% effective in preventing RSV related hospitalization and a small number of patients have palivizumab resistant viruses after therapy. Candidate vaccines, which are based on the historical GA1 genotype of RSVA, might provide variable levels of protection dependent on the subgroup and genotype of the infecting virus. Our objective is to develop a bivalent RSV-F vaccine that is composed of a consensus F protein for the RSV-A genotypes (Fa) and consensus F protein for the RSV-B genotypes (Fb). The G and F genes of RSV isolates (n=118) stored at BCM were sequenced. The F and G sequences of 118 isolates from GenBank were obtained. Together, these sequences were used to generate phylogenetic trees based on the G gene. The F sequences were then used to create a consensus sequence for each of the 7 RSV-A genotypes and 4 RSV-B genotypes identified in the phylogenetic trees. The genotype consensus sequences were then used to generate an overall consensus sequence of F protein for RSV-A (Fa) and RSV-B (Fb). We identified 169 RSV-A sequences (GA1=10, GA2=94, GA5=36, GA7=5, NA1=10, SAA1=2, ON=12) and 67 RSV-B sequences (BA=41, GB1=3, GB3=10, GB4=13). Among the genotype consensus sequences of each subclass, there is approximately 98% nucleotide conservation. However, this number decreases 5% when comparing contemporary to historical genotypes. There is approximately 90% amino acid and 80% nucleotide conservation between Fa and Fb. Amino acid variation is greatest in the signal peptide (64%), no annotation domain 2 (23%), antigenic site ø (17%), transmembrane domain (12%), and cytoplasmic domain (12%). Although the F protein is generally well conserved between the genotypes of each subclass, there is variability in domains that contribute to the overall structure of the protein. Differences in protein domains could contribute to changes in quaternary structure or accessibility of the antigenic sites. We hypothesize that a bivalent vaccine (Fa & Fb) based on consensus of the F proteins will provide broader protection among all circulating genotypes when compared to a monovalent RSV-F (Fa) vaccine derived from historical virus (GA1 genotype). Contributors: Piedra, Pedro A. ; Avadhanula, Vasanthi 109 BAYLOR COLLEGE OF MEDICINE RARE VARIANT EXTENSIONS OF THE TRANSMISSION DISEQUILIBRIUM TEST DETECTS ASSOCIATIONS WITH AUTISM EXOMES SEQUENCE DATA Zongxiao He Department of Molecular & Human Genetics Advisor: Suzanne Leal, Ph.D.-Department of Molecular & Human Genetics Autism spectrum disorder (ASD), a heterogeneous disorder with substantial heritability, is defined by impaired social communication, deficits in language development, and the presence of restricted interests and/or stereotyped repetitive behaviors. Genome-wide association, de novo mutation and copy number variant studies have reported more than 100 different genes and genomic regions to be associated with this complex trait, but for at least 70% of autism cases the underlying genetic component remains unexplained. Under the hypothesis that rare variants may be involved in the etiology of ASD, we applied the rare variant extension of transmission disequilibrium test (RV-TDT; He et al. 2014) on the exomes of 2,377 parent-child trios exhibiting sporadic ASD. TDT tests whether or not the frequency of transmitted alleles is the same as the alleles not transmitted to an affected child, and an excess of an allele of one type transmitted to the affected offspring indicates a disease-susceptibility locus for the trait that is both linked and associated with the marker. To enrich the association signal from rare variants in sequence data, RV-TDT incorporates four commonly used rare variant association methods: CMC (Li & Leal, 2008), WSS (Madsen & Browning 2009), BRV (Morris & Zeggini, 2010) and VT (Price et al. 2010). After a stringent quality control, genotypes were accurately phased by BEAGLE (Browning & Browning, 2009). The gene-based RV-TDT analysis was performed on rare and low frequency [minor allele frequency (MAF) < 5%] nonsynonymous and putative splices site variants. Strong associations were observed for a burden of rare variants within gene IFT20 on genomic region 17q11.2 (p value = 4.8x10-4) and gene CPNE2 on genomic region 16q13 (p value = 9.6x10-4). Our results show that RV-TDT analysis of trio-based sequence data is a powerful tool to identify ASD genes and suggest that rare variants are highly involved the etiology of ASD. Contributors: Zong-Xiao He, Suzanne M. Leal 110 2014 GRADUATE STUDENT SYMPOSIUM A TAIL-LIKE ASSEMBLY AT THE PORTAL VERTEX IN INTACT HERPES SIMPLEX TYPE-1 VIRIONS Corey W Hecksel Department of Molecular Virology & Microbiology Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology Latent Herpes Simplex Virus type 1 (HSV-1) infections are a special public health concern as asymptomatic viral shedding is believed to be responsible for over 70% of primary infections and has been shown to increase the rate of HIV transmission in patients with concurrent infections. HSV shares a common evolutionary origin with tailed bacteriophages, which contain a DNA packaging motor, or portal, at one vertex of the icosahedral particle. The bacteriophage tail is a complex structure that assembles at the portal vertex and provides the mechanism for delivering the viral genome into the infected cell. Using electron cryo-tomography (cryo-ET), we have identified a previously unsuspected tail-like structure at the portal vertex (termed PVAT) in the HSV-1 virion. The location of this tail-like structure suggests that it is important for virion architecture and may play a role in herpesvirus infection. Contributors: Hecksel, Corey; Schmid, Michael; Rochat, Ryan, Bhella, David; Chiu, Wah; Rixon, Frazer 111 BAYLOR COLLEGE OF MEDICINE PARITY LOWERS THE APOPTOTIC BARRIER TO TUMORIGENESIS IN THE BREAST Sarah Marie Hein Department of Molecular & Cellular Biology Advisor: Yi Li, Ph.D.-Department of Molecular & Cellular Biology An early aged pregnancy leads to a reduced proliferative rate in the parous breast and to an overall reduction in the lifetime risk of breast cancer, but a late aged pregnancy leads to increased long term breast cancer risk. As increasing numbers of women choose to delay childbirth, it is vitally important to understand how a pregnancy may promote or inhibit breast cancer so that effective prevention strategies may be developed. We have reported that pregnancy can instigate preexisting early lesions in their progression to cancer. However, it is not clear whether pregnancy causes the breast epithelium to become more or less susceptible to oncogenic mutations that will be gained later in life. We utilized the RCAS-TVA viral vector system to introduce the oncogene ErbB2 into similar numbers of mammary epithelial cells in fully-involuted and age-matched nulliparous mice. We performed a tumor latency study and compared the fully developed tumors. We find that a prior pregnancy accelerates tumorigenesis of oncogene activated cells. Tumor phenotype does not appear to be affected by parity. We subsequently evaluated precancerous lesions at multiple stages to determine how tumor initiation is being accelerated. We found that early lesions are initiated at similar rates in both cohorts of mice. However, lesions in the parous mice advance more rapidly than those in the nulliparous group, and lesion advancement is associated with reduced rates of apoptosis. Collectively, these data suggest that parity reprograms the mammary epithelium to lower the apoptotic anticancer barrier to oncogenic mutations and to become more susceptible to tumorigenesis initiated by an oncogenic mutation. These findings stand in direct contrast to all known mechanisms of parity acceleration of tumorigenesis, which assume that the initiating insult must occur prior to pregnancy. Therefore, these findings uncover a novel mechanism by which pregnancy regulates breast cancer risk. Contributors: Hein, Sarah; Haricharan, Svasti; Huang, Shixia; Li, Yi 112 2014 GRADUATE STUDENT SYMPOSIUM DEVELOPMENT OF AN INDUCIBLE DENDRITIC CELL IMMUNOTHERAPY TARGETING MAGE-A+ LUNG CANCER Amanda Y. Hendrix Program in Translational Biology & Molecular Medicine Advisor: Farrah Kheradmand, M.D.-Department of Medicine Anita Sabichi, M.D.-Department of Medicine The number one cause of cancer death for men and women in the US is lung cancer. The poor 16.8% 5-year relative survival rate is attributed to the late-stage diagnosis of lung cancer; 57% of lung cancer patients are initially diagnosed with metastatic disease. The MAGE-A protein family of cancer/testis antigens is expressed in 59-69% of non-small cell lung cancers and is associated with decreased overall survival. In addition, expression increases proportionally as the disease progresses, interfering with the p53 tumor suppression transcriptional program. Advances in bicistronic adenoviral (Adv) vector-mediated activation of in vivo dendritic cells (DCs) provide a promising platform for a novel adjuvant therapy against metastatic lung cancer. This system is a departure from recent clinical trials involving expensive and lengthy ex vivo DC-maturation protocols that have produced suboptimal clinical outcomes; in these trials, DCs express low levels of co-stimulatory molecules and TH1-polarizing cytokines, show poor migration to lymphoid tissues, insufficient longevity, and potentially tolerogenic outcomes. In contrast, in vivo bicistronic delivery of our drug-inducible composite activation receptor with a tumor-specific antigen activates DCs and has been shown to successfully enhance potent CD8+ T-cell responses against antigen-specific tumor growth. Coupled with the widely expressed lung tumor antigen MAGE-A, we hypothesize that our bicistronic Adv will target DCs for potent activation of specific anti-tumor T cell responses against Mage-a+ lung tumors in vivo. Mage-a3 and Mage-a4 murine mRNA have been found to be expressed in more than one lung tumor model. We have designed and produced the bicistronic adenoviral vector iMC-Mage-a, combining the drug-inducible composite activation receptor MyD88 and CD40, “iMC,” with the tumor-specific antigen Mage-a. The murine Mage-a3 and -a4 proteins are fused using a glycine flexible linker. Initial functional studies show protein expression of the iMC activation receptor and the Mage-a3 and Mage-a4 antigens, and the construct is able to induce nucleofactor-kappa B signaling critical to the adjuvant function and anti-tumor response. A high titer of the vaccine is currently being produced by a third party. Future work will evaluate murine in vitro and in vivo T cell tumoricidal function following activation of iMC-Magea bicistronic Adv-transduced DCs. Then we will evaluate in vivo anti-tumor function and efficacy of the iMC-Magea bicistronic Adv using murine lung tumor models expressing murine Mage-a. Contributors: Hendrix, AY; Spencer, D; DeMayo, F; Levitt, J; Kheradmand, F in collaboration with the Helis Foundation. 113 BAYLOR COLLEGE OF MEDICINE CHOLINERGIC BASAL FOREBRAIN FEEDING CIRCUIT MODULATES APPETITE SUPPRESSION Alexander Michael Herman Program in Developmental Biology Advisor: Benjamin Arenkiel, Ph.D.-Department of Molecular & Human Genetics Acetylcholine has long been proposed to influence feeding behavior. However, this idea has been largely based on anecdotal evidence associated with tobacco users who attest to the appetite-suppressing effect that tobacco has on the body. This link between acetylcholine and tobacco is important to consider because nicotine, the addictive component in tobacco that is believed to act as an appetite suppressant, acts on the same receptors in the body as acetylcholine. Therefore, endogenous acetylcholine may act in a similar way to curb appetite under normal physiological conditions that nicotine does in tobacco users. Thus far, however, a source site of acetylcholine in the brain that might play a homeostatic role in suppressing appetite is unknown. Using conditional genetic approaches to impair cholinergic signaling from the diagonal band of Broca (DBB) – the major component constituting the cholinergic basal forebrain (CBF) – we show that adult mice lacking cholinergic DBB neurons display increased food intake and develop severe obesity. Furthermore, conditional removal of cholinergic neurotransmission from the DBB by targeted knockout of the choline acetyltransferase (Chat) gene recapitulates these findings. In addition, viral-mediated mapping strategies uncovered presynaptic inputs into the cholinergic DBB originating predominantly from the hypothalamus, the master-regulatory site of feeding behavior in the brain. In turn, feeding-associated postsynaptic projections from the DBB converged in the median eminence, another site important in regulating food intake. Taken together, these data reveal a novel role of cholinergic CBF neurons in modulating satiety in order to promote normal feeding behavior. Contributors: Herman, Alexander; Ortiz-Guzman, Joshua; Garcia, Isabella; Arenkiel, Benjamin 114 2014 GRADUATE STUDENT SYMPOSIUM CARDIAC AUTONOMIC DYSFUNCTION IN MICE LACKING METHYL CpG BINDING PROTEIN 2 Jose A. Herrera Program in Translational Biology & Molecular Medicine Advisor: Jeffrey Neul, M.D./Ph.D.-Department of Pediatrics Jeffrey Kim, M.D.-Department of Pediatrics Background: Rett Syndrome (RTT) is an X-linked dominant neurodevelopmental disorder primarily caused by mutations in Methyl-CpG-binding protein 2 (MECP2), a transcriptional regulator. Twenty-five percent of all deaths in RTT are sudden and unexpected, and autonomic nervous system (ANS) dysfunction is hypothesized to be the cause of sudden deaths in RTT. However, the role of MeCP2 in cardiac autonomic function has not been investigated in depth. Design/Methods: Male (2 months) and female (9 months) Mecp2 deficient and wildtype mice were implanted with ETA-F10 telemeters and 24h recordings were taken to calculate heart rate, heart rate variability, and incidence of arrhythmic events. Effects of pharmacological stimulants or inhibitors on heart rate were normalized to saline injection response. Results: Male Mecp2 deficient mice (NULL) have a decreased heart rate (WT = 634 ±3, NULL = 518 ±8, p<0.001), while female deficient mice (NULL/+) do not present with a decreased heart rate. Interestingly, NULL and NULL/+ mice both have a high incidence of sinus pauses and AV block (WT 1/5, NULL 6/6, NULL /+ 4/4, p<0.05). Additionally, both NULL and NULL/+ mice have increased bradycardia events defined as a heart rate below 200 bpm, (WT = 17 ±6, NULL = 307 ±94, NULL/+ = 198 ±67, p<0.05) and an increased heart rate variability. The normalized heart rate response to atropine, a parasympathetic blocker, of NULL mice was above wildtype levels (p<0.05) Chemical denervation by combined antagonism of parasympathetic (atropine) and sympathetic (propranolol) resulted in NULL mice having an increased normalized heart rate response compared to wildtype (p<0.05). Finally, the incidence of AVB/sinus pauses counts per 30 minutes post-acute treatment with atropine and chemical denervation (NULL saline 49 ±10, NULL atropine 8 ±5, NULL atropine/propranolol 0.5 ±0.5 p<0.05) decreased the severity of the cardiac phenotype in NULL mice. Conclusions: In summary, Mecp2 is required for a normal heart rhythm. Loss of Mecp2 causes bradycardia, sinus pauses, AV block, and increased heart rate variability that may be attributed by aberrant innervation. Acute atropine and chemical denervation was therapeutic and ameliorated the cardiac phenotype observed in Mecp2 deficient mice. Contributors: Herrera J, Ward, CS, Neul, JLN 115 BAYLOR COLLEGE OF MEDICINE USE OF GENE EDITING FOR GENE THERAPY OF AUTOSOMAL DOMINANT RETINITIS PIGMENTOSA Tyler John Hilton Integrative Program in Molecular and Biomedical Sciences Advisor: Theodore Wensel, Ph.D.-Department of Biochemistry & Molecular Biology John Wilson, Ph.D.-Department of Biochemistry & Molecular Biology Retinitis pigmentosa (RP) is a neurodegenerative disease that causes progressive loss of vision due to defective retinal cell function. RP is a very heterogeneous disease, with over 60 genes associated with the disease phenotype. Of these genes, rhodopsin is linked mostly to Autosomal Dominant RP (ADRP), and accounts for about 10% of all cases of RP. Mutations in this gene alter rhodopsin function or localization, resulting in progressive degeneration of the rod photoreceptors in the retina. Past experiments in the lab have used zinc-finger nucleases to cause a double strand break in the last exon of the rhodopsin gene, which was found to be repaired predominantly by non-homologous end-joining (NHEJ) and produce an insertion or deletion. If such a frameshift site were inserted upstream of the mutations in rhodopsin associated with ADRP, it could potentially attenuate the effect of the toxic protein products. Combined with a functional copy of the rhodopsin gene resistant to the double-strand break agent, this “kill and replace” gene therapy strategy may be used to treat the ADRP phenotype from the associated rhodopsin mutations. Thus far, we are focusing on using TALENs and CRISPR/Cas9 systems to target sites in the first and second exons of the rhodopsin gene, to introduce a double-strand break that will be repaired by NHEJ in the rod photoreceptors. These double-strand break agents will be tested in HEK 293 cells to determine activity before they will be packaged into rAAV and used in murine models of ADRP. Contributors: Hilton, Tyler; Wilson, John H.; Wensel, Theodore G. 116 2014 GRADUATE STUDENT SYMPOSIUM ESTROGEN-RESPONSIVE NEURONS IN THE MEDIAL AMYGDALA PREVENT STRESS-INDUCED HYPERTENSION Antentor Othrell Hinton Integrative Program in Molecular and Biomedical Sciences Advisor: Yong Xu, Ph.D.-Department of Pediatrics Background: Hypertension is the leading cause of cardiovascular disease worldwide. The etiology of essential hypertension is unknown. Psychological stress contributes to development of hypertension in humans. The effects are mediated at least partly in the amygdala which is part of the brain limbic system that mediates emotional and hormonal responses to stress. In humans, mice and rats, neural activities in the medial amygdala (MeA) are positively associated with levels of blood pressure (BP) during stress and estrogens attenuate stress-induced c-fos expression in the MeA. Estrogen receptor-α (ERα) is highly expressed in the MeA. Thus, we hypothesize that ERα expressed by MeA neurons mediate the anti-hypertensive effects of estrogen during stress. Methods: Experiment 1: SIM1-Cre mice were subjected to bilaterally stereotaxic injections with AAV-hM3Dq-mCherry into the MeA and after two weeks for the virus to replicate, mice were inserted with telemetry probes. After seven days recovery, mice received ip injections of CNO or saline and blood pressure was recorded for 3 hours to establish baseline and then mice were subjected to 1 hour stress restraint. Experiment 2: C57BL/6 female mice were subjected to bilateral ovariectomy (OVX) and given a vehicle (V) or 17-β-Estradiol pellet (E). One week later, female OVX + V and E mice received either no stress or 1 hr stress and were perfused and stained for c-fos. Experiment 3: We used the Cre-LoxP system to remove ERα from specific MeA neurons in mice (ERαlox/lox/SIM1-Cre). Female ERαlox/lox/SIM1-Cre mice and controls (ERαlox/lox) were subjected to bilateral ovariectomy (OVX), given a vehicle (V) or 17-β-Estradiol pellet (E), and inserted with telemetry probes on day one. After seven days recovery, blood pressure was recorded for 3 hours to establish baseline and then mice were subjected to 1 hour stress restraint. Experiment 4: C57BL/6 Female mice received OVX surgery and were given V and E therapy. 1 week later, mice received either no stress or 1 hr stress and then amygdala tissue was harvested for western blot analysis of glutamate receptors. Results: We found that the basal BP and HR (before stress started) were significantly elevated by CNO compared to saline injections. In addition, all mice showed increased BP/HR upon restraint stress, and CNO-treated mice displayed significantly potentiated stress-induced responses compared to saline-treated mice. C57BL/6 OVX + V and OVX + E mice receiving no stress had comparable c-fos activity. However, stress-restraint C57BL/6 OVX + V mice showed increased c-fos activity in the MeA whereas stress-restraint C57BL/6 OVX + E mice had attenuated c-fos in the MeA. ERαlox/lox OVX + V mice showed increased mean arterial blood pressure (MAP) and systolic arterial blood pressure (SAP) in response to stress. Conversely, ERαlox/lox OVX+ E mice had blunted stressinduced hypertension (MAP and SAP). Interestingly, ERαlox/lox/SIM1-Cre OVX + E and OVX + V group both showed increased MAP, SAP, and RPP in response to stress. AMPA Receptor Subunits GluR1 (GluA1) and GluR2 (GluA2) increase after stress in OVX+V group, responses which appeared to be blunted in OVX+E group. OVX+E group shows higher membrane expression of GluA1 and GluA2 levels in basal condition compared to OVX+V group. Conclusions: We demonstrated that selective activation of SIM1 neurons in the MeA increases basal BP and potentiates hypertensive responses provoked by psychological stress (restraint) in conscious mice. We also demonstrated that estrogens protect against increased neural activity under stress. Also, ERα in the MeA is required to mediate estrogenic actions to prevent stress-induced hypertension. Our results also indicate that estrogen increases glutamatergic membrane receptors, which may mediate the anti-hypertensive effects of estrogen during stress. Contributors: Hinton,Jr., Antentor; Xia, Yan; Henderson, Alexander; Xu, Pingwen; Saito, Kenji; Cao, Xuehong; Yan, Xiaofeng; Reynolds, Corey; Xu, Yong 117 BAYLOR COLLEGE OF MEDICINE KIN RECOGNITION PROTECTS SOCIAL AMOEBA FROM CHEATERS Hsing-I Ho Department of Molecular & Human Genetics Advisor: Gad Shaulsky, Ph.D.-Department of Molecular & Human Genetics The evolution of sociality and altruism is enigmatic because cooperators are constantly threatened by cheaters who benefit from cooperation without incurring its full cost. Kin recognition is the ability to recognize and cooperate with genetically close relatives. It has also been proposed as a potential mechanism that limits cheating, but there has been no direct experimental support for that possibility. Here we show that kin recognition protects cooperators against cheaters. The social amoebae Dictyostelium discoideum cooperate by forming multicellular aggregates that develop into fruiting bodies of viable spores and dead stalk cells. Cheaters preferentially differentiate into spores while their victims die as stalk cells in chimeric aggregates. We found that the victims escaped exploitation by different types of non-kin cheaters. This protection depends on kin-recognition-mediated segregation, because it is compromised when we disrupt strain segregation. The genetic variability in this recognition system is puzzling because cells expressing incompatible cues are expected to be eliminated during development. We show that rare recognition variants are excluded at first, but rejoin the cooperative aggregate and produce spores due to subsequent suppression of kin recognition. Cheating through several strategies is also reduced at late development, thus limiting the threat posed by chimerism. These findings provide direct evidence for the role of kin recognition in cheater control, and suggest a mechanism for the maintenance of stable cooperative systems. Contributors: Ho, Hsing-I; Hirose, Shigenori; Kuspa, Adam; Shaulsky, Gad 118 2014 GRADUATE STUDENT SYMPOSIUM A HIERARCHY OF ANKYRIN/SPECTRIN COMPLEXES CLUSTERS SODIUMCHANNELS AT NODES OF RANVIER Szu-Yu Ho Program in Developmental Biology Advisor: Matthew Rasband, Ph.D.-Department of Neuroscience The scaffolding protein ankyrinG is required for Na+ channel clustering at axon initial segments. It is also considered essential for Na+ channel clustering at nodes of Ranvier to facilitate fast and efficient action potential propagation. However, in contrast to these widely accepted roles, we show here that ankyrinG is dispensable for nodal Na+ channel clustering in vivo. Surprisingly, without ankyrinG, erythrocyte ankyrin (ankyrinR) and its binding partner βI spectrin substitute and rescue nodal Na+ channel clustering. In addition, channel clustering is also rescued after loss of nodal βIV spectrin by βI spectrin and ankyrinR. Thus, ankyrinR/βI spectrin protein complexes function as secondary reserve Na+ channel clustering machinery. In mice lacking both ankyrinG and ankyrinR, Na+ channels fail to cluster at nodes. Thus, two independent ankyrin/spectrin protein complexes exist in myelinated axons to cluster Na+ channels at nodes of Ranvier. Contributors: HO, Tammy Szu-Yu; Zollinger, Daniel R.; Chang, Kae-Jiun; Xu, Mingxuan; Cooper, Edward C.; Stankewich, Michael C.; Bennett, Vann; Rasband, Matthew N. 119 BAYLOR COLLEGE OF MEDICINE ACTIVATION OF THE TRANSLATION PATHWAY CORRELATES WITH BREAST CANCER RECURRENCE MEDIATED BY FGFR1 SIGNALING IN A PRECLINICAL MOUSE MODEL Xue B Holdman Integrative Program in Molecular and Biomedical Sciences Advisor: Jeffrey Rosen, Ph.D.-Department of Molecular & Cellular Biology Background: Tumor dormancy appears to play a critical role in the late recurrence of luminal breast cancers. Fibroblast growth factor receptor 1 (FGFR1), amplified in more than 20% of luminal B breast cancers, is correlated with endocrine resistance, poor prognosis and increased recurrence. However, the molecular basis of the recurrence remains elusive. Experimental Design and Methods: We have employed a transplantable luminal B breast cancer genetically engineered FGFR1/Wnt1-induced mouse mammary tumor model to study the mechanisms involved in dormancy, resistance and tumor recurrence. Tumor bearing mice treated with a pan-FGFR specific inhibitor BGJ398 resulted in rapid tumor regression due to both inhibition of proliferation as well as increased apoptosis. Results: Following 10 days of treatment with BGJ398 the tumors disappeared, and treatment was stopped. After approximately 60 days of dormancy, the tumors recurred. The recurrent tumor cells were still sensitive to BGJ398 treatment, and again could be eliminated following treatment, but again recurred when treatment was stopped. Interestingly, the second recurrence occurred sooner (25 days) than the first recurrence. Western blot analysis shows that the recurrent tumors had enhanced phosphorylation of the mTOR kinase and 4EBP1 as compared to the primary tumors, suggesting that there are escape pathways activated to circumvent the effect of BGJ398. Conclusion: In summary, we have demonstrated that mTOR kinase can be phosphorylated through FGFR1 signaling and that the increased activation of the protein translation pathway is correlated with recurrence. Future studies are focused on the molecular characterization of the dormant cells after BGJ398 treatment. Supported by NIH grant CA16303. Contributors: Holdman, Xue; Rosen, Jeffrey 120 2014 GRADUATE STUDENT SYMPOSIUM AN ATLAS OF TRANSCRIPTION FACTOR DNA BINDING ACTIVITY IN TOTIPOTENT STEM CELLS Matthew Valle Holt Department of Biochemistry & Molecular Biology Advisor: Jun Qin, Ph.D.-Department of Biochemistry & Molecular Biology Recent advances in stem cell research have shown intricate and robust mechanisms which are responsible for the establishment and maintenance of pluripotency: the ability to generate any embryonic germ layer. The addition of only four transcription factors is capable of reprogramming somatic cells into a pluripotent state. Embryonic stem cells lack the capability of significantly contributing to extra-embryonic lineages such as the trophectoderm, and thus are different than totipotent cells which can contribute to both. However, the addition of two inhibitors (CHIR99021 and PD 0325901) has been shown to cause embryonic stem cells to express a trophectoderm marker in conjunction with pluripotency markers and actively contributing both embryonic and extra-embryonic lineages. The primary objective of this project is to understand the key players involved in the response to this inhibitor treatment and which are the key transcription factors involved in totipotency. Transcription factor DNA binding activity and abundance can be determined at the protein level by using transcription factor response elements as an affinity reagent to pull-down DNA binding proteins (catTFRE pull-down). These proteins can be subsequently analyzed by liquid chromatography and mass spectrometry, and through our database software package (iSPEC and Align!) to determine which transcription factors and associated proteins are activated in a totipotent state. We have identified 436 DNA binding transcription factors in pluripotent and totipotent stem cells. Furthermore, distinct transcription factor patterns arise from each individual inhibitor treatment potentially underscoring the transcription factors directly involved in the transition from pluripotency to totipotency. Contributors: Holt, Matthew; Yucer, Nur; Choi, Jong Min; Jain, Antrix; Kim, Beom-Jun; Chan, Doug; Malovannaya, Anna; Wang, Yi; Qin, Jun 121 BAYLOR COLLEGE OF MEDICINE SEEING THE PORTAL IN MEMBRANE-CONTAINING BACTERIOPHAGE PRD1 BY CRYO-EM Chuan Hong Program in Structural and Computational Biology and Molecular Biophysics Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology The functional and structural knowledge of assembly principles of viruses, often dsDNA viruses composed of only proteins and nucleic acids, have extended our understanding of viral capsid maturation and genome packaging processes. Viruses with lipids possess an additional complexity when exploring the mechanistic and structural properties of such fundamental functions. PRD1 phage, which is known to infect Salmonella typhimurium, is a good candidate for a possible antibacterial agent. PRD1 is an icosahedral dsDNA bacterial virus with an inner membrane (Tectiviridae family). Based on previous X-ray crystallographic results of PRD1, its major capsid protein has similar fold as those of several other viruses such as adenovirus, PBCV-1 and STIV. However, the structure of the non-icosahedrally arranged portal complex anchored in the inner membrane remains elusive. Biochemical and immuno-electron microscopic studies have identified four proteins in the portal complex: the packaging ATPase P9, the packaging efficiency factor P6, and the integral membrane proteins P20 and P22. The goal of our study is to reveal the structures of the portal machinery of PRD1. We used single-particle electron cryo-microscopy (cryo-EM) to study the mature virion and three procapsid mutants of PRD1 using symmetry-free reconstructions. Their density maps allow us to conclude the locations and features of the four portal proteins at a unique vertex. The P20 and P22 form a hexamer of dimers embedded in the viral membrane and function as a conduit for the DNA packaging. The P20 or P22 cannot exist alone without the other. The P6 and P9 form a 12-mer of a portal complex with ATPase activity similar to other phage portal protein complex. This is the first structural evidence of the PRD1 packaging complex operating at a specific vertex, and shows the connection between the membrane and the capsid shell providing a conduit for DNA translocation in an ATPase-driven reaction. Contributors: Hong, Chuan; Liu, Xiangan; Jakana, Joanita; Oksanen, Hanna; Bamford, Dennis; Chiu, Wah 122 2014 GRADUATE STUDENT SYMPOSIUM A RARE VARIANT LOCAL HAPLOTYPE SHARING METHOD WITH APPLICATION TO ADMIXED POPULATIONS Stanley Eugene Hooker Department of Molecular & Human Genetics Advisor: Suzanne Leal, Ph.D.-Department of Molecular & Human Genetics With the advent of next generation sequencing there is great interest in studying the involvement of rare variants in complex trait etiology. For many complex traits sequence data is being generated on DNA samples from African Americans and Hispanics to elucidate rare variant associations. Analyses of admixed populations present special challenges due to spurious associations which can occur because of confounding. However using information on admixture and local ancestry can also be highly beneficial and increase the power to detect associations in these populations. Here a local haplotype sharing (LHS) method (Xu and Guan 2014) was extended to test for rare variant (RV) associations in admixed populations. Previously the Weighted Haplotype and Imputation-based Test (WHAIT) (Li et al. 2010) was proposed to test for rare variant associations using haplotype data. The RV-LHS method unlike WHAIT, does not require reconstruction of haplotypes which can be both computationally intensive and error prone. Additionally the RV-LHS uses information on local ancestry which is particularly advantageous when analyzing admixed populations. Results will be shown from simulation studies performed for rare variant data from an admixed population. Both Type I and II errors are evaluated for the RV-LHS method. Additionally the power of the RV-LHS method is compared to WHAIT as well as several other nonhaplotype-based rare variant association methods including the combined multivariate collapsing (CMC) (Li and Leal, 2008), Variable Threshold (VT) (Price et al. 2010) and Sequence Kernel Association Test (SKAT) (Wu et al. 2010). Several heart, lung and blood phenotypes were analyzed using sequence data on African-Americans from the NHLBI-Exome Sequencing Project to better evaluate the performance of the RV-LHS compared to other rare variant association methods. Contributors: Hooker, Stanley; Wang, Gao T; Li, Biao; Guan, Yongtao; Leal, Suzanne M 123 BAYLOR COLLEGE OF MEDICINE EFFECT OF DNA VECTOR LENGTH ON CELL TRANSFECTION Benjamin Daniel Hornstein Department of Molecular Virology & Microbiology Advisor: E. Zechiedrich, Ph.D.-Department of Molecular Virology & Microbiology Gene therapy requires the delivery of nucleic acid to replace, regulate, or correct genes to treat diseases. Viral vectors deliver DNA efficiently to cells, but are often unsuitable for therapeutic applications because their integration into the host genome can disrupt or dysregulate essential genes or proto-oncogenes. Nonviral vectors do not have these issues, but many human cell lines are refractory to transfection with plasmid. Our laboratory developed minimized nonviral vectors that can be as small as 250 bp, called minivectors. Minivectors are devoid of bacterial sequences, and are unlikely to be silenced. Published and preliminary data show that minivectors transfect hard-totransfect cell types, including primary cells and cells in suspension, and cause no toxicity. During transfection, a DNA vector needs to enter the cell, translocate to the nucleus, and be expressed to mediate an effect. Whereas minivectors get into cells extremely well, the observed minivector-mediated knockdown was less efficient than plasmid when the same number of molecules were transfected. I hypothesized that smaller vectors get expressed less efficiently than larger ones. To test how vector length affects transfection, I created DNA vectors expressing shRNA against GFP. The lengths were 383, 735, 1,026, 1,869, 2,844, 3,913, 4,265, and 4,556 base pairs (bp). I tested transfection efficiency by measuring GFP knockdown in HeLa cells stably expressing GFP. By using HeLa cells, which are easily transfected, we can test how vector length affects nuclear localization and expression without including cell entry as a variable. Flow cytometry and fluorescence microscopy were used to quantify the data. Indeed, the smallest vector showed the least knockdown, and the largest three vectors showed similar knockdown. To test directly whether these differences in GFP knockdown are a consequence of inhibition of RNA polymerase, I am quantitatively measuring in vitro transcription. Because each clinical application is different, understanding the effect of vector size on transfection is necessary for calculating the amount of DNA vector needed to elicit the appropriate expression level for any given application. The ultimate goal of this project is to find the optimal minivector size for transfection so we can design a nonviral gene therapy vector that maximizes expression while minimizing toxicity from transfection vehicle and vector. Contributors: Arevalo-Soliz, Lirio Milenka; Szafran, Adam; Mancini, Michael; Zechiedrich, Lynn 124 2014 GRADUATE STUDENT SYMPOSIUM FOOD AVOIDANCE AND USE OF DIETARY SUPPLEMENTS AMONG PATIENTS WITH INFLAMMATORY BOWEL DISEASE Jason K Hou Clinical Scientist Training Program Advisor: Hashem El-Serag, M.D./M.P.H.-Department of Medicine Purpose: Patients with inflammatory bowel disease (IBD) perceive that diet impacts their gastrointestinal symptoms. There are conflicting data regarding the effect of dietary modifications on IBD activity; however, patterns and patient perceptions of food aversions among IBD patients are not well defined. Unnecessary food aversion may lead to macro- and micro- nutrient deficiencies. The aim ofthis study was to evaluate the frequency and patient perceptions of food aversions among patients with IBD. Methods: A case-control study of 104 patients with an established diagnosis of IBD and 101 healthy controls was performed. Patients with known diagnosis of Ulcerative Colitis (UC) and Crohn’s disease (CD) were recruited in consecutive fashion from an IBD referral center. Controls were healthy volunteers without IBD, family history of IBD, or a diagnosis of irritable bowel syndrome. A 38 question selfadministered food frequency questionnaire was administered to patients and controls. Chart review of all cases was performed to confirm diagnosis of IBD and obtain clinical characteristics using a standardized abstraction form. Comparisons of patterns of use of vitamins, supplements, and food aversions were performed using Chi square and Fisher-exact test. Multivariate logistic regression was performed to adjust for age, sex, and ethnicity. Results: A total of 104 IBD cases (50 UC, 54 CD) and 101 healthy controls were included. After adjusting for age, sex, and ethnicity, IBD patients were more likely to take vitamin/mineral supplements (OR 5.30, 95% CI 2.41-11.66) and use liquid meal replacements (OR 4.66, 95% CI 1.76-12.37). IBD patients were also more likely to have food aversions overall (OR 3.49, 95% CI 1.47-8.30), and specifically to fruit/juice (OR8.24, 95% CI 2.98-22.81), vegetables (OR 6.40, 95% CI 2.72-15.05), pork/beef/poultry (OR 2.83, 95% CI 1.26-6.34), and peanuts/nuts (OR 11.79, 95% CI 4.00-34.80) compared to healthy controls. Exacerbation of IBD symptoms was the most commonly patient-reported reason for food avoidance amongst IBD patients. Conclusions: Avoidance of fruit, vegetables, meat, and nuts are more common among patients with IBD than healthy controls. Patients perceive that these food groups exacerbate their IBD symptoms. Dietary restrictions may increase risk of nutritional compromise in these vulnerable patients and increase unnecessary stress among IBD patients. Further studies on the perceptions of dietary intake on IBD symptoms may identify adjunct therapy such as elimination or exclusion diets to control symptoms, prolong remission, and involve patients actively in their care. Contributors: Chen, Tien-Chun; Cruz, Guillermina; Sellin, Joseph; Hou, Jason 125 BAYLOR COLLEGE OF MEDICINE VALIDATION OF NEAR-ATOMIC RESOLUTION MACROMOLECULES TO IDENTIFY STRUCTURES FOR DRUG DESIGN Corey F. Hryc Program in Structural and Computational Biology an Molecular Biophysics Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology Structural biology plays a key role in the development of novel biological inhibitors and drug delivery methods. Recently, structures of large macromolecular complexes ranging from 400 kDa to tens of MDa have been solved by single particle electron cryo-microscopy, resulting in near-atomic resolution structures, however lacking validation. This absence of validation devalues the derived models and their usefulness in drug discovery. Large datasets exist for these structures in various databases, including the Protein Data Bank (PDB) and Electron Microscopy Data Bank (EMDB), but this data must be validated to annotate the level of structural uncertainty prior to their use in translational medicine. By understanding this structural variability, such as atom locations and interactions between molecules, pharmaceutical chemist can target specific domains and interfaces for drug design with confidence. We have repurposed existing crystallographic software for our near-atomic resolution structures determined without crystallography. In an attempt to understand the model on a per-atom basis, we developed a number of model building and optimization protocols to interrogate the expansive data set that comprises cryo-EM density maps. These protocols are currently being tested with a known virus, in addition to various unknown structures. We can then apply rigorous validation test, as in crystallography, for structures commonly used in drug design. Using this approach, we can analyze and improve model validity for current and future structures. Our early results provide a statistical assessment that can be used in conjunction with fit-todensity and stereochemistry scores to assess model uncertainty. This research was funded by a training fellowship from the Keck Center of the Gulf Coast Consortia, on the Training Program in Biomedical Informatics, National Library of Medicine (NLM) T15LM007093-21, PI - G. Anthony Gorry. Contributors: Hryc, Corey; Chen, Dong-hua; Wang, Zhao; Afonine, Pavel; Baker, Matthew; Adams, Paul; Chiu, Wah 126 2014 GRADUATE STUDENT SYMPOSIUM THE PURPOSE OF GRANULE CONVERGENCE IN NK CELL CYTOTOXICITY Hsiang-Ting Hsu Department of Pathology & Immunology Advisor: Jordan Orange, M.D.-Department of Pediatrics Natural killer (NK) cells are cytolytic immune lymphocytes that survey the host environment for stressed or diseased cells and are required for maintenance of human health. NK cell activation triggers a stepwise series of events leading to degranulation of specialized lysosomal-related organelles called lytic granules. Prior to degranulation, preformed lytic granules rapidly converge from dispersed cytoplasmic locations to the microtubule-organizing center (MTOC), and then polarize to the immunological synapse (IS), which is defined as the contact site with the target cell. Once polarized, lytic granules fuse with NK cell membrane and release their contents onto the target cell to trigger target death. Our lab has previously identified that granule convergence requires LFA-1 signaling and Src family kinase activities, is dependent on dynein motor function but not on actin reorganization, and does not require commitment to NK cell cytotoxicity. Despite these advances the purpose of lytic granule convergence in NK cell cytotoxicity is unknown. We hypothesize that granule convergence can improve efficiency of targeted lytic granule secretion and prevent collateral damage to neighboring tissue. By using a Drosophila S2 cell-based expression system which, unlike mammalian cells, does not express ligands for adhesion and activation receptors of human NK cells we aim to control signal inputs from a target cell to precisely control granule convergence and degranulation. Specifically, engagement of LFA-1 induces convergence of granules, but not degranulation, whereas ligation of CD16 triggers undirected degranulation. Thus, we propose to test our hypotheses using S2 cells expressing only intercellular adhesion molecule (ICAM)-1 as ligand of LFA-1, S2-specific IgG as ligand for the Fc receptor CD16, or ICAM-1+IgG. By mixing differentially labeled IgG-loaded and IgGunloaded target cells we determined the efficiency and specificity of individual NK cells activated through LFA-1, CD16 alone, or LFA-1+CD16. Indeed, we showed that NK cells activated by anti-S2 IgG had diffuse subcellular granule localization and undirected lytic granule release causing non-specific killing of neighboring cells. In contrast, NK cells triggered by S2-ICAM1 cells resulted in granule convergence but not cytotoxicity, whereas NK cells receiving both signals killed targets more precisely and efficiently. By understanding the purpose of granule convergence, we believe we will be able to harness a powerful regulatory step in NK cell cytotoxicity that can be exploited to tailor immunological therapies. Contributors: Hsu, Hsiang-Ting and Orange, Jordan S. 127 BAYLOR COLLEGE OF MEDICINE IMPACT OF GENETIC VARIATIONS ON PROTEIN FUNCTION AND ON FITNESS Teng-Kuei Hsu Department of Biochemistry & Molecular Biology Advisor: Olivier Lichtarge, M.D./Ph.D.-Department of Molecular & Human Genetics Whole genome sequencing uncovers a plethora of genetic alterations that are often with unknown functional and clinical impact. A major challenge is to estimate the consequences of these alterations. As a complement to traditional computational tools that follow machine learning, statistical and biophysical modeling approaches and trained with numerous available data, we sought to assess the impact of novel mutations from a phylogenomic perspective, namely by relating specific genotype variations to specific speciation events. In our retrospective data analysis, this approach i) correlated with the loss of protein function in viral, prokaryotic and eukaryotic proteins, ii) separated the disease-associated mutations from the benign, iii) matched the morbidity of monogenic disorders. When this approach was tested by independent judges at the two most recent international CAGI contests, it also consistently achieved top ranking. In order to assess its practical value in a complex disease, we asked whether it could prognosticate for poor outcomes in head and neck squamous carcinoma (HNSCC), which often bear TP53 mutations. We found in a small cohort but at a statistically significant level that cancer patients can be further stratified into subgroups by the impact of mutations in TP53 determined by our method, such that patients with greater deleterious impact in TP53 mutations associate with the poorest clinical outcomes. Together, these results may open a novel and complementary approach to assess genetic variations at the molecular level and may eventually play a role in the stratification of cancer patients for precise treatment selection. Contributors: Katsonis, Panagiotis; Lichtarge, Olivier 128 2014 GRADUATE STUDENT SYMPOSIUM ONCOGENIC MYC INDUCES A DEPENDENCY ON THE SPLICEOSOME IN HUMAN BREAST CANCER Tiffany Hsu Integrative Program in Molecular and Biomedical Sciences/M.D.-Ph.D. Program Advisor: Thomas Westbrook, Ph.D.-Department of Biochemistry & Molecular Biology c-MYC (MYC) overexpression or hyperactivation is one of the most common drivers of human cancer. Despite intensive study, the MYC oncogene remains recalcitrant to therapeutic inhibition. Like other classic oncogenes, hyperactivation of MYC leads to collateral stresses onto cancer cells, suggesting that tumors harbor unique vulnerabilities arising from oncogenic activation of MYC. Herein, we discover the spliceosome as a new target of oncogenic stress in MYC-driven cancers. We identify BUD31 as a MYC-synthetic lethal gene, and demonstrate that BUD31 is associated with the core components of the human spliceosome. These BUD31-associated core spliceosomal factors (SF3B1, U2AF1, and others) are also required to tolerate oncogenic MYC. Notably, MYC hyperactivation induces an increase in total pre-mRNA synthesis, suggesting an increased burden on the core spliceosome to process premRNA. In contrast to normal cells, partial inhibition of the spliceosome in MYChyperactivated cells leads to global intron retention, widespread defects in pre-mRNA maturation, and deregulation of many essential cell processes. Importantly, genetic or pharmacologic inhibition of the spliceosome in vivo impairs survival, tumorigenicity, and metastatic proclivity of MYC-dependent breast cancers. Collectively, these data suggest that oncogenic MYC confers a collateral stress on splicing and that components of the spliceosome may be therapeutic entry points for aggressive MYC-driven cancers. Contributors: Hsu, Tiffany Y.-T.; Simon, Lukas M.; Neill, Nicholas; Marcotte, Richard; Sayad, Azin; Bland, Christopher S.; Sun, Tingting; Dominguez-Vidana, Rocio; Kurley, Sarah J.; Tyagi, Siddhartha; Karlin, Kristen L.; Hartman, Jessica D.; Renwick, Alexander; Bernardi, Ronald J.; Skinner, Samuel O.; Jain, Antrix; Orellana, Mayra; Lagisetti, Chandraiah; Golding, Ido; Jung, Sung Y.; Neilson, Joel R.; Zhang, Xiang; Webb, Thomas R.; Neel, Benjamin G.; Shaw, Chad A.; Westbrook, Thomas F. 129 BAYLOR COLLEGE OF MEDICINE CRYSTAL STRUCTURES OF A SINGLE DOMAIN IN PKGI REVEAL THE MOLECULAR MECHANISM OF CGMP SELECTIVITY Gilbert Huang Department of Biochemistry & Molecular Biology Advisor: Choel Kim, Ph.D.-Department of Pharmacology Cyclic guanosine monophosphate (cGMP) is a key secondary messenger that is produced in response to nitric oxide. One of the key mediators of cGMP signaling, cGMP-dependent protein kinase (PKG), is activated upon binding to cGMP and phosphorylates downstream substrates in a process required for important physiological processes such as vasodilation, nociception, and memory formation. PKGs are also known to mediate most effect of drugs that increase cellular cGMP levels, including nitric oxide-releasing agents and phosphodiesterase inhibitors, which are used for the treatment of angina pectoris and erectile dysfunction, respectively. We have investigated the mechanism of cyclic nucleotide selectivity by PKG by determining crystal structures of the cGMP-selective carboxyl-terminal cyclic nucleotide-binding domain (CNBD-B) of human PKG I bound to cGMP and in the apo form. Our crystal structure of CNBD-B with bound cGMP reveals that cGMP adopts the syn configuration in the binding pocket and is coordinated by a previously unidentified arginine residue. Furthermore, comparison of the cGMP-bound crystal structure of the apo structure suggests a role for a C-terminal tyrosine residue in capping the nucleotide into the binding pocket. The interaction of this tyrosine residue with cGMP appears to result in conformational rearrangement of the C-terminal helix, suggesting a mechanism for kinase activation by cGMP. Contributors: Huang, Gilbert; Kim, Jeong Joo; Reger, Albert; Casteel, Darren; Bertinetti, Daniela; Lorenz, Robin; Zhao, Chi; Moon, Eui-Whan; Melacini, Giuseppe; Herberg, Friedrich; Kim, Choel 130 2014 GRADUATE STUDENT SYMPOSIUM DISTINCT CONNECTIVITY PATTERNS AND PLASTICITY OF GRANULE CELLS AND EPL INTERNEURONS IN THE OLFACTORY BULB Longwen Huang Department of Neuroscience Advisor: Benjamin Arenkiel, Ph.D.-Department of Molecular & Human Genetics The exquisite balance between excitation and inhibition determines information processing in neural circuits, where inhibitory neurons play significant roles in sculpting principle neuron output and maintaining proper brain function. In prominent models of olfactory bulb circuitry, mitral cells, the principle neurons, receive inhibitory input from granule cells, which are considered to mediate lateral inhibition, facilitate information processing, and/or control the gain of mitral cell activity. Additionally, we recently reported a novel population of GABAergic interneurons located in the external plexiform layer (EPL) of the olfactory bulb, which make reciprocal connectivity with mitral cells. However, the connectivity map between EPL interneurons/granule cells and mitral cells, and how these interneurons contribute to odor processing remain unknown. Using optogenetics, electrophysiology, and fast scanning microscopy, we show that EPL interneurons make spatially broader connectivity with mitral cells, and their activity have subtractive effects on mitral cell’s tuning to odorants; while granule cell-mitral cell circuitry show distinct connectivity patterns, which is much narrower and specific. Furthermore, we are currently implementing light-assisted circuit mapping techniques and behavioral studies to investigate the inhibitory circuit plasticity of EPL interneurons/granule cells; these studies will provide more insight onto the principle underlying the organization and dynamics of inhibitory circuitry in the brain. Contributors: Huang, Longwen; Garcia Isabella; Saggau, Peter; Arenkiel Benjamin. 131 BAYLOR COLLEGE OF MEDICINE ROLES OF MECP2 IN THE AUTONOMIC NERVOUS SYSTEM Teng-Wei Huang Program in Developmental Biology Advisor: Jeffrey Neul, M.D./Ph.D.-Department of Pediatrics Rett syndrome (RTT) is a dominant X-linked neurodevelopmental disorder caused by MeCP2 mutations. RTT associated with loss of communication and purposeful hand skills as well as several autonomic deficits including abnormal temperature regulation, decreased heart rate variability, and respiratory deficits. These autonomic disorders may contribute to the sudden death observed in a fraction of people with RTT. We found that MeCP2 is both necessary and sufficient within the brainstem and spinal cord for normal lifespan, control of heart rate, and respiratory response to hypoxia. These results suggest MeCP2 is critical within autonomic and respiratory control centers for survival, and suggest a possible correlation between abnormal cardiorespiratory regulation and premature lethality. To study MeCP2 functions in specific circuits in the brainstem, HoxA4-Cre line was generated to target caudal medulla and spinal cord. HoxA4-Cre line 1 conditional knockout (CKO) mice have deficient motor functions, increased ventilation hypoxic response, and early lethality. Re-expression of MeCP2 in HoxA4 domain rescued basic motor coordination, forelimb grapping strength, and survival. HoxA4 domain including important component of respiratory network: nucleus of the solitary tract (NTS), PreBötzinger Complex (preBötC) and ventral respiratory column (VRC). Interestingly, although previous study indicates that losing MeCP2 function in the pontine respiratory group results in increased baseline breathing rate, the development of this phenotype in HoxA4 rescue animals is much slower than that in non-rescue animals. In addition, HoxA4 rescue animals had apnea at 7 weeks old but the level of apnea was ameliorated with age. These findings suggest that MeCP2 re-expression in the downstream component of the respiratory network allowed it to adopt and/or resist the mis-regulated inspiratory/expiratory transition signal and maintain a relative normal pacing rhythm. MeCP2 expression in HoxA4 domain partially rescued the decreased heart rate. ECG recording by implanted probe is now performed to determine the detail of cardiac phenotype. Contributors: Huang, Teng-Wei; Ward, Christopher; Neul, Jeffrey Lorenz 132 2014 GRADUATE STUDENT SYMPOSIUM αII-SPECTRIN IS ESSENTIAL FOR PNS NODE OF RANVIER SUBDOMAIN FORMATION Yu-Mei Huang Department of Neuroscience Advisor: Matthew Rasband, Ph.D.-Department of Neuroscience Spectrins are a family of cytoskeletal proteins, which provide structural support of cells, link membrane-associated proteins to actin and serve as platforms for cell signaling. Spectrins are classified into α and β subunits, forming heterotetramers to function as a complex. Among the spectrins, αII-spectrin is highly expressed in both neurons and myelinating glia. It is also implicated in the neurological disorder, West syndrome, which is the infantile spasm with symptoms including cerebral hypomyelination, epilepsy and brain atrophy. Moreover, the importance of αII-spectrin in the nervous system is emphasized by embryonic lethality of constitutive knockout mice with nervous system malformation. Owing to the fact that αII-spectrin constitutive knockout mice die in utero whereas myelination and node formation do not take place until birth, there is no mouse model to study αII-spectrin in myelination and node-related structure formation. Therefore, I generated αII-spectrin conditional knockout (cko) mice to bypass the embryonic lethality and to identify the roles of neuronal and glial αII-spectrin. When αII-spectrin is deleted in the CNS (Nestin-cre), mutant animals have profound phenotypes and die perinatally. Nestin-cre cko mice are smaller and weaker and they have motor function deficits. Besides, immunohistochemistry shows that neurodegeneration occurs throughout the brain and cko mice have defects in neuronal migration in the cerebral cortex while Purkinje cells are dramatically reduced in the cerebellum. Furthermore, axon initial segments are fragmented and remarkably reduced. However, the role of αII-spectrin in CNS myelin and node formation remains unknown due to the early perinatal lethality. To eliminate αII-spectrin exclusively in myelinating glia, αII-spectrin floxed mice were crossed with CNP-cre mice. Surprisingly, myelin and the nodes of Ranvier subdomains can still form. In addition, βII-spectrin remains at bands of Cajal in Schwann cells despite loss of αII-spectrin, suggesting that the spectrin network may work in a different fashion in myelinating glia. On the other hand, when neuronal αII-spectrin is knocked out only in the pheriphery sensory nervous system (advillin-cre), animals have hind leg clasping and significantly worse motor function. Immunohistochemistry shows that paranodes are extensively disrupted and even invade into nodal regions. Moreover, potassium channel clustering is aberrant. This study will reveal the function of αII-spectrin and provide insights into the different roles of spectrin networks in neurons and in myelinating glia. Contributors: Huang, Yu-Mei; Rasband, Matthew 133 BAYLOR COLLEGE OF MEDICINE SMALL WONDERS: CARBON NANOTECHNOLOGY TO TREAT AUTOIMMUNE DISEASES Redwan Huq Department of Molecular Physiology & Biophysics Advisor: Christine Beeton, Ph.D.-Department of Molecular Physiology & Biophysics Autoimmune diseases, such as multiple sclerosis and rheumatoid arthritis, are mediated by a type of white blood cell—T lymphocytes. Current treatments for these diseases are broad-spectrum immunosuppressants associated with life-threatening side-effects, necessitating the development of new therapeutic strategies. The inflammatory microenvironment in these diseases generates large quantities of harmful reactive oxygen species (ROS). However, low levels of intracellular ROS act as signaling molecules necessary for T lymphocyte activation. Therefore, intracellular ROS represent attractive targets for modulating T lymphocyte activity and for treating autoimmune diseases. Carbon nanoparticles can scavenge ROS with higher efficacy than dietary and endogenous antioxidants. The affinity of carbon nanoparticles for specific cell types represents an emerging tactic for targeted therapy. Here, we show that nontoxic poly(ethylene)-glycol-functionalized hydrophilic carbon clusters (PEG-HCCs) are the first carbon nanoparticles to be preferentially internalized by T lymphocytes over other splenic immune cells. We use this selectivity to attenuate T lymphocyte function in vitro without affecting major functions of macrophages, immune cells crucial for the physiological activation of T lymphocytes. We demonstrate the effectiveness of PEGHCCs in reducing T lymphocyte-mediated inflammation in delayed-type hypersensitivity and in ameliorating experimental autoimmune encephalomyelitis and pristane-induced arthritis, animal models of multiple sclerosis and rheumatoid arthritis, respectively. Our results suggest that the remarkable selectivity of PEG-HCCs for T lymphocytes is a novel and promising route for treating T lymphocyte-mediated autoimmune diseases without inducing broad-spectrum immunosuppression. Contributors: Huq, Redwan; Samuel, Errol L.G.; Lee, Thomas; Tanner, Mark R.; Khan, Fatima S; Tajhya, Rajeev B; Inoue, Taeko; Pautler, Robia; Tour, James M.; Beeton, Christine 134 2014 GRADUATE STUDENT SYMPOSIUM HIGH-THROUGHPUT DISCOVERY OF NOVEL PHAGE-DISPLAYED PEPTIDE REAGENTS FOR THE DETECTION OF NOROVIRUS Amy Marie Hurwitz Program in Translational Biology & Molecular Medicine Advisor: Timothy Palzkill, Ph.D.-Department of Pharmacology Robert Atmar, M.D.-Department of Medicine Norovirus (NoV) infections cause over 96% of non-bacterial gastroenteritis and lead to about 21 million new cases annually in the United States alone. Existing diagnostics have significant limitations in feasibility for point-of-care applications, so there is a clear need for more reliable, rapid, and simple-to-use diagnostic tools to prevent epidemic outbreaks and to inform appropriate actions. In this study, phage display technology was used to screen libraries of phages displaying random 12-mer peptides for those that bind to NoV virus-like particles (VLPs). Since NoV strains classified in genogroups I and II cause the majority of human outbreaks, we focused initially on the prototypical Norwalk strain within genotype GI.1. After five rounds of library selection against GI.1, we amplified the phage DNA from phage populations eluted from each round for high throughput sequencing. Using an in-house computational algorithm for analysis, a consensus motif was identified from thousands of sequences for binding GI.1. Peptides containing this consensus motif were then validated for their ability to bind GI.1 VLPs in multiple formats. Overall, the work here provides a proof-of-concept for the use of high-throughput phage display sequence analysis for the identification of peptide motifs with specific binding affinity for a target protein of interest. Contributors: Hurwitz, Amy; Huang, Wanzhi; Atmar, Robert; Palzkill, Timothy 135 BAYLOR COLLEGE OF MEDICINE NORWALK VIRUS SHEDDING DURATION IS ASSOCIATED WITH CHANGES IN THE INTESTINAL MICROBIOME Diane Smith Hutchinson Program in Translational Biology & Molecular Medicine Advisor: Joseph Petrosino, Ph.D.-Department of Molecular Virology & Microbiology Stephen Pflugfelder, M.D.-Department of Ophthalmology Background: Norovirus (NoV) pathogenesis is not fully understood, and the associations and potential role of the gut microbiota in NoV infection have not been described thoroughly. While the gut microbiome contributes to the development of gut immunity, it has also been demonstrated to enhance enteric viral replication and systemic pathogenesis. We assessed the interaction between NoV, the intestinal microbiota, and the human host in fecal samples collected from the Norwalk virus (NV) challenge study performed at Baylor College of Medicine. Methods: The study population consisted of 55 individuals who participated in an experimental challenge with NV. Fecal samples collected from all subjects at 6 timepoints (days -7, 2, 4, 8, 21, and 56) were analyzed to assess changes in the fecal microbiota related to NV infection. Of the 55 subjects included in our study, 35 were uninfected (including 9 non-secretors and 6 placebo) while 20 subjects were infected. 16S rDNA sequencing was performed on the V4 hypervariable region of 328 samples using the Illumina MiSeq platform. Results: Pre-challenge fecal microbiome composition was not associated with susceptibility to NV infection. Following challenge with NV, infection did not induce changes in the structure of the fecal microbiome when compared to uninfected controls. Among infected individuals, the microbiome of long shedders was more similar to the pre-challenge microbiome composition than that of short shedders. Additionally, long shedders exhibited increased abundance of Subdoligranulum, a butyrate producer generally thought to improve intestinal health, compared to short shedders. During infection, the presence of gastroenteritis or specific symptoms was not associated with microbiome composition. Unrelated to NV infection, secretor positive subjects exhibited either high or low abundance of Prevotella, while secretor negative subjects only exhibited low abundance of Prevotella. Conclusions: These results indicate that NV infection does not alter the composition of the fecal microbiome with the exception of short shedders. This suggests that long shedders are able to maintain a healthy microbiome throughout infection. Currently, we are performing metagenomic sequencing to examine gene content in long and short shedders. Further study of the host genetics underlying shedding duration and its association with microbiome stability may lead to better intervention/prevention strategies for NoV infection. Contributors: Hutchinson, Diane S; Ajami, Nadim J; Finkbeiner, Stacy R; Neil, Frederick H; Opekun, Antone R; Metcalf, Ginger A; Muzny, Donna M; Gibbs, Richard A; Graham, David Y; Atmar, Robert L; Estes, Mary K; Petrosino, Joseph F 136 2014 GRADUATE STUDENT SYMPOSIUM SEMANTIC SIMILARITY ANALYSIS OF PATIENT PHENOTYPES FOR GENOME WIDE GENETIC DIAGNOSTICS Regis Aaron James Program in Structural and Computational Biology and Molecular Biophysics Advisor: Chad Shaw, Ph.D.-Department of Molecular & Human Genetics Genome wide genotype data are increasingly important in the molecular diagnosis of human disease. However, interpretation of these genome wide data is challenged by the large number of variants observed in individual patients. We hypothesize that efficient integration of available clinical data characterizing patient indications for genetic testing can improve the diagnostic workflows, physician communication with the diagnostic lab and, ultimately, prioritization of variants. We have developed scalable quantitative analysis tools for mining indication content. Fundamentally, our approach exploits semantic similarity computed on patient phenotypes to comprehensibly represent patient features in simple composite scores. Our tool can help to elucidate cohort substructure by segmenting large, heterogeneous groups of subjects into subgroups based on shared multidimensional phenotypes. Identification of this structure can help to identify and establish cohorts for associative or experimental study, accelerating the investigation of genetic variation. Identification of cohorts can also lead to discovery of novel functional variants that drive complex phenotypes. We have also developed a prototype graphical interactive web-based viewer exploiting our computational toolkit. This viewer can aid decision support through visual query and analysis of structured input data. This tool can help simplify identification of clinically relevant variants by facilitating indirect matching of patient phenotypes to cataloged variants already known to be associated with semantically similar patients or reported OMIM diseases. We use our tools, in the context of the Human Phenotype Ontology representation of patient indications, to analyze a pilot cohort assembled from retrospective data from the Whole Genome Laboratory at Baylor College of Medicine. We demonstrate the performance of our method for matching to known diseases by comparing our computational OMIM disease matches for subjects against the findings reported in exome reports. These tools can help improve the quality of clinical care by more effectively utilizing available indication data, increasing the value of the electronic health record (EHR), and better illuminating the correspondence between the requisition content and the ultimate genetic diagnosis. Contributors: James, Regis; Bainbridge Matthew; Eng, Christine; Shaw, Chad 137 BAYLOR COLLEGE OF MEDICINE ARV1 AS A NOVEL REGULATOR OF ENERGY HOMEOSTASIS Kelsey Elizabeth Jarrett Integrative Program in Molecular and Biomedical Sciences Advisor: William Lagor, Ph.D.-Department of Molecular Physiology & Biophysics Obesity is a major problem in the United States; it is estimated that over 34% of adults are obese and at increased risk for cardiovascular disease, stroke, and diabetes. While several key genetic factors contributing to obesity are known, far less is known of genes that contribute to leanness. ARE2 Required for Viability 1 (ARV1), a transmembrane protein of the endoplasmic reticulum (ER), is a putative sterol transporter. Loss of ARV1 in yeast results in buildup of early sterol intermediates and ceramides in the ER, as well as reduced synthesis of complex sphingolipids. Yeast lacking Arv1 have disorganized ER structure, increased ergosterol in the ER membrane, and constitutive activation of the unfolded protein response. We found that germline deletion of Arv1 in mice results in a 25% reduction in body weight, greatly reduced white adipose tissue, increased energy expenditure, and improved glucose tolerance. These data are mirrored in mice with conditional deletion of Arv1 by AP2-cre, a cre recombinase primarily targeting adipose tissue; however, AP2-cre also deletes in other tissues, particularly the brain. In contrast, deletion of Arv1 with Adiponectin-cre, which is adipose-specific and does not delete in brain, fails to reproduce the phenotype observed with AP2-cre mediated deletion. Given the importance of cholesterol and sphingolipids in neuronal function, we hypothesize that loss of Arv1 in the brain may alter sympathetic outflow to control whole body energy expenditure. To test this, we are generating mice with targeted deletion of Arv1 in key neuronal populations. Contributors: Gupta, Rajat; Fields, David; 138 2014 GRADUATE STUDENT SYMPOSIUM THE FUNCTIONAL ROLE OF ATOH1 IN HAIR CELL FORMATION AND REGENERATION Hsin-I Jen Program in Developmental Biology Advisor: Andrew Groves, Ph.D.-Department of Neuroscience The majority of hearing loss is due to irreversible hair cell death in the cochlea of the inner ear. Hair cells (HCs) are the sensory receptor cells for sound, and are surrounded by supporting cells (SCs). During ear development, the differentiation of HCs requires a transcription factor, Atoh1, which is evolutionarily conserved and is necessary and sufficient for hair cell formation. With the ability to generate cochlear HCs from SCs. Atoh1 based gene therapy has thought to be a promising treatment of deafness. However, its ability to induce trans-differentiation is limited to early postnatal stages. Ectopic expression of Atoh1 in mature SCs does not induce trans-differentiation. To successfully regenerate HCs in the adult cochlea, it is important to understand the mechanisms that prevent Atoh1 from inducing HCs in the mature animal. Since Atoh1 is a transcription factor, we hypothesize that Atoh1 is unable to activate at least some HCspecific genes in adult SCs. This may be due to the alterations in the epigenetic state of its target genes, or absence of transcriptional co-activators, or both. We have identified 10 Atoh1 direct target genes in the mammalian ear using RNA-seq, in situ screen and Atoh1 ChIP-PCR. We aim to study how these targets are epigenetically regulated in neonatal and adult supporting cells. In addition, we are also studying another important HC specific transcription factor, Gfi1, which has been shown to synergize with Atoh1 and recruit repressive chromatin remodeling complexes. Our preliminary data shows that Gfi1 may interact with Atoh1 and alter Atoh1 transcriptional activity. Base on these finding, we hypothesize that Gfi1 might be an important factor in HC formation and maturation, possibly through interaction with Atoh1. Contributors: 139 BAYLOR COLLEGE OF MEDICINE QUANTITATIVE IMAGING OF GLUTATHIONE IN LIVE CELLS USING A REVERSIBLE REACTION-BASED RATIOMETRIC FLUORESCENT PROBE Xiqian Jiang Department of Pharmacology Advisor: Jin Wang, Ph.D.-Department of Pharmacology Glutathione (GSH) plays an important role in maintaining redox homeostasis inside cells. Currently, there are no methods available to quantitatively assess the GSH concentration in live cells. In order to minimize the perturbance on the biological system in live cell imaging, the probe concentration needs to be significantly lower than the analyte concentration. Because of this, any irreversible reaction-based GSH probe will exhibit the maximum response regardless of the GSH concentration. A reversible reaction based probe with an appropriate equilibrium constant allows measurement of an analyte at much higher concentration, and thus is a prerequisite for GSH quantification inside cells. In this contribution, we report the first fluorescent probe CouBro for quantitative imaging of intracellular GSH in live cells and demonstrate the importance of using reversible reactions for ratiometric probe design in order to quantitatively measure the analyte concentrations in biological systems. Contributors: Jiang, Xiqian; Yu, Yong; Zhao, Mingkun; Matzuk, Alexander J.; Chen, Jianwei; Tan, Xiao; Sizovs, Antons; Wang, Meng C.; Wang, Jin 140 2014 GRADUATE STUDENT SYMPOSIUM TORC2 REGULATION OF AGING AND AGE-RELATED MEMORY IMPAIRMENT Jennifer Leigh Johnson Department of Neuroscience Advisor: Mauro Costa-Mattioli, Ph.D.-Department of Neuroscience As our population ages, cognitive decline and dementia are becoming more prevalent. In the United States, there are currently 4.7 million people with dementia. By 2050, the number is expected to double. Memory declines with age indicating crosstalk between these two processes. Yet, mechanisms are not fully understood nor have shared components been identified. This information is critical as different mechanisms of lifespan extension may benefit certain functions but be detrimental to others such as cognition. One important example is the Target of Rapamycin Complex 2 (TORC2) signaling pathway. Given the evolutionary conservation of TORC2 between flies and humans and the relatively short lifespan of Drosophila melanogaster, we measured lifespan in flies lacking rictor and sin1, essential components of TORC2 formation. We found that TORC2 mutants are significantly long lived, suggesting that TORC2 regulates lifespan in the fly. In addition, given that TORC2 is required for long-term memory (LTM) and mTORC2 activity declines with age in mice, we asked whether TORC2 dysregulation contributes to age-related memory impairment. Intriguingly, we found that a specific small molecule that activates TORC2, A-443654, restored the impaired LTM in aged wild-type flies and mice. These results suggest that TORC2 plays a role in age-related memory impairment via an evolutionarily conserved mechanism. Ongoing work will determine if TORC2-regulated memory and aging processes can be uncoupled by targeting distinct cell populations or neural circuitries. The knowledge obtained in this study will lead to a better understanding of the molecular, cellular and neuronal circuit mechanisms underlying aging and age-related cognitive impairment. In addition, this study may also lead to the development of TORC2-based treatments to boost memory in the old but also enhance lifespan. Contributors: Johnson, Jennifer; Huang, Wei; Dierick, Herman; Roman, Gregg; Costa-Mattioli, Mauro 141 BAYLOR COLLEGE OF MEDICINE ROLE OF PROLACTIN PATHWAY IN PREGNANCY STIMULATION OF BREAST CARCINOGENESIS Alyssa N Johnston Program in Translational Biology & Molecular Medicine Advisor: Yi Li, Ph.D.-Department of Molecular & Cellular Biology In 2014, approximately 297,000 new breast cancer cases will be diagnosed in the United States alone with an estimated 39,620 deaths by year’s end. Breast cancer is the most common cancer among women, as well as the second leading cause of cancer death of women in the United States; reducing breast cancer incidence will have a profound impact on saving lives and reducing the tremendous cost associated with treatment. Epidemiological studies indicate that there is a link between increased risk of breast cancer and pregnancy. Previous studies in the Li lab discovered that pregnancy stimulates the progression of pre-existing early lesions to cancer via activation of STAT5, which is normally expressed during pregnancy to maintain lactation. In normal breast epithelium; pSTAT5 is down regulated after lactation to allow the breast to return to a more normal, pre-pregnancy state. However, some pre-cancerous lesions can activate STAT5 in response to pregnancy and lactation hormones and aberrantly maintain high levels of pSTAT5 even after involution. The prolactin receptor (PRLR) is an upstream regulator of pSTAT5 that is activated during pregnancy and later inactivated following lactation. After ligand-mediated activation of PRLR, Janus kinase (JAK) family tyrosine kinases can phosphorylate and activate STAT family proteins to transduce cytokine-mediated signals. We believe that treatment of antipsychotic drugs that suppress dopamine can increase breast cancer risk as these drugs remove the negative feedback of prolactin. Based on previous studies we hypothesize the following: Pregnancy promotes breast cancer via aberrant maintenance of PRLR-JAK/STAT pathway and drugs that increase PRL levels, such as Pimozide, increase breast cancer incidence while drugs that block PRLR/JAK/STAT signaling can prevent it. Aim one: Determine the role of PRLR in pSTAT5 maintenance in parous early lesions. PRLR will be selectively knocked down and then separately over expressed in our mouse model to determine its effect on breast cancer development in the presence of an oncogenic insult. Aim two: Elucidate the role of antipsychotic drug, Pimozide, on breast cancer risk. Nulliparous mice were burdened with an oncogenic insult and then treated with either Pimozide or DMSO control. Aim 3: Examine the dosage and efficacy of the clinically used JAK inhibitor Ruxolitinib. We are currently testing whether Ruxolitinib treatment for a short window of time can prevent STAT5 dependent parity-stimulation of tumorigenesis in mice. This will also move to a phase two clinical trial in high risk women. Contributors: Johnston, Alyssa; Li, Yi 142 2014 GRADUATE STUDENT SYMPOSIUM CRYOEM STRUCTURE OF A NOVEL DINOVERNAVIRUS REVEALS THE SIMPLEST NATURAL REOVIRUS CAPSID Jason T Kaelber Department of Molecular Virology & Microbiology Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology The family Reoviridae contains dsRNA viruses with 9-12 genome segments. In some turreted reoviruses, 260 copies of the peripheral trimer protein cover the inner shell with a T=13l second layer. In others, as few as 60 copies of the trimer are present. Presently, the only exception has been the genus Cypovirus which lacks trimer altogether; Yu and co-workers (2011) have proposed that an extra domain in the T=2 major capsid protein compensates for its absence by stabilizing the single-shelled capsid. We describe the isolation and characterization of the first natural isolate of a nine-segmented reovirus, tentatively designated Fako virus. Fako virus is a mosquitospecific reovirus that was isolated from several mosquito species and there is evidence of vertical transmission of Fako virus in nature. Fako virus is a member of the genus Dinovernavirus, sister clade to Cypovirus. We obtained a cryo-EM structure of Fako virus capsids at subnanometer resolution. Like Cypovirus, Fako virus is a single-layered turreted reovirus and has no peripheral trimers. However, it does not contain the compensatory extra domain found in Cypovirus. Fako virus lacks the clamp protein at the icosahedral three-fold axis, even though this feature is conserved among all other turreted reoviruses. Evolutionary analysis reveals that Dinovernavirus and Cypovirus evolved from a more complex double-layered ancestor through stepwise loss of structural features. The Fako virus capsid contains fewer polypeptides than any other reovirus capsid and does not compensate with domain insertions. The structure and genome of Fako virus demonstrate the dispensability of many conserved reoviral features previously observed. Contributors: Kaelber, Jason; Auguste, A. Jonathan.; Liu, Xiangan; Fokam, Eric; Guzman, Hilda; Jakana, Joanita; Tesh, Robert; Weaver, Scott; Chiu, Wah 143 BAYLOR COLLEGE OF MEDICINE PROTEIN FOLDING AND COLLAPSE: THERMODYNAMICS OF AGGREGATION OF GLY5 VS CONCENTRATION IN SOLUTION Deepti Karandur Program in Structural and Computational Biology and Molecular Biophysics Advisor: B. Pettitt, Ph.D.-Biochemistry Jianpeng Ma, Ph.D.-Department of Biochemistry & Molecular Biology Intrinsically disordered proteins (IDPs) are proteins that do not fold into a stable, three-dimensional, structure, and may only undergo ordering when interacting with other molecules. IDPs tend to be rich in amino acids like glycine, which favor the proteins’ disorderliness. Hence, oligoglycines are a good model to study the behavior of IDPs in aqueous solvent. Experimentally, the solubility of oligoglycine in water decreases as its length increases until, when the peptide contains 5 glycines, it aggregates and falls out of solution at mM concentration. We present results of large scale simulations of over 3 million atoms of several hundred short (five residue) oligoglycines at varying concentrations in explicit solvent. We find that intermolecular interactions between oligoglycines are favored over interactions between oligoglycine and water, leading to their aggregation, viz. concentration effects play a significant role in driving oligoglycines to aggregate and/or collapse. However, the interaction driving peptide associations, liquid-liquid phase separation, are not predominantly hydrogen bonding. We hypothesize that the thermodynamics of aggregation of short oligoglycines is equivalent to the thermodynamics of collapse of longer oligoglycines and similar disordered domains in water. We compare the aggregation of short oligoglycines with the collapse of longer, single oligoglycines in water. Contributors: Karandur, Deepti; Pettittt, B. Montgomery 144 2014 GRADUATE STUDENT SYMPOSIUM UGT2B28 IS A KEY REGULATOR OF ANDROGEN SIGNALING IN PROSTATE CANCER Akash Kumar Kaushik Department of Biochemistry & Molecular Biology Advisor: Arun Sreekumar, Ph.D.-Department of Molecular & Cellular Biology Androgen signaling is the central modulator of prostate cancer (PCa) development and progression. UDP-glucuronosyltransferases (UGTs) are the major glucuronidation enzymes known to participate in the inactivation of androgens in prostate cancer. Previously, we nominated UGT2B28 to be associated with biochemical recurrence in PCa patients and verified its higher expression in castration-resistant or metastatic PCa compared to androgen dependent (AD) localized tumors. To investigate the role of UGT2B28 in AD PCa we knockdown (KD) the gene using two independent lentivirus shRNA clones in LNCaP PCa cells. The KD resulted in an increased expression of androgen receptor (AR), elevated levels of testosterone and its precursors as well as elevated levels of prostate specific antigen (PSA), a classical AR regulated gene. Consistent with these, microarray analysis suggested increased expression of genes involved in biosynthesis of androgens from cholesterol and concomitant down regulations of genes regulating the conversion of cholesterol to bile acids. This apparent increased flux towards steroid biosynthesis was consistent with low steady state levels of bile acids in the KD cells. Interestingly, the KD cells exhibited significantly higher rate of proliferation and migration in vitro while forming higher number of tumors in vivo. Mining the microarray data further suggested that this tumor promoting function of UGT2B28 is potentially caused by rewiring of metabolism towards a more bioenergetic state constituted by altered mitochondrial activity. Targeted metabolic analysis showing elevated levels of citrate, isocitrate, glutamate and glutamine further supported the increased mitochondrial activity. In addition to increase in energy production, the microarray data also described the existence of a senescence signature in the KD cells. Androgen action on prostate cancer cells has been long known to cause proliferation and more recently suggested to be involved in inducing senescence. This is most evident in the biphasic effect seen in LNCaP prostate cancer cells treated with higher concentrations of androgen. This alludes to the importance of fine tuning androgen levels during PCa development with higher levels potentially causing inhibition of cell growth or senescence. Our data described above suggests higher levels of testosterone in UGT2B28 KD cells. In light of the microarray-derived senescence signature, we reasoned that the KD cells may be more sensitive to growth inhibition by low levels of exogenous androgens compared to controls, which was indeed the case when tested. This novel finding is currently being validated in vivo. If verified this will reveal a new therapeutic modality for early stage PCa involving down regulation of UGT2B28 expression to perturb the androgen homeostasis coupled to testosterone therapy to induce tumor growth arrest. Overall, our data for the first time suggests UGT2B28 to be a key regulator of androgen levels in prostate cancer, modulating its proliferative and anti-proliferative activities. Contributors: Kaushik, Akash; Sonavane, Rajni; Putluri, Vasanta; Manikkam, Mohan; Gohlke, Jie; Nagireddy Putluri; Nancy, Weigel; Michailidis, George; Palapattu, Ganesh; Sreekumar, Arun 145 BAYLOR COLLEGE OF MEDICINE HIPPOCAMPAL CIRCUIT DEFICITS IN TWO MOUSE MODELS OF INTELLECTUAL DISABILITY Sara Elizabeth Kee Department of Neuroscience Advisor: Huda Zoghbi, M.D.-Department of Pediatrics Learning and memory deficits are characteristic of Intellectual Disability Disorders (IDDs), however, little is known about how this phenotype arises in so many disorders with vastly different genetic causes. Our hypothesis is that different genetic defects can cause functional deficits in the hippocampal (HP) circuit, during at least one of the stages of memory processing: memory formation, consolidation, or retrieval. HP neurons (place cells) encode spatial memories by firing at specific locations (place fields) in an environment. Thus, to test our hypothesis we used tetrode recording to examine the HP place cells in the CA1 region of naturally behaving animal models of two clinically similar IDDs that are caused by different genes, Rett Syndrome (RTT) and Angelman Syndrome (AS). Available rodent models of RTT (Mecp2+/-) and AS (Ube3amat-/pat+) recapitulate the learning and memory deficits observed in human patients. We have implanted tetrode hyperdrives on four pairs of RTT mice with wildtype (WT) controls and three pairs of AS mice with WT controls. Preliminary examination of AS model place fields suggests reduced spatial information (SI) of place fields as animals run on a novel linear track (p = 0.0001, rank-sum) as compared to WT animals. There was also reduced correlation between place fields in a familiar open field with a cue card and the same field with the cue card removed in AS mice, compared with that in WT mice (Pearson correlation, p = 0.03, rank-sum). These results suggest impaired memory formation and recall, respectively, in AS mice. Preliminary results from RTT mice suggest that the amplitude of ripples, high frequency, high amplitude events that occur in the local field potential of HP CA1, was decreased during slowwave sleep (p < 0.0001, rank-sum), as compared to WT. This suggests a deficit in memory consolidation in RTT mice. Therefore, spatial memory code is disrupted in both animal models, but in a different fashion. Our data suggest that genetic alterations in both the Mecp2 and Ube3a genes can cause functional changes in HP, however, the seemingly similar behavioral deficits caused by these two genetic manipulations are mediated by different circuit mechanisms within the HP. Contributors: KEE, SARA; ZOGHBI, HUDA; JI, DAOYUN 146 2014 GRADUATE STUDENT SYMPOSIUM HIGH LEVELS OF CIRCULATING CXCL10 AT INITIAL DIAGNOSIS ARE ASSOCIATED WITH POOR PROGNOSIS OF OSTEOSARCOMA PATIENTS Aaron Josef Kelly Program in Structural and Computational Biology and Molecular Biophysics Advisor: Tsz-Kwong Man, Ph.D.-Department of Pediatrics Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents. Although survival rates have been relatively constant at 60% to 70% over the past few decades, the prognosis of patients with metastasis at diagnosis is much worse. Identification of biomarkers besides clinically observable metastatic lesions will help to facilitate the risk-stratification of patients more effectively and identify the most appropriate treatment option to improve their outcomes. However, due to complex and highly variable genomic characteristics, coupled with a relatively low prevalence inherent in childhood cancers, it has been exceedingly difficult to discover tumor biomarkers in OS. Circulating cytokines and chemokines provide an excellent alternative to tumor biomarkers since they can be detected non-invasively through patients’ peripheral blood samples. Levels of these known cancerrelated proteins may be tumor derived or they may correspond to immune responses that occur during tumor pathogenesis. In this study, a luminex assay was employed to analyze 39 chemokines/ cytokines in two cohorts comprised of totally 290 OS patients. A regressive partitioning method was used to binarize the variables in the training cohort (40 samples). 24 of the candidate biomarkers were significant with respect to overall survival. The cutoffs derived from the training cohort were applied to the validation cohort (250 samples) for each of the 24 variables. After Benjamini-Hochberg correction, CXCL10 significantly correlated with overall survival (p=.047). After controlling for the known prognostic factor, i.e. initial metastasis, CXCL10 remained significant (CXCL10: p=0.0037, Metastasis: p=0.00012), indicating that CXCL10 is an independent prognostic factor. Furthermore, when the two prognostic factors were combined, we found that the patients with no initial metastasis and low CXCL10 had a 71% survival rate, those with initial metastasis and high CXCL10 had 31% survival rate and those with one or the other had 54% survival rate at 5 year follow-up. In the non-metastatic patients alone, CXCL10 levels were able to further significantly risk stratify patients (p=0.019), indicating that high CXCL10 can help to identify high-risk patients that did not have detectable metastatic lesions at diagnosis. Lastly, compared to non-tumor controls, the “high CXCL10” group of patients had significantly higher protein expression (p=3.4 e-05) whereas “low CXCL10” patients were not significantly different (p=0.16). These analyses indicate that circulating CXCL10 is a novel prognostic factor that is independent of metastasis at diagnosis. Our results showed that they can be used in conjunction to provide a more accurate riskstratification of OS patients in a non-invasive manner. Finally, CXCL10 is a ligand of CXCR3, which is known to be related to cancer progression and metastasis. A majority of primary OS tumors express CXCR3, which is targetable and may lead to a personalized therapy for patients with high levels of CXCL10. Contributors: Kelly, Aaron; Flores, Ricardo; Nakka, Manjula; Li, Yiting; Lau, Ching; Hicks, John; Man, Tsz-Kwong 147 BAYLOR COLLEGE OF MEDICINE CIRCADIAN HOMEOSTASIS OF LIVER METABOLISM SUPPRESSES HEPATOCARCINOGENESIS Nicole M Kettner Department of Molecular & Cellular Biology Advisor: Loning Fu, Ph.D.-Department of Pediatrics Hepatocellular carcinoma (HCC), a major type of liver tumor in humans, was previously considered a rare cancer in Western countries, but shows an average increase of 3.5% annually among men and woman since 1992 and is one of the top cancer-related deaths in the U.S. This increase in HCC in the U.S. parallels the obesity epidemic, and obesity-related metabolic disorders including nonalcoholic fatty liver disease (NAFLD) and diabetes mellitus which have recently been identified as novel risk factors for HCC worldwide. We have previously reported that chronic disruption of circadian light cues following a schedule similar to human night shift-work schedule promotes tumor development, and that HCC is the second most commonly observed primary lesion induced by circadian disruption in mice. Further study of the mechanism of chronic circadian dysfunction-induced cancer risk has led to the discoveries that chronic circadian disruption increases the risk of obesity by disruption of homeostasis of neuroendocrine function, which induces liver metabolic malfunction prior to HCC development, including a dramatic increase in the risk of hepatomegaly, NAFLD, liver inflammation, and fibrosis. Genome-wide microarray analysis identified a large number of deregulated pathways prior to HCC detection in the livers of mice lacking circadian homeostasis, including all core clock genes, well-established molecular markers for human HCC, and those controlling the biosynthesis and metabolism of redox, steroids, vitamins, nucleotides, carbohydrates, triglycerides, bile acids, and glycogen leading to global deregulation of liver metabolism as detected by metabolomics studies. Thus, we conclude that chronic circadian disruption is an independent risk factor for HCC. Contributors: Kettner, Nicole M.; Katchy, Chinenye A; Finegold, Milton J.; Moore, David D.; Fu, Loning 148 2014 GRADUATE STUDENT SYMPOSIUM IDENTIFICATION OF KID3 AS A POTENTIAL REGULATOR OF OSTEOCYTE DIFFERENTIATION Jordan Kho Program in Developmental Biology Advisor: Brendan Lee, M.D./Ph.D.-Department of Molecular & Human Genetics Osteocytes are the most abundant component of bone in the body, making up nearly 95% of all bone cells. They are derived from osteoblasts and become embedded under the mineralized bone matrix upon differentiation. Recent studies over the past several years unraveled the diverse physiological roles of osteocytes in regulating bone remodeling, mineral homeostasis, mechanosensing, hematopoietic stem cell mobilization, and most recently, fat metabolism. Despite their important roles, little is known about the molecular mechanisms that govern osteocyte differentiation. To identify candidate transcription factors that are involved in regulating osteocyte differentiation, we performed TRANSFAC analysis of the promoter regions of known osteocyte-specific genes. We identified a 5-base pair CCACA motif that is highly enriched in the promoter region of all of these genes. This motif is known to be the consensus binding site for Kid3 (also called Zfp354c and AJ18), a Krüppel associated box (KRAB) domain-containing C2H2 zinc finger transcription factor. Gene expression analysis revealed that Kid3 is highly expressed in osteoblast and its expression is significantly down-regulated upon osteocyte differentiation. To investigate how Kid3 regulates the expression of osteocyte genes, we generated a stable osteocyte cell line (Ocy454) overexpressing Kid3 in an inducible manner. We found that expression of sclerostin (SOST) and Dmp1, well-known markers of mature osteocytes, were significantly decreased in Kid3-overexpressing Ocy454 cells at the late stage of differentiation, suggesting a potential role of Kid3 in suppressing osteocyte differentiation. To further understand in vivo roles of Kid3 during osteocyte differentiation, we generated transgenic mice overexpressing Kid3 specifically in osteoblast (driven by 2.3 kb mCol1a1 promoter) or osteocyte (driven by 10 kb Dmp1 promoter). Our current work focuses on the phenotypic characterization of these transgenic mice. Contributors: Bertin, Terry; Chen, Yuqing; Zeng, Huan-Chang; Bae, Yangjin; Spatz, Jordan; Pajevic, Paola; Brunetti-Pierri, Nicola; Lee, Brendan 149 BAYLOR COLLEGE OF MEDICINE TRANSCRIPTIONAL PROGRAMS GOVERNING ACTIVITY-DEPENDENT MATURATION OF ADULT-BORN NEURONS Cynthia J Kim Department of Molecular & Human Genetics/M.D.-Ph.D. Program Advisor: Benjamin Arenkiel, Ph.D.-Department of Molecular & Human Genetics Our lab aims to uncover new regulatory pathways that control activity-dependent survival and integration of neural precursors in the adult brain. These studies may be useful towards development of cell repair or renewal approaches in neuro-regenerative therapy. The rodent olfactory system features robust and lifelong neurogenesis and thereby serves as a well-characterized model to examine the mechanisms of synaptogenesis and circuit rewiring in the adult brain. We recently reported a novel mechanism that influences adult-born neuron circuit integration. Namely, adult-born neurons that receive pre-synaptic input from local interneurons, which secrete the neuropeptide corticotropin-releasing hormone (CRH), show increased propensity for survival and integration. These studies defined a new role for CRH, apart from its wellknown functions in the systemic stress response. Microarray gene expression analysis of gain- and loss-of-function animal models of local CRH signaling revealed that local CRH signaling dynamically regulates expression of the brain-specific Homeoboxcontaining transcription factor Brain-5 (BRN5). Overactive CRH signaling upregulates transcription of BRN5, whereas decreased CRH signaling downregulates transcription of BRN5. BRN5 is described as a pro-neural differentiation factor in embryonic neural precursors, and its expression remains high in the neurogenic olfactory areas of the adult brain. However, little is known about the function of BRN5 in adult-born neuron development. To elucidate the role of BRN5 in neuronal development in the adult brain, we use biochemical, genetic, and electrophysiological techniques to test the hypothesis that BRN5 influences synaptic maturation in adult-born neurons. Contributors: Kim, Cynthia; Garcia, Isabella; Arenkiel, Benjamin R 150 2014 GRADUATE STUDENT SYMPOSIUM TUMOR-INDUCED HETEROGENEITY OF MYELOID CELLS AND THEIR ROLE IN BREAST CANCER PROGRESSION Ik Sun Kim Integrative Program in Molecular and Biomedical Sciences Advisor: Xiang Zhang, Ph.D.-Department of Molecular & Cellular Biology Background: Tumors evolve immunosuppressive microenvironment to subvert anti-tumor immunity. Among cancer promoting inflammatory effectors, tumor-associated myeloid cells (TAMCs) represent a heterogeneous group that predominantly orchestrates tumor-induced immunosuppression as well as many other hallmarks of cancer. It has become increasingly clear that these immunosuppressive mechanisms elicited by TAMCs are a major barrier to effective anti-tumor therapies. What remains poorly understood, however, is why and how individual tumors evolve to employ different mechanisms to suppress anti-tumor immunological response. Experimental Design and Methods: To delineate the immune system disorder in the context of heterogeneous breast cancer, we utilized p53-null syngeneic mammary tumor models that highly resemble different human breast cancer subtypes. Various lymphoid and myeloid cells have been profiled by multi-color flow cytometry in blood, bone marrow, and primary tumor at different stages of tumor growth. Results: We have demonstrated heterogeneous immunosuppression phenotype across p53-null tumor models conferred by various expansion and differentiation of CD11b+ Gr1+ myeloid cells. Interestingly, these myeloid cells exhibited inter- and intratumoral heterogeneity in terms of the initial expression of surface antigens, cellular plasticity upon infiltrating tumors, and functional roles in tumor progression. Conclusions: This work highlights the dramatic phenotypical and functional heterogeneity of TAMCs among different subtypes of breast cancer. Identification and intervention of such tumor-specific immune aberrations will likely provide a rationale to classify patients for different cancer immunotherapies. Contributors: Kim, Ik Sun; Welte, Thomas; Rosen, Jeffrey; Zhang, Xiang 151 BAYLOR COLLEGE OF MEDICINE THE IMPACT OF OPTION B+ ON THE ANTENATAL PMTCT CASCADE IN LILONGWE, MALAWI Maria Hyoun Kim Clinical Scientist Training Program Advisor: Elizabeth Chiao, M.D./M.P.H.-Department of Medicine Objective: In July 2011, Malawi implemented Option B+ (B+), lifelong antiretroviral therapy (ART) for pregnant and breast-feeding women. We aimed to describe changes in service uptake and outcomes along the antenatal PMTCT cascade post B+ implementation. Design: Pre/post study using routinely collected program data. Methods: We compared testing of HIV-infected pregnant women at antenatal care, enrollment into PMTCT services, receipt of ART and six month ART outcomes pre- (Oct 2009-Mar 2011) and post- (Oct 2011-Mar 2013) B+. Results: A total of 13,926 (pre) and 14,532 (post) women presented to antenatal care. Post-B+ a smaller proportion were HIV tested (99.3% vs. 87.7% post-; p<0.0001). There were 1654 (pre) and 1535 (post) HIV-infected women identified, with a larger proportion already known to be HIV-infected (18.1% vs. 41.2% post-; p<0.001) and on ART post-B+ (18.7% vs. 30.2% post-; p<0.001). A significantly greater proportion enrolled into the PMTCT program (68.3% vs. 92.6% post-; p<0.001) and was retained through delivery post-B+ (51.1% vs. 65% post-; p<0.0001). Amongst those not already on ART at enrollment there was no change in the proportion newly initiating ART/ARVs (79% vs. 81.9% post-; p=0.11); although median days to initiation of ART decreased (48d [19,130] vs. 0d [0,15.5] post-; p<0.001). Amongst those newly initiating ART, a smaller proportion was alive and on ART six months post-initiation (89.3% vs. 78.8% post-; p=0.0004). Conclusion: While several improvements in PMTCT program performance were noted with implementation of B+, challenges remain at several critical steps along the cascade requiring innovative solutions to ensure an AIDS-free generation. Contributors: Kim, Maria H. *1,2,§; Ahmed, Saeed*1,2; Hosseinipour, Mina C. 3,4; Giordano,Thomas P. 5; Chiao, MD, Elizabeth Y. 5; Yu, Xiaoying 6; Nguyen, Chi6; Chimbwandira, Frank 7; Kazembe, Peter N. 2; Abrams, Elaine J. 8,9 1Baylor College of Medicine, Department of Pediatrics, Section of Retrovirology, Houston, Texas, USA 2Baylor College of Medicine-Abbott Fund Children’s Clinical Center of Excellence, Lilongwe, Malawi. 3University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA. 4UNC Project, Lilongwe, Malawi 5Department of Medicine, Baylor College of Medicine, Houston TX 6Department of Pediatrics, Epidemiology Center, Baylor College of Medicine, Houston TX 7Department of HIV and AIDS, Ministry of Health, Lilongwe, Malawi 8ICAP-Columbia University, Mailman School of Public Health, New York NY, USA 9College of Physicians & Surgeons, Columbia University, New York, NY, USA 152 2014 GRADUATE STUDENT SYMPOSIUM MIR-200C ALTERS THE EPITHELIAL PROGRAM AND DIFFERENTIATION STATUS WITHIN A CLAUDIN-LOW BREAST CANCER MODEL, IMPAIRING CHEMORESISTANCE AND METASTATIC POTENTIAL IN VIVO Jana Knezevic Department of Molecular & Cellular Biology Advisor: Jeffrey Rosen, Ph.D.-Department of Molecular & Cellular Biology Claudin-low tumors are a highly aggressive breast cancer subtype with no targeted treatments and a clinically documented resistance to chemotherapy. The significant enrichment in Cancer Stem Cells (CSCs) makes claudin-low tumors an attractive model forstudying CSC behavior in order to develop different approaches to minimize CSC therapy resistance. The epithelial-mesenchymal transition (EMT) is one mechanism by which CSCs arise, and its reversal may provide a potential therapeutic approach for increasing tumor chemosensitivity. We therefore investigated the miR-200 family of microRNAs in the regulation of the epithelial state within a primary p53null claudin-low tumor model, normally deficient in miR-200 expression, and its subsequent impact on stem-like properties and therapeutic response in vivo. We show that restoration of miR-200c cluster within this claudin-low model changes the epithelial state, and consequently, impedes CSC behavior within these mesenchymal tumors. Moreover, these state changes are accompanied by a decrease in proliferation and differentiation status. MiR-200c expression also forces a significant reorganization of tumor architecture, affecting important cellular processes involved in cell-cell contact, cell adhesion and motility. Accordingly, the chemosensitivity of the p53null claudin-low tumors was significantly enhanced upon restoration of miR200c expression, along with a decrease in the metastatic potential. Collectively, our data suggest that miR-200c reexpression in claudin-low tumors offers a potential therapeutic application to disrupt the EMT program on multiple fronts within this mesenchymal tumor subtype, altering tumor growth, chemosensitivity and metastatic potential in vivo. Contributors: Knezevic, Jana; Pfefferle, Adam; Petrovic, Ivana; Perou, Charles; Rosen, Jeffrey 153 BAYLOR COLLEGE OF MEDICINE THE HUMAN ADENOVIRUS E4-ORF1 PROTEIN HIJACKS DISCS LARGE 1 TO MEDIATE MEMBRANE RECRUITMENT AND DYSREGULATION OF PHOSPHOINOSITIDE 3-KINASE Kathleen Kong Department of Molecular Virology & Microbiology Advisor: Ronald Javier, Ph.D.-Department of Molecular Virology & Microbiology Human adenoviruses cause acute illnesses associated with respiratory, gastrointestinal, and ocular infections. In addition to their use as vectors for vaccination and gene and cancer therapy, adenoviruses also serve as tools for revealing mechanisms of cancer due to their tumorigenic potential in experimental animals. The human adenovirus E4-ORF1 gene encodes an oncoprotein that enhances viral replication by activating cellular phosphoinositide 3-kinase (PI3K). While the underlying mechanism of activation is not known, this activity depends on a complex formed between E4-ORF1 and the cellular PDZ protein Discs Large 1 (Dlg1). Mass spectrometry analysis of cellular proteins associated in vitro with an E4-ORF1 protein fused to glutathione S-transferase identified the PI3K regulatory subunit p85 as a candidate binding partner of E4-ORF1. Confirming and extending this observation, I demonstrated that in human epithelial cells, E4-ORF1 directly interacts with both the p85 regulatory and p110 catalytic subunits of PI3K and elevates their levels, the latter effect of which, like PI3K activation, requires Dlg1. I also showed that E4-ORF1, PI3K, and Dlg1 assemble into a ternary complex located at the plasma membrane. At this site, Dlg1 co-localized with the activated PI3K effector protein Akt, supporting the idea that the ternary complex mediates PI3K signaling. Signifying the functional significance of the ternary complex, the capacities of E4-ORF1 to induce soft agar growth and focus formation in cells were ablated either by a mutation that prevents E4-ORF1 binding to Dlg1 or by a PI3K inhibitor drug. These results reveal that E4-ORF1 interacts with Dlg1 and PI3K to assemble a ternary complex where E4-ORF1 subverts the Dlg1 oncogenic function to relocate cytoplasmic PI3K to the membrane for constitutive activation. This novel mechanism may serve as a paradigm to understand PI3K activation mediated by other pathogenic human viruses, including influenza A virus, human papillomavirus, and human T-lymphotropic virus type I, which likewise target Dlg1 in infected cells. Thus, studies of human adenovirus E4-ORF1 may yield mechanistic insights that aid development of new therapeutics to treat both viral infections and human cancers. Contributors: Kong, Kathleen; Kumar, Manish; Taruishi, Midori; Javier, Ronald T. 154 2014 GRADUATE STUDENT SYMPOSIUM STUDY OF ENTRY MECHANISM AND RESTRICTION FACTORS OF THE HEPATITIS B VIRUS IN MICE Robert Layne Kruse Program in Translational Biology & Molecular Medicine/M.D.-Ph.D. Program Advisor: Karl-Dimiter Bissig, M.D./Ph.D.-Department of Molecular & Cellular Biology Fasiha Kanwal, M.D.-Department of Medicine Hepatitis B Virus (HBV) is a major global health problem with 350 million people chronically infected, resulting in increased risk of cirrhosis and hepatocellular carcinoma. In order to evaluate new vaccines, drugs, and immunotherapies against HBV, testing in an immunocompetent small animal model resembling human infection is crucial – a model that currently does not exist since human HBV only productively infects humans and chimpanzees. The holy grail of HBV research would be the establishment of a transgenic immunocompetent mouse model permissible to all stages of natural HBV infection. After years of limited understanding of HBV entry, the HBV field was transformed over past two years with the discovery of the first validated receptor for HBV, sodium taurocholate cotransporting polypeptide (NTCP). The discovery of the receptor prompted speculation that a small animal model infectious for HBV by expressing human NTCP (hNTCP) could be generated. This goal is similar in spirit to the successful entry and infection of human hepatitis C virus into mice expressing human CD81 and human occludin (Dorner et al, Nature 2013). Unfortunately, published results by several groups have indicated that human NTCP alone does not confer permissiveness to HBV infection in murine liver cell lines, in contrast to the results with hepatitis delta virus (HDV) harboring the same viral envelope (Li et al, Cell Mol Immunol 2014). Our preliminary results in mice in vivo have confirmed these results. Given that once the HBV genome is introduced inside the mouse hepatocyte nucleus all subsequent steps of the viral life cycle are functional, this suggests that one or more of the steps during HBV entry and covalently closed circular DNA (cccDNA) genome formation are restricted in mice. Our research seeks to study the basic biology of these potential restriction factors, in order to discover the point of block during infection. We hypothesize that HBV is not restricted at the steps of nuclear import and cccDNA formation and amplification, but rather an intracellular fusion step is limiting. A novel intracellular receptor triggering PreS domain rearrangement and exposure of the fusion loop within the viral envelope may be lacking in mouse hepatocytes. This hypothesis does not exclude, however, that these other processes are inefficient and might be limiting as well inside mouse hepatocytes. Our first aim will be to characterize HBV entry mechanisms using our humanized mouse model and virion labeling in vitro. To study intracellular restriction, I will create novel mouse cell lines to study cccDNA in the murine context. Contributors: Kruse, Robert; Minor, Marrisa; Slagle, Betty; Bissig, Karl-Dimiter 155 BAYLOR COLLEGE OF MEDICINE REACTIVATION OF EMBRYONIC GENE SOX11 PROMOTES SKIN TUMORIGENESIS Amy Tsu Ku Program in Translational Biology & Molecular Medicine Advisor: Hoang Nguyen, Ph.D.-Department of Molecular & Cellular Biology Abdul Diwan, Ph.D.-Department of Pathology & Immunology Embryonic stem cell signature genes are activated in various types of cancers. Sox11, a SRY box-containing transcription factor, plays a critical role in embryonic development. Recent studies show that Sox11 has a tumor-promoting role in mantle cell lymphoma, breast cancer and ovarian cancer. Since I found that Sox11 expression is reactivated in the murine skin squamous cell carcinoma (SCC), I hypothesize that aberrant expression of embryonic gene Sox11 contributes to skin tumorigenesis. To determine the oncogenic potential of Sox11, I tested how gain and loss of function of Sox11 affects skin tumorigenesis using the two-stage chemical carcinogenesis model of skin SCC. I found that Sox11 overexpression increases tumor incidence and multiplicity; whereas Sox11 deficiency reduces tumor formation and growth. These results demonstrate that Sox11 has a tumor-promoting role in skin SCC. I then performed transcriptional profiling to identify downstream target genes that are altered by Sox11 overexpression. I identified Lef/Tcf genes, which are known as β-catenin DNA-binding partners in the canonical Wnt signaling pathway, as the downstream targets of Sox11. I also found that Sox11 overexpression increases β-catenin transactivation activity. Our data suggest that Sox11 reactivation promotes tumor growth by altering β-catenin activity. In summary, our novel tet-inducible Sox11 transgenic mouse model, in combination with the previously described Sox11 conditional knockout mouse, allow us to establish for the first time the role of Sox11 as a tumor promoter in skin SCC. Our findings could directly impact diagnosis, prognosis and treatment of this common malignant form of skin cancer. Contributors: Ku, Amy T; Miao, Qi; Nguyen, Hoang 156 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF CD1D-RESTRICTED NKT CELLS IN THE IMMUNE RESPONSE TO SALMONELLA-BASED RECOMBINANT CANCER VACCINE Suhrab Kurbanov Department of Pathology & Immunology Advisor: Leonid Metelitsa, M.D./Ph.D.-Department of Pediatrics Our group is developing a novel therapeutic cancer vaccine platform, which utilizes molecular machinery of Salmonella for effective delivery of tumor-associated antigens into the cytosol of the antigen presenting cells in situ. An attenuated strain of S. typhimurium, MvP728 has been engineered to express human survivin and the resulted vaccine (MvP728-survivin) induced potent CD8 T cell-mediated anti-tumor responses in a highly aggressive murine A20 lymphoma model. The vaccine therapeutic efficacy was further enhanced in combination with an exogenuous ligand for Natural Killer T cells, leading to complete regression of 5-day established A20 tumor grafts in mice and tumor-free survival for at least 60 days in 8 of 8 vaccinated animals. Although this data indicates that exogenuous stimulation of NKT cells can augment the vaccineinduced anti-tumor immune response, the mechanistic role of the NKT/APC interaction in response to Salmonella-based cancer vaccine remains elusive. To assess the direct contribution of NKT cells in response to MvP728, the vaccine-specific CD8 T cellmediated immune response generation were compared in Ja18-/- (lacks type-I NKT cells), Cd1d-/- (lacks all NKT cells), and WT mice. Surprisingly, we found that NKT deficiency significantly augments the Salmonella-based cancer vaccine-induced immune response, resulting in higher antigen-specific CD8 T cell-mediated immune response in the blood and spleen of Black 6 mice. We also found that NKT deficiency results in higher persistence of the vaccine in the spleen and mesenteric lymph nodes. In summary, our results so far show that both exogenuous stimulation with potent NKT ligands as well as the NKT deficiency enhance the vaccine-induced immune response. We hypothesize that exogenuous stimulation of NKT cells with potent ligands renders NKT cells unresponsive for any stimulation in the course of vaccination imitating the NKT deficiency. Therefore, we will further investigate the mechanism of exogenuous stimulation of NKT cells at different time points during the course of vaccination with the MvP-728. Contributors: Suhrab Kurbanov, Xin Xu, Amy Courtney, Linjie Guo, Xiuhua Gao, and Leonid Metelitsa 157 BAYLOR COLLEGE OF MEDICINE ENHANCING CHEMOTHERAPEUTIC RESPONSE BY BLOCKING PGE2 – MEDIATED RECRUITMENT OF CANCER STEM CELLS FROM REPOPULATING RESIDUAL TUMORS Antonina V Kurtova Program in Translational Biology & Molecular Medicine Advisor: Keith Chan, Ph.D.-Department of Urology Seth Lerner, M.D.-Department of Urology Cytotoxic chemotherapy remains a major therapeutic option for a wide spectrum of epithelial cancers including urothelial carcinoma. While chemotherapy is highly effective in debulking the tumor mass and improving survival, certain patients show initial response but their tumors eventually become unresponsive after multiple chemotherapy cycles. Currently the identity of repopulating cancer cells following chemotherapy is unknown, and the underlying molecular mechanisms that initiate tumor repopulation remain poorly understood. Here we hypothesize that cancer stem cells (CSCs) may be recruited to repopulate chemotherapy induced damage within residual tumors, similar to how normal resident tissue stem cells mobilize to wound sites for tissue repair. To test this hypothesis we utilized our previous data showing that cytokeratin 14 (CK14) marks the most primitive urothelial carcinoma cells and abundance of CK14+ cancer cells in patients correlates with poor survival. We found that exposure to one cycle of gemcitabine and cisplatin (GC) chemotherapy effectively reduced the size and growth rate of tumors in vivo. We then followed the regular clinical regimen with a gap period to allow recovery of normal tissues and found a generalized expansion of CK14+ cancer cells in residual tumors during the time between treatment cycles. Next we investigated whether cancer cells are stimulated to proliferate in between chemotherapy courses to repopulate residual tumors and demonstrated the increase of proliferating CK14+ cells. We also found that prostaglandin E2 (PGE2) released by neighboring apoptotic cells induced CSC expansion in a paracrine manner. This repopulation could be abrogated by a PGE2 neutralizing antibody and Celecoxib drug-mediated blockade of PGE2 signaling. In vivo administration of Celecoxib significantly attenuated progressive manifestation of chemoresistance in xenograft tumors, including primary xenografts derived from a patient who failed chemotherapy. Here we demonstrated a new mechanism by which CSCs contribute to therapeutic resistance, via repopulating residual tumors between chemotherapy cycles. This repopulation occurs by the recruitment of quiescent CSCs to divide and “repair” chemotherapy-induced damage. Celecoxib blocked CSC repopulation following the first chemotherapy cycle and consequently improved chemotherapeutic response in a second cycle of chemotherapy. These findings reveal a new mechanism in the development of clinically relevant chemoresistant cancer, and provide a new paradigm to enhance chemotherapeutic response by abrogating early “wound repair”-like CSC repopulation. Contributors: Kurtova, Antonina; Xiao, Jing; Pazhanisamy, Senthil; Ho, Philip; Krasnow, Ross; Lay Erica; Chan, Keith 158 2014 GRADUATE STUDENT SYMPOSIUM METABOLIC REGULATION BY THE MeCP2 TRANSCRIPTIONAL COREPRESSOR COMPLEX POINTS TO NEW THERAPEUTIC TARGETS IN RETT SYNDROME Stephanie Marie Kyle Department of Molecular & Human Genetics Advisor: Monica Justice, Ph.D.-Department of Molecular & Human Genetics David Nelson, Ph.D.-Department of Molecular & Human Genetics Metabolic dysregulation can lead to downstream pathogenesis in nearly all tissues and organ systems. In recent decades, a large body of data has implicated metabolic perturbations in neurological development and degeneration. In particular, dysregulation of cholesterol trafficking and biosynthesis are responsible for the onset of Neimann-Pick type C and Smith-Lemli Opitz syndrome, respectively. Furthermore, Fragile X syndrome, Alzheimer, Parkinson, and Huntington diseases have all been linked to aberrant cholesterol homeostasis. Rett syndrome (RTT) is a progressive neurodevelopmental disorder of females primarily caused by mutations in the X-linked gene encoding methyl-CpG binding protein 2 (MECP2). To identify pathways in disease pathology for therapeutic intervention, we carried out a dominant random mutagenesis suppressor screen in Mecp2 null mice1. One suppressor identifies a stop codon mutation in a rate-limiting enzyme in cholesterol biosynthesis, which ameliorates RTTlike symptoms and increases longevity in Mecp2 null mice by altering brain cholesterol homeostasis. Although RTT has been classically labeled a neurological disorder, these studies suggest that a metabolic component contributes to pathology. Here we show that the Mecp2 mutation induces metabolic defects in mice including fatty liver, increased lipolysis, and insulin resistance in muscle and adipose. These studies inform highly targetable therapeutic pathways relevant to treating RTT; remarkably, statin drug administration improves motor symptoms and confers increased longevity in Mecp2 null mice. The suppressor mutation also suggests that symptoms may be modified in patients by mutations in genes that affect metabolism. In support of this idea, a subset of RTT patients has increased serum cholesterol and triglycerides, independent of body mass index. Our ongoing studies point to additional metabolic pathways that are prime targets in the pursuit of preventing morbidities associated with Rett syndrome. Contributors: Kyle, Stephanie M.; Buchovecky, Christie, M.; Justice, Monica J. 159 BAYLOR COLLEGE OF MEDICINE INTEGRATING MAGNETIC RESONANCE SPECTROSCOPY AND DIFFUSION TENSOR IMAGING INTO A MULTIMODAL MODEL OF MULTIPLE SCLEROSIS BIOMARKERS Andrew Marc Laitman Program in Structural and Computational Biology and Molecular Biophysics/M.D.-Ph.D. Program Advisor: Mirjana Maletic-Savatic, M.D./Ph.D.-Department of Pediatrics Multiple Sclerosis is an autoimmune neurodegenerative disorder that affects the central nervous system and is characterized by demyelinating lesions separated by space and time. Conventional Magnetic Resonance Imaging (MRI) can detect demyelinating lesions in white matter areas, but only after significant pathology has occurs. Similarly, Magnetic Resonance Spectroscopy (MRS) is able to distinguish metabolic changes in between the white matter of the frontal lobe in healthy controls (CTWM), the frontal lobe in patients with MS that appears to be normal by conventional MRI (NAWM - normal appearing white matter), and periventricular non-enhancing lesions (NELES). Furthermore, Diffusion Tensor Imaging (DTI) has been able to identify abnormal diffusivity in NAWM in MS patients. Each modality seems to be able to detect abnormalities that are not detectable by normal MRI. None of the current imaging biomarkers for MS are sensitive nor specific enough to detect the pathological process before symptoms occur. Since MS is characterized by recurrent attacks on the nervous system, a way to predict the subsequent attack would be incredibly useful. We propose to integrate these different modalities into a holistic model to classify patients. In MS patients and healthy controls, we have collected MRI, MRS, and DTI data in the same patient. While each modality has been used to compare MS patients and and healthy controls, no model has been able to combine all 3 imaging modalities. Contributors: Laitman, Andrew; Liu, Zhandong; Maletic-Savatic, Mirjana 160 2014 GRADUATE STUDENT SYMPOSIUM WHAT LIMITS BEHAVIORAL PRECISION? BAD NOISE OR BAD DECODING? Janakiraman Kausik Lakshminarasimhan Department of Neuroscience Advisor: Dora Angelaki, Ph.D.-Department of Neuroscience Single neurons in the macaque medial superior temporal (MSTd) and ventral intraparietal (VIP) cortices are tuned to the animal’s heading direction, but their relative roles in heading perception are unknown. We inactivated area VIP in monkeys trained to discriminate heading and found no deficits in performance. In contrast, earlier results demonstrated that inactivating area MSTd is detrimental to behavior. This is surprising because responses of VIP neurons are more correlated with behavioral choices than those in MSTd. We attempted to resolve this paradox by analyzing neural data recorded from these two areas to evaluate potential decoding strategies, in the context of a mathematical theory of population codes. First, we demonstrate that the observed pattern of choice probabilities (CPs) rules out strictly optimal decoding. Second, we evaluate the degree of optimality by considering two encoding models with fundamentally different noise correlations, one model having extensive information that scales with population size, and the other having information that saturates to a smaller value. We combine each model with the observed CPs to infer decoding weights and decoding efficiency, and predict consequences of inactivating MSTd or VIP. The decoding model with limited information is only mildly suboptimal, whereas the model with extensive information is terribly suboptimal. Although both population models are qualitatively consistent with inactivation results, the latter can account for our inactivation results only under a fragile coincidence that offers a clear and surprising prediction for future experiments: weak inactivation of VIP should improve performance, while strong inactivation should not change performance. Contributors: Lakshminarasimhan, Janakiraman Kaushik; Liu, Sheng; Gu, Yong; DeAngelis, Gregory; Pitkow, Xaq; Angelaki, Dora 161 BAYLOR COLLEGE OF MEDICINE EX VIVO EXPANDED MULTI-SPECIFIC CYTOTOXIC T CELLS DERIVED FROM HIV+ PATIENTS USING GMP-COMPLIANT METHODOLOGIES RECOGNIZE MULTIPLE HIV ANTIGENS AND SUPPRESS HIV REPLICATION Sharon Lam Department of Pathology & Immunology Advisor: Catherine Bollard, M.B.,B.Ch.-Department of Pediatrics Cliona Rooney, Ph.D.-Department of Pediatrics Anti-retroviral therapy (ARVs)(??ART??) does not eliminate HIV from latently -infected reservoirs, has long-term toxicities and fails to fully prevent immune attenuation. Therefore there is a need for alternative therapies that will decrease dependency on ARTVs. Previous studies have demonstrated the safety and feasibility of infusing single-epitope specific CD8 T cells or artificial T cell receptor- transduced T cells to HIV+ patients. However, these T cells were restricted to a single HLA restricted epitope and had limited persistence in vivo. Hence, we hypothesized that broadly HIV-specific T cells could be expanded from patients on ARTV to effectively target HIV infection using a non-HLA restricted, GMP-compliant approach. We developed a method by which PBMCs from patients on ARTV were stimulated with antigen presenting cells pulsed with gag, pol, and nef peptide libraries (pepmixes) in the presence of growth, Th1, and memory promoting cytokines and co-cultured with co-stimulatory K562 cells. Starting from 60 to 100 mL of blood, T cells expanded to clinically relevant numbers (Mean=1.62e8 cells, Range (3.72e7, 2.87e8 cells), n=7) in the presence of ART to prevent possible viral spread during expansion. The majority of the expanded T cells had an effector memory phenotype (CD3+CD45RO+CD62L-) with approximately 10% suggestive of a central memory phenotype (CD3+CD45RO+CD62L+) which is important for long-term persistence of T cells in vivo. Post-expansion, 5 of 7 patient sample lines showed specific activity to all 3 HIV antigens in IFNγ ELISPOT assays, with the remaining 2 showing specificity to 1 of 3 antigens. The T cell lines were broadly -specific to gag (mean=99.33 SFC/10e5 cells), pol (mean=131.11 SFC/10e5 cells) and nef (mean=337.26 SFC/10e5 cells), and polyclonal as shown by flowbased Vβ usage analysis (mean usage= 14.67 of the 24 Vβ analyzed). Importantly, T cells expanded from both ART patients were cytotoxic, as e. Expanded T cells lysed antigen loaded autologous targets (mean=67.55% specific lysis compared to absence of antigen: mean=0.46% at 10:1 effector target ratio) in 51Cr release assays. Expanded T cells from ART patients also showed a greater ability to suppress HIV outgrowthreplication in vitro compared to unexpanded CD8 T cells when co-cultured with reactivated resting CD4+ T cells from ART-suppressed HIV+ patients, the authentic latently infected cells that define viral latent reservoirs in treated patients compared to unexpanded CD8 T cells, which presumably contains pre-existing HIV-specific T cells. In 5 patients on ART a statistically lower recovery of virus from resting CD4+ cells was seen in the presence of CTLs as compared to no effectors (p<0.006 by Mann Whitney), while the unexpanded CD8 cells showed only a modest trend towards decreased recovery that was not statistically significant (p>0.9). We have developed robust GMP-compliant methodologies for expanding functional HIV-specific T cells from both HIV+ patients and HIVneg for autologous and third-party use, respectively. We now plan to translate our approach to the clinical setting where we will testuse HIV-polyspecific T cell products as a part of a strategy to fully eradicate HIV infectionsterilizing therapeutic for patients with HIV. Contributors: Lam, Sharon; Sung, Julia; Cruz, Conrad RY; Castillo, Paul; Ngo, Minh T; Rooney, Cliona M; Margolis, David M; Bollard, Catherine M 162 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF NFIA IN REMYELINATION Dylan James Laug Program in Developmental Biology Advisor: Benjamin Deneen, Ph.D.-Department of Neuroscience Oligodendrocytes are responsible for providing the myelin sheath that is essential for the rapid and efficient propagation of an action potential down the axon of a neuron. Understanding the processes that regulate the differentiation of oligodendrocytes and their myelination of neurons is important for determining possible treatments for demyelinating disorders, such as multiple sclerosis and cerebral palsy. Specifically, the progression of chronic multiple sclerosis is partly due to the failure of intrinsic mechanisms for the replacement of lost myelin and the leading cause of cerebral palsy is the loss of myelin during early post-natal due to white matter injury. Previously our lab demonstrated that NFIA is dynamically expressed in differentiating oligodendrocytes in the embryonic spinal cord, where it is co-expressed with Olig2+, oligodendrocyte precursors within the pMN domain of the embryonic spinal cord and downregulated as these precursor populations differentiate into myelinating oligodendrocytes. These observations suggest that NFIA functions to suppress oligodendrocyte precursor differentiation. Our lab demonstrated that NFIA suppresses oligodendrocyte differentiation during embryogenesis in mouse, chick, and in vitro oligodendrocyte precursor models. Also, our lab found that overexpression of NFIA in an adult mouse model of white matter injury and remyelination, suppressed oligodendrocyte precursor differentiation and remyelination. This, coupled with our observation that NFIA is expressed in oligodendrocyte precursors in human multiple sclerosis lesions and neonatal white matter injury, suggests that it may also play an important role in the suppression of differentiation and the failure of remyelination in these disorders. Late embryonic lethality of the NFIA knockout mice has limited our basic studies to embryonic stages. However, the clinical application of these findings is that reduced NFIA expression in oligodendrocyte precursors stimulates remyelination in cases of white matter injury and multiple sclerosis in post-natal or adult humans. Therefore, studies in the post-natal and adult mouse are crucial to assess whether the loss of NFIA stimulates remyelination following injury. Recently, we have generated a conditional, floxed NFIA allele that we have crossed with an oligodendrocyte precursor-specific cre to delete NFIA in this population to study its role in adult myelination and remyelination during white matter injury. In order to address the remyelinating capabilities of oligodendrocyte precursors, we will use lysolecithin injection in the spinal cord of oligodendrocyte precursor-specific NFIA knockout mice. Following lysolecithin-induced demyelination, we will analyze for myelin repair through both staining for mature oligodendrocyte markers, such as MAG and PLP, and observe the remyelination by electron microscopy. Contributors: Laug, Dylan; Glasgow, Stacey; Deneen, Benjamin 163 BAYLOR COLLEGE OF MEDICINE HIPPO AND WNT SIGNALING IN CARDIAC REGENERATION John Preston Leach Department of Molecular Physiology & Biophysics Advisor: James Martin, M.D./Ph.D.-Department of Molecular Physiology & Biophysics The leading cause of death in the United States is heart failure brought on by heart disease, and a loss of functional cardiac muscle. Because heart muscle regenerates poorly, loss of cardiomyocytes leads to a weakening of the heart culminating in heart failure. There is still paucity in understanding endogenous mechanisms preventing heart regeneration; our objective is to clarify the molecular pathways preventing meaningful cardiomyocyte renewal. The Hippo signaling pathway and its down stream effector Yap are known regulators of intrinsic organ size by modulating proliferation during development. Wnt/β catenin signaling has been well characterized in stem cells, and modulation of Wnt signaling like Hippo signaling presents an interesting potential for treating cardiac disease. Multiple studies have established a link between Hippo and Wnt signaling through activation and interaction of the down-stream transcription factors Yap and βcatenin. However the interaction of these two pathways during cardiac regeneration is poorly understood. Thus, the specific effect of Hippo and Wnt signaling activity in cardiomyocytes following myocardial infarction has yet to be determined. Thus, we hypothesize Hippo/Wnt signaling effectors Yap/β-catenin regulate cardiomyocyte regeneration. We have used two methods of cardiac damage in the mouse: Apex resection (AR) and LAD-ligation (MI). In a cardiomyocyte-specific inducible Salvador-knockout we demonstrated Hippo signaling inhibits cardiac regeneration. In he same mice adult cardiomyocytes re-enter the cell cycle as indicated by EDU assays and proliferation markers: pHH3, AurkB, and Ki67. Furthermore, we see functional recovery and reduced fibrosis in both the post-natal AR and MI models as well as the adult MI model. Others have now shown similar results looking at over-expression and GOF for Yap. Our current studies aim to clarify the role of beta-catenin during this Hippo knockout mediated cardiac regeneration. Thus far, our findings have uncovered Hippo signaling as an endogenous repressor of adult cardiomyocyte renewal and regeneration. Current experiments aim to evaluate Wnt signaling using both a β-catenin LOF and GOF approach. Contributors: Leach, John; Heallen, Todd; Tao, Ge; Morikawa, Yuka; Zhang, Min; Martin, James 164 2014 GRADUATE STUDENT SYMPOSIUM ANTI SCLEROSTIN ANTIBODY COULD BE A POTENTIAL TREATMENT OF OSTEOGENEISIS IMPERFECTA CAUSED BY WNT1 MUTATIONS Yi-Chien Lee Integrative Program in Molecular and Biomedical Sciences Advisor: Brendan Lee, M.D./Ph.D.-Department of Molecular & Human Genetics Osteogenesis Imperfecta (OI) is a brittle bone disease characterized by low bone mass, bone deformities, and multiple bone fractures. Our laboratory and other groups have found WNT1 mutations in OI patients. To establish a mouse model of OI caused by WNT1 mutations, we studied swaying mice that carry a spontaneous single base deletion in the Wnt1 gene. Interestingly, we found spontaneous bone fractures with severe osteopenia in swaying mice. We believe that the swaying mouse model could recapitulate the human phenotypes of OI caused by WNT1 mutations. Current treatment options for OI mostly focus on bisphosphonate therapy. Because of its questionable efficacy in mild OI patients and concerns about long-term administration, it is necessary to develop a new treatment. Studies have shown that sclerostin could directly inhibit WNT signaling through binding to LRP5/6, the co-receptor of WNT signaling. Therefore, utilizing an antibody targeting sclerostin to enhance WNT signaling and further increase bone formation becomes a potential therapeutic approach. We hypothesize that anti-sclerostin antibody could be beneficial for treating WNT1 related OI. In this study, we treat swaying mice and wild-type littermates with anti-sclerostin antibody twice per week for six weeks. At six weeks, we assess the bone phenotypes of mice. First, anti-sclerostin treated swaying mice exhibit reduced spontaneous fractures. From MicroCT imaging and analysis, we find swaying mice treated with anti-sclerostin antibody show significant increase of bone volume verses tissue volume in both femurs and spines. Moreover, by three point bending analysis on femur, anti-sclerostin antibody treated swaying mice show increased maximum load indicating the bone strength is increased. Finally, we evaluate the biomechanical properties of bone and find antisclerostin antibody treated swaying mice have increased collagen in bone. The results suggest that anti-sclerostin antibody treatment partially rescued the low bone mass phenotype in swaying mice. Anti-sclerostin antibody may be a potential treatment for OI caused by WNT1 mutations. Contributors: Joeng, Kyu Sang; Bi, Xiaohong; Ambrose, Catherine; Lee, Brendan H 165 BAYLOR COLLEGE OF MEDICINE THE ROLE OF RNF197 ON THE GROWTH OF BREAST CANCER Yu-Ju Lee Program in Translational Biology & Molecular Medicine Advisor: Weei-Chin Lin, M.D./Ph.D.-Department of Medicine Mothaffar Rimawi, M.D.-Department of Medicine Breast cancer is the most common cancer and the second leading cause of cancer death among American women. About 12% women in the US will develop invasive breast cancer during their lifetime. Although the early diagnosis and target therapy have significantly decreased the mortality rates of breast cancer, there is no effective treatment of ER, PR and HER2 triple-negative breast cancer (TNBC) and metastatic breast cancer. To develop a better treatment of breast cancer, we need to understand the molecular mechanisms of the initiation and progression of breast cancer. RNF197, also known as CGRRF1, is a p53-induced cell growth regulator with RING-finger domain. p53 is a tumor suppressor and approximately 20~35% of breast cancer patients have p53 mutations. Since RNF197 is implicated in the suppression of cell growth and is regulated by p53, it might be related to cancer development. Indeed, over-expression of RNF197 inhibits colony formation of colon carcinoma, ovarian carcinoma and glioblastoma cell lines. However, the role of RNF197 on breast cancer is still uncharacterized. We analyzed the correlation between the level of RNF197 and the survival rates of breast cancer, and found that the low expression of RNF197 is strongly correlated with a shorter survival. Compared with matched normal tissues, the level of RNF197 is lower in several tumor types including breast cancer. The analysis of the level of RNF197 in different subtypes of breast cancer showed a lower level of RNF197 in basal-like, mainly triple-negative, breast cancer. RNF197 contains a C3HC5 RINGfinger domain which is a binding motif of ubiquitin-conjugating enzymes. The activity of most E3 ubiquitin ligases is specified by the RING-finger domain. However, studies have not been done to examine the potential E3 ubiquitin ligase activity of RNF197. Based on this information, the central hypothesis of my research is that RNF197 regulates the growth of breast cancer. In particular, I hypothesize that RNF197 inhibits the growth of breast cancer through cell cycle regulation. To understand the mechanism behind RNF197-inhibited cell growth, I hypothesize that RNF197 is an E3 ubiquitin ligase and is able to degrade or regulate the biological function of its substrates associated with breast cancer cell proliferation. Studying the potential E3 ubiquitin ligase activity of RNF197, as well as its role in the development of breast cancer may help us to identify potential therapeutic targets or pathways that would aid in breast cancer treatment. Contributors: Lee, Yu-Ju; Lin, Weei-Chin 166 2014 GRADUATE STUDENT SYMPOSIUM KU PRIMARILY IMPACTS TELOMERE LENGTH IN SACCHAROMYCES CEREVISIAE VIA EST1 RECRUITMENT TO THE TELOMERE Laramie Denise Lemon Integrative Program in Molecular and Biomedical Sciences Advisor: Alison Bertuch, M.D./Ph.D.-Department of Pediatrics Telomeres are nucleoprotein complexes that cap the ends of linear eukaryotic chromosomes, protecting them from degradation, recombination and fusion, thus contributing to genome stability. Telomeres are maintained by telomerase which, in Saccharomyces cerevisiae, consists of an RNA component, TLC1, the reverse transcriptase Est2, and Est1 and Est3, which are required for telomerase activity in vivo, but not in vitro. Est1 recruits telomerase to the telomere during late S phase by interacting with Cdc13, a single-stranded telomeric DNA binding protein. The evolutionarily conserved Ku heterodimer also contributes to telomere length maintenance by associating with TLC1 at a unique stem loop structure. In contrast to strains lacking telomerase components, telomeres are short yet stable and cells do not senesce in the absence of Ku or Ku:TLC1 interaction. Strains lacking Ku or Ku:TLC1 binding, such as yku80-135i, fail to localize TLC1 to the nucleus and experience reduced levels of Est2 and Est1, possibly secondary to less Est2, at the telomere. Thus, the primary role of Ku in telomere elongation has yet to be fully elucidated. Our data support a model in which Ku’s major function in telomere length regulation is via effects on Est1. Unlike Est2, when we tethered Est1 to the telomere via Cdc13, Ku’s role in telomere elongation was bypassed, with telomeres progressively elongating in yku80∆ or yku80-135i strains comparable to wild type. Furthermore, we found Ku in a complex with Est1; this interaction was dependent on Ku’s ability to bind TLC1, as the interaction was abolished in a yku80-135i strain or upon treatment with RNase A. Although the presence of both Est1 and Est2 at the telomere is mutually dependent, we found tethering Est2 to the telomere via Cdc13 did not rescue Est1 levels at telomeres in a yku80∆ or yku80-135i strain, despite partial restoration of TLC1 in the nucleus. These data suggests that Ku’s primary role in telomere elongation lies in its interaction with Est1, and not with Est2 recruitment or TLC1 nuclear localization. The mechanism behind the role of Ku in Est1 recruitment to the telomere is currently under investigation. Although the Ku:TLC1 interaction is important for Est1 localization to the telomere, it remains unknown whether Ku binding to the DNA end is also required for Est1 association. The Est1:Cdc13 interaction at the telomere is promoted by Cdc13 phosphorylation at T308 and telomeres exhibit the same degree of shortness in yku80∆ cdc13-T308A and yku80∆ mutants. This suggests that Ku and the phosphorylation of Cdc13 at T308 function in the same pathway at the telomere. We are currently testing whether Ku DNA end binding activity modulates Cdc13 phosphorylation, thereby promoting Est1 association with the telomere. Contributors: Williams, Jaime; Ouenzar, Faissal; Chartrand, Pascal; Bertuch, Alison 167 BAYLOR COLLEGE OF MEDICINE AN ALTERNATIVE TRAFFICKING PATHWAY FOR NEWLY SYNTHESIZED PERFORIN Kelsey Elizabeth Lesteberg Department of Pathology & Immunology Advisor: George Makedonas, Ph.D.-Department of Pediatrics Background: One mechanism by which a cytotoxic T lymphocyte (CTL) kills its target relies on the exocytosis of specialized lysosomes called lytic granules, which contain the pore-forming protein perforin. Traditional dogma states that a CTL may only replenish its perforin content upon proliferation. However, it was demonstrated that CTL may rapidly upregulate perforin within hours of stimulation, and that it is targeted to the immunological synapse independently of lytic granules. In the current study, we aim to define the means by which this newly synthesized perforin traffics to the immunological synapse. Methodology: Antigen-specific CTL were generated by stimulating human leukocytes with peptides from CMV, EBV, or influenza viruses. After 10 days of expansion, the cells were re-stimulated with their cognate antigen for 2-6 hours. We then utilized imaging flow cytometry to determine the intracellular trafficking mediators involved in the transport of newly synthesized perforin. The cells were stained with antibodies against CD8 and CD56, a panel of rab and SNARE proteins, and antiperforin clones D48 and δG9—D48 recognizes both new perforin and lytic granuleassociated perforin, whereas δG9 recognizes only granule-associated perforin. The localization of both perforin types with rab and SNARE proteins was assessed through the use of bright detail similarity (BDS) analysis, which assigns a score to each cell based on the overlap of the probes of interest. Results: We defined cells displaying significant colocalization between perforin and trafficking molecules as those having BDS scores greater than 1.5. Newly synthesized perforin (D48+ δG9-) localized to recycling endosome compartments, as identified by rab8, vti1b, and the transferrin receptor (CD71). In addition, new perforin also colocalized with rab37 and VAMP4, which are involved in transport to the synapse and to endosomes, respectively. Conclusions: Our data suggest that newly synthesized perforin is incorporated into recycling endosomes, where it may be sorted to subsequent destinations: the immunological synapse for immediate use, or to lysosomes to refill the cells’ store of lytic granules. These results define an alternative intracellular trafficking pathway for perforin that functions to sustain CTL cytotoxicity. Future work will investigate the determinants of perforin sorting through this alternative pathway. Contributors: Lesteberg, Kelsey; Makedonas, George 168 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF FXR-GANKYRIN-PROTEASOME PATHWAY IN LIVER CANCER, PROLIFERATION AND LIVER INJURY Kyle Joseph Edward Lewis Department of Molecular & Cellular Biology Advisor: Nikolaj Timchenko, Ph.D.-Department of Pathology & Immunology Richard Sifers, Ph.D.-Department of Pathology & Immunology Biological processes in the liver are regulated by a complex cooperation of multiple signaling pathways. Disorganization of this cooperation is associated with development of liver diseases such as liver cancer and non-alcoholic fatty liver disease. We have previously found that alterations in the ubiquitin-proteasome system are involved in development of liver cancer. A small subunit of the 26S proteasome, Gankyrin (Gank), is activated during development of liver cancer and eliminates tumor suppressor proteins Rb, p53, C/EBPα, and HNF4α through UPS-mediated degradation. Quiescent livers express relatively low levels of Gank due to repression by Farnesoid X Receptor (FXR) signaling. We found that long-lived, growth hormone-deficient mice have expressed high levels of FXR and do not develop liver cancer with age and under conditions of Diethylnitrosamine (DEN)-mediated carcinogenesis. Because these mice do not increase levels of Gank, this led us to the hypothesis that Gank plays a key role in liver proliferation and liver cancer. To further investigate the role of Gank in liver biology, we have generated mice with liver-specific deletion of Gank, Gank LKO mice. We first examined the role of Gank in liver regeneration after partial hepatectomy (PH). These studies showed that the deletion of Gank leads to a dramatic reduction of liver proliferation which is associated with much lower and delayed DNA replication and almost complete inhibition of mitosis. We found that a powerful activator of liver proliferation, cyclin D1, is identically elevated in both WT and Gank LKO mice; however, this elevation is not sufficient to initiate DNA replication in Gank LKO livers. In addition, we found that expression of PCNA and cdc2 is significantly reduced in livers of Gank LKO mice after PH. Examination of liver injury and recovery after carbon tetrachloride (CCl4) treatments also showed that deletion of Gank changes liver response to CCl4. Taken together, our results showed that the FXR-gankyrin pathway plays an essential role in liver cancer and in liver proliferation after surgical resections and injury. Contributors: Lewis, Kyle; Jiang, Yanjun; Moore, David; Timchenko, Nikolai 169 BAYLOR COLLEGE OF MEDICINE HIV VLPs CONJUGATED TO LIPID VESICLES CONTAINING MPLA, A TLR4AGONIST, INDUCES A POTENT AND ENDURING HIV-SPECIFIC IMMUNE RESPONSE Phoebe Elnora Lewis Program in Translational Biology & Molecular Medicine Advisor: Qizhi Yao, M.D./Ph.D.-Department of Surgery Shital Patel, M.D.-Department of Medicine Human Immunodeficiency Virus (HIV) continues to be a major public health concern with almost 1.2 million people currently infected with the virus. Despite 3 phase III clinical trials, an effective prophylactic HIV vaccine candidate has yet to be identified. Results from previous clinical trials have revealed major safety concerns involving the use of viral delivery vectors and their potential to increase risk of HIV infection. VirusLike-Particles (VLPs) offer a safe alternative for viral-vector based strategies as this platform does not require the use of viral genetic material that may have the potential to increase the risk for HIV infection. The translatable potential of VLP based vaccines have been demonstrated with the FDA approved Gardasil ™ vaccine against HPV. VLPs are structurally similar to native virions and have the capability of eliciting both cellular and humoral immune responses. Our lab has successfully engineered chimeric VLPs containing combinations of SIV or HIV gag, various strains of HIV env, and immuno-modulatory molecules such as CD40 ligand (CD40L) or influenza hemagglutinin (HA). Adjuvants provide an additional platform to further increase the immunogenicity of our chimeric VLPs. In this study, we evaluated the potency and longevity of HIV-specific immune responses induced by HIV GagIIIB/EnvBaL VLPs conjugated to Conjugatable Adjuvant Lipid Vesicles (CALVs) containing a Toll-like-receptor 4 (TLR4) agonist in a C57B6 mouse model. We demonstrate that the TLR4-agonist potently enhanced HIV-1IIIB Gag, HIV1BaL Env , and Gag/Env-VLP-specific total IgG responses up to 10-fold after intranasal priming followed by three intracheek boosts with VLPs +/- TLR4 agonists. Furthermore, our TLR4-agonist conjugated VLPs induced a predominant IgG1 subtype response with levels 4-fold higher than specific IgG2a responses. Intracellular Cytokine Staining demonstrated that Env and Gag peptide stimulation induced a potent IL-2 response, the primary cytokine involved in immunological memory formation. Mice immunized with VLPs + TLR4-agonist had 3 fold higher IL-2 producing CD8+ cells than the VLP only immunization group upon Env- peptide stimulation. Preliminary data also suggests that TLR-4 agonist conjugated to VLPs can elicit persistent long-term antibody responses as high levels of HIV-specific IgG1 were detected 90 days after the last immunization. Our results indicate that TLR-4 agonists conjugated with our VLPs induce potent and enduring IgG responses with a predominant IgG1 subtype. Contributors: Lewis, Phoebe; Poteet, Ethan; Ho, Sam; Fujii, Gary; Chen, Changyi;Yao, Qizhi 170 2014 GRADUATE STUDENT SYMPOSIUM SUPPRESSORS OF THE TGRC1– PHENOTYPE: SIGNAL TRANSDUCTION AND EVOLVABILITY Cheng-Lin Li Department of Molecular & Human Genetics Advisor: Gad Shaulsky, Ph.D.-Department of Molecular & Human Genetics The cell-cell adhesion molecules, TgrB1 and TgrC1, mediate allorecognition and cell differentiation during development in Dictyostelium. TgrB1 and TgrC1 are essential for proper development as single-gene deletions (tgrB1-null or tgrC1-null) or singlegene replacement (e.g., replacing tgrC1AX4 with tgrC1QS45) lead to a developmental arrest at the loose aggregation stage and lack of cell-type differentiation. Moreover, TgrB1 and TgrC1 function as a receptor-ligand pair for kin recognition and are highly polymorphic in natural populations. Cells carrying diverged allelic pair of tgrB1-tgrC1 segregate from one another during streaming. The TgrB1-TgrC1 system presents a novel model to study the molecular basis of rapid diversification of adhesion receptors. It is intriguing how adhesion receptors can evolve rapidly despite the fact that they have essential functions in development. To elucidate the evolvability of this system, we are dissecting the signal transduction pathways that mediate TgrB1-TgrC1 signaling. We have devised screens for genetic suppressors that rescue the developmental defects in tgrC1-null cells. We discovered mutations in several candidate genes, including a glycosyltransferase (alg9) and stcA (suppressor of tgrC1–), that suppress some of the tgrC1-null phenotypes. Inhibition of N-linked glycosylation by insertional mutation in alg9 or by Tunicamycin treatment allow tgrC1-null cells and the single gene replacement strain (tgrB1AX4 tgrC1QS45) to sporulate (~10% sporulation efficiency). The mutation stcAins increases the sporulation efficiency of tgrC1-null to 7% through an unknown mechanism. Co-development of the suppressor mutants and the parental tgrC1-null cells rescued the development of the latter, suggesting that suppression acts through a non-cell autonomous mechanism. The essential function of TgrB1 and TgrC1 in development and the selection for maintaining the compatibility between these two proteins restrict the molecular evolution of the tgrB1-tgrC1 genes. However, genetic suppressors, such as alg9 and stcA, may play a role in buffering the effects of novel tgrB1-tgrC1 mutations and allow the organism to tolerate the associated fitness disadvantages. Therefore, these mechanisms may promote allelic diversification and expand the allelic repertoires of tgrB1-tgrC1. Contributors: Li, Cheng-Lin; Wang, Yue; Kuspa, Adam and Shaulsky, Gad 171 BAYLOR COLLEGE OF MEDICINE PITX2 IS ASSOCIATED WITH CARDIAC ARRHYTHMIA AND CARDIAC DEVELOPMENT Lele Li Program in Cardiovascular Sciences Advisor: James Martin, M.D./Ph.D.-Department of Molecular Physiology & Biophysics Pitx2 is localized on chromosome 4q25, variants of which are associated with increased risk of atrial fibrillation. This locus locates close to the pituitary homeobox2 (Pitx2) gene. Pitx2 is a transcription factor, which plays a critical role in left-right asymmetry establishment and maintenance of the heart. Our previous studies indicate that Pitx2+/− mice show symptoms of atrial fibrillation when given programmed electrical stimulation. However, role of Pitx2 in heart function is still not clear. Therefore, we hypothesize that Pitx2 deficiency leads to cardiac arrhythmia and ARVC generation. Conditional knockout mouse Pitx2flox/flox MCK-Cre mice, in which Pitx2 expression is specifically disrupted in mice cardiac/skeletal muscle after birth showed sinus node dysfunction. ChIP-sequencing and microarray assay is done to find target genes of Pitx2 and determine changes in genes expression levels. Finally, we find RNA level of genes including genes related to ion channels, signaling pathways, transcription factors is more than two fold in mutant than in wildtype mice. Majority of the genes screened have an increase in RNA level in mutant mice, indicating Pitx2 may function mainly as a repressor for the candidate genes. RNA level of a number of candidate genes, which regulates cardiac function, are analyzed using qRT-PCR. Result indicated upregulation of RNA level of majority of candidate genes and down regulation of a small number of genes, indicating Pitx2 may function mainly as repressor in regulation of genes transcription in cardiac function. Luciferase reporter assay was conducted and confirmed Pitx2 targets. TEM showed that Pitx2 CKO mice have damaged mitochondria and disrupted intercalated disc structure, suggesting Pitx2 regulates heart function by stabilize integrity of intercalated disc structure. Pitx2 F/+Nkx2.5-Cre adult mice showed phenotype of AV block in telemetry, indicating the defect of Pitx2 is associated with cardiac conduction system. In situ hybridization and immunostaining showed PItx2 F/+ Nkx2.5-Cre mice have abnormal expression of cardiac conduction system markers, including HCN4 and Tbx3. ChIP-seq and microarray overlay screened a set of genes related to mitochondria. Confirmation by qPCR showed increased level of these genes in mutant mice. Contributors: Tao, Ye; Tao, Ge; Zhang, Min; Martin, James 172 2014 GRADUATE STUDENT SYMPOSIUM THE E3 UBIQUITIN LIGASE UBR3 REGULATES USHER SYNDROME TYPE 1 PROTEINS IN DROSOPHILA AND ZEBRAFISH AUDITORY ORGANS Tongchao Li Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics Andrew Groves, Ph.D.-Department of Neuroscience The auditory organs of Drosophila and vertebrates have a number of molecular and functional similarities despite being widely separated in evolutionary time. To identify novel genes that regulate the development and function of auditory organs, we performed a mosaic screen of X-chromosome linked lethal mutations in Johnston’s organ, the auditory organ of Drosophila. We identified mutations in ubr3, an E3 ubiquitin ligase, that cause a detachment of the sensory scolopidia in Johnston’s organ. This phenotype is similar to that seen in mutations in Drosophila myosin7a. Here, we show that ubr3 genetically interacts with myo7a and that Ubr3 regulates Myo7a monoubiquitination via Cullin 1 (Cul1), another E3 ligase. Myosin7a mutants in humans are associated with Usher Syndrome 1B, and we show that Ubr3, Cul1 and Myo7a physically and genetically interact with Drosophila homologues of two other Usher syndrome proteins, PCDH15 and Sans. Finally, we show loss of Ubr3 in zebrafish leads to the mislocalization of three Usher syndrome proteins, Myo7a, Harmonin and PCDH15, and causes hair-bundle and hearing defects. Our results demonstrate the conservation of Usher syndrome protein interactions between vertebrates and invertebrates in the assembly and function of mechanosensitive organs and identify a novel pathway in which a cascade of E3 ligases regulates protein abundance through ubiquitination. Contributors: Tongchao Li, Bernardo Blanco, Nikolaos Giagtzoglou, Shinya Yamamoto, Manish Jaiswal, Sonal N. Jaiswal, Daniel F. Eberl, Dorothea Godt, Monte Westerfield, Andrew K. Groves, Hugo J. Bellen 173 BAYLOR COLLEGE OF MEDICINE NECROTIC CELLS ACTIVELY ATTRACT PHAGOCYTES THROUGH THE COLLABORATIVE ACTION OF TWO DISTINCT PS-EXPOSURE MECHANISMS Zao Li Department of Biochemistry & Molecular Biology Advisor: Zheng Zhou, Ph.D.-Department of Biochemistry & Molecular Biology Cells injured pathologically undergo necrosis, a type of cell death distinct from apoptosis in both morphology and mechanism. Like apoptotic cells, necrotic cells must be swiftly removed from animal bodies to prevent harmful inflammatory and autoimmune responses. In the nematode Caenorhabditis elegans, gain-of-function mutations in certain ion channel subunits result in necrotic-like cell death of six touch neurons. Necrotic touch neurons are engulfed and degraded inside engulfing cells. It is unclear how necrotic cells are recognized by engulfing cells. Phosphatidylserine (PS) is an important apoptotic cell surface signal that attracts engulfing cells. Using ectopically expressed MFG-E8, a high-affinity PS-binding protein, we observed that PS was present on the surface of necrotic touch neurons. In addition, phagocytic receptor CED1, whose function is needed for the efficient clearance of necrotic cells, clusters around necrotic cells. The extracellular domain of CED-1 associates with PS as determined in assays for an enzyme-linked immunosorbent assay-like solid-phase reaction and surface plasmon resonance. We further identified a necrotic cell-specific function of CED-7, the worm homolog of mouse ABC1 transporter, in promoting PS-exposure on necrotic cell surfaces. In addition to CED-7, ANOH-1, the C. elegans homolog of mammalian Ca2+-dependent phospholipid scramblase TMEM16F, plays an independent and unique role in promoting PS-exposure on the surface of necrotic but not apoptotic cells. The combined activities from CED-7 and ANOH-1 ensure sufficient PS exposure for necrotic cells to attract their phagocytes. Our work demonstrates that cells killed by different mechanisms (necrosis or apoptosis) expose a common “eat me” signal that attracts their common phagocytic receptor CED-1. Furthermore, unlike previously believed, we discovered that PS is actively exposed onto the outer surface of necrotic cells through two distinct molecular mechanisms, rather than being leaked out passively. Contributors: Li, Zao; Venegas, Victor; Raghavan, Prashant; Nakanishi, Yoshinobu; Zhou, Zheng 174 2014 GRADUATE STUDENT SYMPOSIUM NEURON-GLIA INTERACTION VIA NFΚB/COMPLEMENT SIGNALING MEDIATES DENDRITIC MORPHOLOGY AND NEURONAL EXCITABILITY: DYSREGULATION IN ALZHEIMER’S DISEASE Hong Lian Department of Molecular & Human Genetics Advisor: Hui Zheng, Ph.D.-Department of Molecular & Human Genetics Abnormal NFκB activation has been implicated in Alzheimer’s disease (AD). However, the signaling pathways governing the regulation and function of NFκB in the brain are poorly understood. Here we demonstrate that NFκB activity is critically controlled by its inhibitor protein IκBα in astrocytes, but not in neurons. We identify complement protein C3 as an astroglial target of NFκB whose levels are increased in response to heightened NFκB activity. Elevated astroglial C3, in a neuronal complement receptor C3aR dependent manner, triggers aberrant intraneuronal calcium levels and disrupts surface AMPA receptor-mediated excitatory synaptic function and dendritic morphology. These effects can be rescued by C3aR blockade. Importantly, we show that astroglial NFκB and C3 can be induced by A and are upregulated in AD brains. Thus, deregulation of neuron-glia interaction through IκBα/NFκB/C3/C3aR/calcium signaling may contribute to synaptic dysfunction occurring in AD and our study provides support that C3aR antagonists may be therapeutically beneficial. Contributors: Li Yang, David Shim, Allysa Cole, Jennifer Rodriguez-Rivera, Giulio Taglialatela, Hui-Chen Lu and Hui Zheng 175 BAYLOR COLLEGE OF MEDICINE THE ROLE OF MULTI-ENZYME AMINOACYL TRNA SYNTHETASE COMPLEX COMPONENT AIMP1/P43 IN TH1 IMMUNITY Dan Liang Department of Pathology & Immunology Advisor: William Decker, Ph.D.-Department of Pathology & Immunology Of professional antigen presenting cells (APC), only the dendritic cells (DC) are regarded as initiators of adaptive immune responses. Previously we identified a Th1promoting phenotype of DC simultaneously loaded with overlapping (homologous) MHC class I and II determinants: tumor specific mRNA and cell lysate. This event, termed “homologous antigenic loading”, augmented DC IL-12 production, generation of CD8+ cytolytic effectors, and IFN-γ T-cell secretion, all downstream hallmarks of Th1 immunity. We further confirmed that this Th1-promoting phenotype exists across a variety of antigenic systems in which DC class I and II are loaded with determinants of homologous origin, indicating DC intrinsic mechanisms to recognize and compare antigenic epitopes. In an effort to characterize underlying mechanisms, we looked for secreted factors in DC culture supernatants and detected high levels of AIMp1/p43 release from DC loaded with homologous lysate/mRNA, single protein antigen, or overlapping viral peptides in comparison to singly-loaded or heterologous controls. AIMp1 is a structural component of the multi-enzyme aminoacyl-tRNA synthetase complex (mARS) which consists of at least 8 aminoacyl-tRNA synthetases and 3 structural proteins. Previous studies have shown that AIMp1 is released by both tumor and macrophages under stress or TLR stimulus, upregulates proinflammatory gene expression in monocytes and macrophages, and induces Th1 polarization of bone marrow derived DC (BMDC). We subsequently established that the level of AIMp1 release is closely correlated to the degree of homology between loaded class I and II epitopes. In vitro studies confirmed that siRNA knockdown and genetic knockout reduced the expression of costimulatory markers and Th1 cytokines from BMDC. Further, AIMp1 deficiency abolished the ability of DC vaccines to mediate Th1 immune response against B16 tumors in vivo. These data suggest secreted AIMp1 is critical for Th1 immune responses induced by homologous loading of DC. AIMp1 also colocalizes with MHC class I and II molecules as determined by coIP and mass spectrometry, suggesting association with antigen recognition machinery. In future studies, we will determine molecular and cellular mechanisms of AIMp1-mediated crosstalk between DC and T-cells. We are looking for candidate cell surface receptors and downstream signal pathways both in DCs and T cells. Additionally, we will determine how AIMp1 release by DC is regulated and the role of other mARS components in this process. Finally, we seek to apply AIMp1 as a novel vaccination adjuvant and will test its potential in anti-tumor immunity and infectious diseases models. Most critically, these studies will underscore the importance of homologous antigenic signals in DC polarization.Contributors: Liang, Dan; Halpert, Matthew M; Konduri, Vanaja; Levitt, Jonathan M; Decker, William K 176 2014 GRADUATE STUDENT SYMPOSIUM THE EFFECT OF BLAST-WAVE EXPOSURE ON THE MOUSE VESITUBLOOCULAR REFLEX Steven Donald Lien Department of Neuroscience Advisor: J. Dickman, Ph.D.-Department of Neuroscience The dramatic escalation of conflict has led to higher incidences of exposure to blast-related traumatic brain injury. Active service members who have been deployed to conflict zones have reported high rates of vestibular dysfunction, such as vertigo, imbalance, and dizziness, accumulating evidence suggests that blast wave trauma causes damage to both the peripheral and central vestibular system. However, the mechanism by which blast-wave exposure induces vestibular dysfunction remains unclear. Previous work has established that blast-wave exposure is capable of causing damage to the auditory hair cells of the inner ear. Our goal for this study is to identify whether blast-wave exposure will damage the vestibular hair cells of the inner ear. To determine whether blast-wave exposure causes damage to the peripheral vestibular system, we plan on analyzing the horizontal vestibulo-ocular reflex (hVOR) and vertical vestibulo-ocular reflex (vVOR) after exposure to a mild blast-wave. Analyzing the hVOR and vVOR may give us further insight into the mechanism of vestibular dysfunction after blast-related traumatic brain injury. Contributors: Lien, Steven; Dickman, David 177 BAYLOR COLLEGE OF MEDICINE OPTIMIZATION OF HSC TRANSFECTION CONDITION Angelique Lin Integrative Program in Molecular and Biomedical Sciences Advisor: Daisuke Nakada, B.A.Sc.-Department of Molecular & Human Genetics Hematopoietic stem cells (HSCs) are the stem cells found in bone marrow that generate all the functional hematopoietic cells. The ability of HSC to self-renew and maintain multipotency is critical to sustain life and thus the identification and study of genes that regulate HSC function are important for HSC research. The recently developed CRISPR/Cas9 system is a powerful gene-editing tool and has been widely used to study gene regulation. Delivering Cas9 mRNA and sgRNA targeting gene-ofinterest into HSCs would provide a fast and precise system to screen for genes that are critical for HSC function. However, the transfection of HSCs is challenged with low efficiency. For this purpose, our lab has optimized the transfection condition for HSCs using the Neon transfection system. As an initial optimization step, we used c-kit positive cells isolated from the bone marrow of ROSA26-LSL-eYFP mouse and transfected Cre mRNA to induce eYFP expression as reporter for transfection efficiency. C-kit positive cells were separated using an autoMACS. Prior to electroporation, c-kit positive cells were cultured for different time periods. The cultured cells were then mixed with different amount of Cre mRNA and transfected by electroporation with Neon transfection system. Transfected cells were cultured overnight and the transfection efficiency was analyzed by flow cytometry for YFP expression. We found that different culture condition time and the amount of mRNA affected transfection efficiency. The transfection efficiency of the bulk population was about 20% for cells cultured for 1 hr prior to electroporation with 1 ug of mRNA. Notably, the efficiency was about 50% for lineage-negative population and about 60% for the LSK (lineage-negative, Sca-1+, ckit+) population. Electroporation of HSCs with Neon transfection system is an effective way to transfect these cells with mRNA directly to study gene function in HSCs. Based on these results, CRISPR/Cas9 system might be used for screening genes important for HSC function by delivering Cas9-mRNA and sgRNA into HSCs. Contributors: Nakada, Daisuke; Kitano, Ayumi; Takeichi, Makiko; Saitoh, Yusuke; Chapple, Richard Harrison 178 2014 GRADUATE STUDENT SYMPOSIUM LIPID METABOLIC REGULATION UPON ENVIRONMENTAL NUTRIENT ANTICIPATION Chih-Chun Lin Department of Molecular & Human Genetics Advisor: Meng Wang, Ph.D.-Department of Molecular & Human Genetics The abilities of environmental sensation and adaption determine the fitness of organisms in the constantly changing world. Whether and how animals anticipate environmental changes remain largely unknown. Among many environmental variables, food availability is the most critical one. In our studies, we utilize Caenorhabditis elegans as a model organism to understand how organisms anticipate nutritional restriction in the environment through perceiving dietary metabolite signals from their preys, and adjust their metabolic strategies for a better survival under food scarcity. As soil-dwelling nematodes, C. elegans experience their diet of bacteria as both a source of nutrition and a source of sensory input. We found that C. elegans fed with bacteria that are grown in a nutrient deprived condition dramatically increases fat storage, and these pre-starved C. elegans survives better upon actual starvation. This presents the first evidence that organisms are able to anticipate environmental changes from interacting with their preys. We further demonstrated that this nutrient anticipation response is mediated by an orphan nuclear hormone receptor, NHR-25, homolog of mammalian LRH-1. We showed that NHR-25 regulates fatty acid de novo synthesis pathway (fat-5, fat-7) and triacylglyceride synthesis pathway (dgat-2), which together lead to increased fat storage. Our data also suggest that oleic acids and phospholipids are the upstream signaling molecules that regulate NHR-25 activity. This integrative gene-environment interaction study unveils a novel mechanism by which organisms sense future environmental starvation from current dietary cues and subsequently remodel their energy metabolic strategy to enable better fitness in the time ahead. Through further dissecting this new molecular mechanism, we expect to identify beneficial probiotic supplements that boost metabolic health in human. Contributors: Wang, Meng 179 BAYLOR COLLEGE OF MEDICINE SEX-SPECIFIC DIFFERENCES IN HYPEROXIC LUNG INJURY IN MICE: ROLE OF CYTOCHROMEP450(CYP) 1A Krithika Lingappan Clinical Scientist Training Program Advisor: Bhagavatula Moorthy, Ph.D.-Department of Pediatrics Background: Sex-specific differences in pulmonary morbidity in adults and preterm infants are well documented. Hyperoxia contributes to lung injury in experimental animals and humans. Cytochrome P450(CYP)1A enzymes have been shown to play a mechanistic role in hyperoxic lung injury (HLI) in animal models. Whether CYP1A enzymes contribute to sex-specific differences in relation to HLI is unknown. In this investigation, we tested the hypothesis that mice will display sexspecific differences in HLI, and that this phenomenon will be altered in mice lacking the genes for Cyp1a1 or 1a2. Methods: Eight week-old male and female wild type (WT) (C57BL/6J) mice, Cyp1a1-/-, and Cyp1a2-/- mice were exposed to 72 hours of hyperoxia (FiO2>0.95). Lung injury and inflammation was assessed and pulmonary and hepatic CYP1A1 and CYP1A2 levels were quantified at the enzyme activity, protein and mRNA level. Results: Upon exposure to hyperoxia, WT males showed greater lung injury and neutrophil infiltration. Analysis of liver and lung microsomal proteins showed higher pulmonary CYP1A1 (apoprotein level and activity) in WT females compared to WT males and a greater induction in hepatic CYP1A2 mRNA levels and activity in WT females after hyperoxia exposure. The sex based female advantage was lost or reversed in Cyp1a1-/- and Cyp1a2-/- mice. Conclusion: This suggests an important role for CYP1A enzymes in the sexspecific modulation of hyperoxic lung injury Contributors: Weiwu, Jiang; Wang, Lihua; Couroucli, Xanthi; Moorthy, Bhagavatula 180 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF COMPLEMENT IN DEVELOPMENT OF TAU-RELATED PATHOLOGY IN ALZHEIMER`S DISEASE Alexandra Litvinchuk Integrative Program in Molecular and Biomedical Sciences Advisor: Hui Zheng, Ph.D.-Department of Molecular & Human Genetics Alzheimer`s disease (AD) is the most common form of dementia that affects millions of people worldwide. The pathological changes of AD include the formation of amyloid plaques of Aβ and neurofibrillary tangles (NFT) of hyperphosphorylated Tau. However, the functional link between Aβ and NFTs is poorly understood. A common feature of AD is the activation of the immune response in the brain. The activation of complement pathway has been implicated in this process. In particular, complement component C3 inhibitor sCrry has been reported to modulate both Aβ and NFT pathologies in mouse models. Moreover, reduced levels of phospho-Tau were shown in a Taurelated model of AD, P301L/sCrry double-transgenic, suggesting the beneficial role of complement inhibition in AD. Ongoing studies in our lab have shown that NF-kB can activate complement C3 component in astrocytes but not neurons. Moreover, elevated astroglial C3 through C3aR in neurons can affect intraneuronal calcium levels, and dysregulation of calcium homeostasis has been implicated in the pathogenesis of AD. Importantly, astroglial NF-kB and C3 can be induced by Aβ and are upregulated in AD brains. These data raise the intriguing possibility that astroglial C3-mediated neuroinflammatory pathway may serve as a functional link between Aβ and NFT pathologies in AD. Therefore, we hypothesize that Aβ-mediated astroglial activation of complement induces Tau phosphorylation in neurons via C3aR and this in turn leads to the formation of neurofibrillary tangles and subsequent neurodegeneration. We further hypothesize that inhibition of the C3/C3aR pathway will be therapeutically beneficial. Aim1. To investigate the role of C3 and C3aR receptor on formation of NFTs and Tau phosphorylation in vitro and vivo. Soluble C3 will be administered to primary neurons from wild-type or PS19 mice, that overexpresses human mutant Tau. Complement inhibition will be achieved by treatment of cells with C3aR antagonist. For in vivo experiments PS19 and C3aR KO/PS19 mouse lines will be generated. Levels of phospho-Tau will be quantified by western blot and immunohistochemistry with total Tau, C3, AT8, PHF1 and MC1 antibodies. NFT formation will be confirmed with ThT staining. Behavioral and electrophysiological studies will be performed to characterize the phenotype of C3aR KO/P301S mice. Aim 2. To identify downstream effectors of C3 and C3aR that promote Tau phosphorylation. The activation of potential kinases that are responsible for increased Tau phosphorylation in complement C3-dependent model will be tested by western blot, immunohistochemistry and ELISA. Validation of potential targets will be achieved by treatment of primary neurons with specific kinase inhibitors or siRNA. Aim 3. To establish the functional link between Aβ and Tau in development of Taurelated pathology in AD through C3 and C3aR. Primary astroglia from wild type mice will be co-cultured with neurons from wild-type, C3aR KO, PS19 and C3aR KO/PS19 mice. Activation of complement will be achieved by administering Aβ oligomers and fibrils to astrocytes prior to starting the co-cultures. In in vivo experiments Aβ microinjections in the hippocampal area of 5-months old PS19 and C3aR KO/PS19 mice will be performed. Levels of total Tau, phospho-Tau, and C3 will be quantified by western blot, immunohistochemistry and ELISA. 181 BAYLOR COLLEGE OF MEDICINE MOLECULAR MECHANISM AND EVOLUTION OF GUANYLATE KINASE REGULATION BY (P)PPGPP Kuanqing Liu Department of Molecular Virology & Microbiology Advisor: Jue Wang, Ph.D.-Department of Molecular & Human Genetics Anthony Maresso, Ph.D.-Department of Molecular Virology & Microbiology The nucleotide (p)ppGpp is a ubiquitous stress resistance mediator in bacteria, but its targets and underlying mechanisms of action vary among bacterial species and remain incompletely understood. Here we characterize the molecular interaction between (p)ppGpp and guanylate kinase (GMK) and reveal its remarkable conservation across many species and its importance in adaptation to starvation. Combining structural and kinetic analyses, we show that (p)ppGpp binds the GMK active site and inhibits the enzyme by a competitive mechanism. Abolishing the (p)ppGpp-GMK interaction results in defective adaptation to amino acid starvation, highlighting its physiological importance. A survey of GMKs from phylogenetically diverse bacteria shows that the (p)ppGpp-GMK interaction is conserved in members of Firmicutes, Actinobacteria, Deinococcus-Thermus, but not in Proteobacteria where (p)ppGpp regulates RNA polymerase (RNAP). We propose that GMK is an ancestral (p)ppGpp target and RNAP evolved more recently as a direct target in Proteobacteria. Contributors: Liu, Kuanqing; Myers, Angela; Claas, Kathy; Satyshur, Kenneth; Keck, James; Wang, Jue 182 2014 GRADUATE STUDENT SYMPOSIUM NEURONAL MITOCHONDRIAL DEFECTS ELEVATE ROS, JNK, SREBP AND SYNTHESIS OF LIPID DROPLETS - PROMOTING NEURODEGENERATION Lucy Liu Department of Neuroscience Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics An increase in lipid droplets (LD) has been implicated in some metabolic disorders but their role in neurodegeneration is ill defined. We show that several genes that affect mitochondrial function lead to an accumulation of LD in glia prior to or at the onset of neurodegeneration in Drosophila. This LD accumulation is caused by increased reactive oxygen species (ROS), which promotes c-Jun-N-terminal Kinase (JNK) and Sterol Regulatory Element Binding Protein (SREBP) activity, and neuronal activation of this pathway is sufficient to cause glial LD accumulation. LD accumulation can be reduced by lowering ROS, JNK, or SREBP levels, or by overexpressing lipases. These manipulations significantly delay the onset of neurodegeneration. Furthermore, a similar pathway leads to glial LD accumulation in Ndufs4 mutant mice, suggesting that LD accumulation following mitochondrial dysfunction is an evolutionarily conserved phenomenon. In summary, our studies show that increased ROS leads to LD accumulation in glia, and that preventing LD accumulation delays neurodegeneration. Contributors: Lucy Liu, Ke Zhang, Hector Sandoval, Shinya Yamamoto, Manish Jaiswal, Elisenda Sanz, Zhihong Li, Brett H. Graham, Albert Quintana and Hugo J. Bellen 183 BAYLOR COLLEGE OF MEDICINE OSTEOCLAST-DEPENDENT EFFECTS OF HYPOXIA ON BREAST CANCER BONE METASTASIS PROGRESSION Flora Lo Integrative Program in Molecular and Biomedical Sciences Advisor: Xiang Zhang, Ph.D.-Department of Molecular & Cellular Biology 20-40% of breast cancer patients eventually develop metastasis to the bone. When presented clinically, breast cancer bone metastases are predominantly osteolytic. Cancer cells secrete factors that activate osteoclasts (bone-breaking cells), and the resulting degradation of bone matrix in turn releases factors that promote cancer cell growth. This forms a vicious cycle that drives aggressive metastatic outgrowth. Interestingly, in early stage metastasis, disseminated cancer cells likely first remain as dormant micrometastases in the bone, and later re-activate and progress to overt lesions. However, very little is known about the regulation of metastasis dormancy and progression. Recent work in our lab suggested that bone metastasis initially reside in a microenvironment with features of osteogenesis, implying it later switches to an osteolytic microenvironment by an unknown mechanism. We are investigating the hypothesis that hypoxia serves as a mechanistic switch for micrometastases to progress to overt metastases via the vicious osteolytic cycle. Why hypoxia? To fuel the vicious cycle, cancer cells that have metastasized to the bone secrete osteolytic factors, such as parathyroid hormone-related protein (PTHrP) and connective tissue growth factor (CTGF). Evidence suggests that the expression of these factors is modulated by hypoxia response proteins in various cell types, but this has not been studied extensively in breast cancer cells. Moreover, our lab discovered that early metastases are found in osteogenic microenvironment niches. These are associated with hypoxic regions in the bone, supporting the role of hypoxia in metastasis progression. It is possible that as the micrometastasis slowly expands, the microenvironment becomes even more hypoxic and promotes the induction of the osteolytic cycle. Using a range of in vitro and in vivo techniques, I aim to identify osteolytic factors that are up-regulated in expression under hypoxic conditions. I will also investigate the effects of hypoxia-mediated changes in osteolytic factor secretion on the bone microenvironment, and cancer cell dormancy and growth. Contributors: Yu, Cuijuan; Wang, Hai; Muscarella, Aaron; Zhang, Xiang 184 2014 GRADUATE STUDENT SYMPOSIUM SPDEF INHIBITS COLORECTAL CANCER CELL GROWTH BY DISPLACING ΒCATENIN FROM CHROMATIN Yuan-Hung Lo Integrative Program in Molecular and Biomedical Sciences Advisor: Noah Shroyer, Ph.D.-Department of Pediatrics Colorectal cancer (CRC) is one of the most lethal cancers worldwide. Under physiological conditions, the Notch and Wingless/Int (Wnt) signaling pathways modulate homeostasis and differentiation of the intestinal epithelium. In intestinal cancers, these pathways are frequently dysregulated. Constitutive activation of canonical Wnt signaling resulting in high β-catenin transcriptional activity is believed to drive CRC tumorigenesis, however current molecular targeted therapies have limited efficacy. Therefore, identifying transcription factor networks between Notch and Wnt pathways associated with β-catenin in CRC is critical for further developing new therapeutic targets and strategies. We have identified that SAM Pointed Domain Ets transcription Factor (SPDEF), a downstream target of ATOH1, plays an important role in Notchdriven cell cycle exit and terminal differentiation in the adult intestinal epithelium. In CRC, we have recently published that SPDEF functions as a tumor suppressor. Our data indicated SPDEF re-expression in colon tumors is sufficient to prevent colon tumor cell proliferation by negatively regulating Wnt/β-catenin signaling, resulting in downregulation of β-catenin target genes in vivo. Moreover, consistent with this observation, transfection of human CRC cell lines with SPDEF inhibits β-catenin transcriptional activity in vitro. Here, we proposed a model that SPDEF inhibits β-catenin transcriptional activity through protein-protein interaction at the chromatin level. To investigate the mechanism of SPDEF-mediated inhibition of β-catenin transcriptional activity, tagged wildtype SPDEF or truncated SPDEF mutants were overexpressed in human CRC cell lines followed by β-catenin transcriptional activity assay, coimmunoprecipitation (co-IP), and immunofluorescent staining. Our data suggested that SPDEF inhibits β-catenin transcriptional activity and the expression of β-catenin targets in human CRC cell lines, which is consistent with our previous studies. Moreover, SPDEF interacts with nuclear β-catenin in both human CRC cell lines in vitro and mouse intestinal crypts under physiological conditions in vivo. The binding regions for SPDEF and β-catenin interaction were mapped by using truncated SPDEF and βcatenin mutants. Interestingly, our results indicated that the DNA-binding domain of SPDEF is unnecessary for it to bind to β-catenin or to inhibit β-catenin transcriptional activity. Finally, our chromatin immunoprecipitation (ChIP) data suggested that SPDEF binds to β-catenin target sites on chromatin, resulting in the disruption of β-catenin— chromatin interaction. Taken together, these results support a model wherein SPDEF may be a pivotal link between Notch and Wnt/β-catenin pathways through interacting with β-catenin in chromatin to repress proliferation and regulate differentiation in the intestine. Contributors: Lo, Yuan-Hung; Noah, Taeko; Shroyer, Noah 185 BAYLOR COLLEGE OF MEDICINE GENETIC INHIBITION OF NOX2 ACTIVITY IMPORVES CONTRACTILE FUNCTION AND PATHOLOGY IN YOUNG AND MATURE MDX MUSCLE James Anthony Loehr Department of Molecular Physiology & Biophysics Advisor: George Rodney, Ph.D.-Department of Molecular Physiology & Biophysics Duchenne Muscular Dystrophy (DMD) is an X-linked progressive degenerative disease caused by a mutation in the gene encoding dystrophin. Dystrophic muscle is characterized by increased reactive oxygen species (ROS) production and Ca2+ influx, decreased contractile function and morphological alterations. In mdx mice, a model of DMD, nicotinamide adenine dinucleotide phosphatase (NADPH oxidase or Nox2) ROS production is elevated early in the disease progression, resulting in increased Ca2+ influx and decreased muscle function. We hypothesized that inhibiting Nox2 ROS would protect against the pathophysiological changes associated with dystrophy, which would be maintained as the mice aged. Genetic inhibition of Nox2 activity resulted in decreased ROS production and partial recovery of force in young (5-7 wks) mice. The increased force was maintained with age (15-18 wks) while mdx force declined. We also demonstrate that decreasing Nox2 ROS reduced Ca2+ influx and central nuclei while maintaining CSA and fiber type distribution in young and mature mice. Our data support our hypothesis that Nox2 inhibition improves the pathophysiology of mdx muscle and identify Nox2 as a potential therapeutic target to preserve muscle function in DMD. Contributors: Pal, Rituraj; Abo-Zahrah, Reem; Rodney, George G. 186 2014 GRADUATE STUDENT SYMPOSIUM BRAF FUSIONS ARE POTENTIAL ONCOGENIC DRIVERS WITH SENSITIVITY TO MAPK PATHWAY INHIBITORS Hengyu Lu Integrative Program in Molecular and Biomedical Sciences Advisor: Kenneth Scott, Ph.D.-Department of Molecular & Human Genetics Fusion genes arising from chromosomal translocations, deletions and inversions play a central role in the development of cancer. Next generation sequencing efforts by the TCGA and others are identifying numerous fusion gene transcripts in tumors, the majority of which remain to be functionally characterized due to significant technical challenges related to their cloning. To circumvent this bottleneck, we have developed a completely novel strategy to build fusion genes that leverages our platform of >32,000 ORF clones and multi-fragment DNA recombination. We have used this approach to successfully validate bona fide fusion drivers (e.g., BCR-ABL, EML4-ALK and ETV6NTRK3) in proof-of-concept studies demonstrating our ability to rapidly deliver fusion genes with functional activity. In a pilot study of a panel of novel fusion genes identified in melanoma, our work revealed potent transforming activity of BRAF fusions, TAX1BP1-BRAF, CDC27-BRAF, and ATG7-BRAF, which strongly activate the MAPK signaling and confers sensitivity to BRAF and MEK inhibitors. This may indicate use of MAPK inhibitors therapy for patients with tumors harboring oncogenic BARF fusions. We anticipate scaling this cloning strategy to functionalize numerous fusion genes discovered by TCGA. Contributors: Scott, Kenneth 187 BAYLOR COLLEGE OF MEDICINE ATXN1-CIC CO-REPRESSOR COMPLEX IS CRITICAL FOR PROPER NEURON/GLIA DIFFERENTIATION IN THE DEVELOPING CORTEX Hsiang-Chih Lu Program in Developmental Biology Advisor: Huda Zoghbi, M.D.-Department of Pediatrics Expansion of glutamine-encoding CAG repeats in ATAXIN-1 (ATXN1) causes spinocerebellar ataxia type 1 (SCA1). Research over the past 20 years has provided insight into SCA1 pathogenesis, whereby genetic studies have demonstrated that SCA1 is mostly caused by a gain-of-function mechanism. Little is known, however, about the specific function of ATXN1. Thus, our lab sought out protein interactors of ATXN1 to understand its function better. We discovered that the most salient of the interactors (that also modify the SCA1 phenotype) are ATXN1’s functionally redundant paralog Ataxin-1-like (ATXN1L) and the transcriptional repressor Capicua (CIC). These three proteins, all highly expressed in brain, form a co-repressor complex in vivo, and depend on one another for stability. Moreover, loss-of-function mutations in ATXN1 or CIC have been implicated in developmental disorders such as autism spectrum disorder and intellectual disability, suggesting that this protein complex is important for the development of the nervous system. Atxn1-/-; Atxn1l-/- (double knockout, DKO) mice and Cic-/- mice die perinatally with defects in multiple organs, demonstrating that these proteins have critical roles during development. We therefore propose that ATXN1-CIC complex is important for the proper development and function of the nervous system. To test this hypothesis and to understand the function of this complex in the brain we must bypass the perinatal lethality. Accordingly, we used the Cre-lox technology to delete the genes encoding this complex conditionally in the nervous system. We generated conditional knockout of either Atxn1-Atxn1l or Cic in the developing forebrain using an Emx1-Cre. Loss of either Atxn1-Atxn1l or Cic causes multiple behavioral abnormalities including increased motor activity, reduced anxiety, and impaired memory. Histological studies showed that while the layering architecture of the cortical neurons is largely preserved in these animals, the thickness of superficial cortical layers (layer II-IV) is reduced. Moreover, there is an increased number of glia, suggesting that there is a disturbance in the balance of neuron/glia differentiation. These results show that ATXN1-CIC complex is indispensable for normal brain development and its absence results in multiple behavioral deficits. We are now analyzing the defects in the timing of neuro/gliogenesis transition in the embryonic cortex. In addition, we are performing transcriptomic analysis to further dissect the molecular mechanism that leads to these defects. Contributors: Lu, Hsiang-Chih; Liu, Xiuyun; Zoghbi, Huda 188 2014 GRADUATE STUDENT SYMPOSIUM MICRORNA-22 ENHANCES TH17 CELL DEVELOPMENT AND EMPHYSEMA THROUGH HISTONE DEACETYLASE 4 Wen Lu Department of Pathology & Immunology Advisor: David Corry, M.D.-Department of Medicine Emphysema is one of the most common causes of death worldwide without any effective treatments. Emphysema is mainly caused by cigarette smoke and particle inhalation and has a strong correlation with lung cancer. Cigarette/particle induced lung antigen presenting cell (APC) activation and downstream T helper 17 cell (TH17) development have been identified as key causative steps of emphysema development. We report that microRNA-22 (Mir22), an oncogenic microRNA identified in lung cancers, is also required for emphysema pathogenesis. Loss of Mir22 protects mice from emphysema. Neutralizing Mir22 in lung using sponge Lock Nucleic Acid (LNA) is able to reverse fully developed emphysema. Mir22 is highly expressed in APCs and its expression in lung APCs correlates with emphysema disease severity in both human and mouse. Mir22 deficient lung APCs induce less TH17 cells correlated with their less pro-inflammatory cytokine secretion and lower expression level of surface costimulatory molecules. Mice with selective Mir22 deficiency in CD11c+ APCs are also protected from emphysema. Adoptively transferring Mir22 sufficient lung APCs from emphysematous donors is able to induce emphysema in Mir22 deficient recipients, suggesting Mir22 in APC is both sufficient and necessary for emphysema development. Utilizing gene expression microarray we identify histone deacetylase 4 (HDAC4) as the target of Mir22 in lung APCs. We further demonstrate HDAC4 negatively regulate c-Jun expression, a critical transcriptional subunit regulating pro-inflammatory gene expression. In this study, Mir22 is the first microRNA functionally linked to emphysema pathogenesis. We have identified a novel Mir22/HDAC4/c-Jun axis regulating APC activation and innate immunity, with great therapeutic potentials treating emphysema and other autoimmune diseases. Contributors: Lu, Wen; You, Ran; Yuan, Xiaoyi; Yang, Tianshu; Tour, James; Kheradmand, Farrah; Corry, David; 189 BAYLOR COLLEGE OF MEDICINE FUNCTIONAL ROLE OF MIR205 IN MAMMARY GLAND DEVELOPMENT AND STEM CELL MAINTENANCE Yang Lu Integrative Program in Molecular and Biomedical Sciences Advisor: Jeffrey Rosen, Ph.D.-Department of Molecular & Cellular Biology While miRNAs play an important role in normal development and cancer, little is known about the functional role of miRNAs in mammary gland development. The analysis of miRNA expression in the mouse mammary epithelial cells has revealed high expression of miR-205 in basal stem cell-enriched population. Consistent with this observation, miR205 over-expression in the mouse mammary epithelial cell (MEC) line COMMA-DβGeo leads to an expansion of the Sca-1+ progenitor cell population, decreased cell size, increased cellular proliferation and colony-forming potential. Based upon these results, we hypothesize that miR205 is required in mammary gland development and stem cell maintenance. By using mice with a conditional floxed miR205 allele containing a Neo-LacZ cassette, we are able to show by X-gal staining that miR205 is preferentially expressed in the cap cell layer of terminal end buds. Furthermore, functional cell transplantation assays of miR205 null cells display severely impaired outgrowths in the first generation. Rudimentary miR205 null transplants lack a K5+ basal cell layer and laminin staining, indicating disruption of basement membrane deposition as well as the loss of a basal stem/progenitor cell population. This might be the underlying reason for the impaired repopulating capacity of miR205 null cells. The observed growth disadvantage was further verified using in vitro mammosphere assays. miR205 null cells show a slight self-renewal defect in primary assays, with mammosphere formation efficiency (MFE) of 0.4%. However, the MFE of miR205 null cells dramatically decreases to 0.005% in the secondary assays, while WT cells still exhibit a 0.1% MFE. In addition, microarray data from Dr. Elsa Flores lab shows that knockout of p63 results in down-regulation of miR205 in skin. Therefore our next step is to study if the p63-miR205 regulatory axis is associated with the ability of miR205 to influence mammary gland stem cell fate. To conclude, our preliminary finding suggests a pivotal role of miR205 in maintaining the mammary gland stem cell population and outgrowth potential. Unraveling miR205’s function in normal development will further help us to study its role in promoting tumorigenesis possibly through regulating the generation and function of cancer stem cells. Supported by NIH grant CA16303. Contributors: Lu, Yang, Rosen, Jeffrey 190 2014 GRADUATE STUDENT SYMPOSIUM THE FRAGILE X PROTEINS FMRP AND FXR2P COOPERATE TO REGULATE GLUCOSE METABOLISM IN MICE Jeannette Galande Lumaban Department of Molecular & Human Genetics Advisor: David Nelson, Ph.D.-Department of Molecular & Human Genetics Fragile X syndrome results from loss of FMR1 expression. Individuals with the disorder exhibit not only intellectual disability, but also an array of physical and behavioral abnormalities, including sleep difficulties. Studies in mice demonstrated that Fmr1, along with its paralog Fxr2, regulate circadian behavior, and that their absence disrupts expression and cycling of essential clock mRNAs in the liver. Recent reports have identified circadian genes to be essential for normal metabolism. Here we describe the metabolic defects that arise in mice mutated for both Fmr1 and Fxr2. These mice have reduced fat deposits compared with age- and weight-matched controls. Several metabolic markers show either low levels in plasma or abnormal circadian cycling (or both). Insulin levels are consistently low regardless of light exposure and feeding conditions and the animals are extremely sensitive to injected insulin. Glucose production from introduced pyruvate and glucagon is impaired and the mice quickly clear injected glucose. These mice also have higher food intake and higher VO2 and VCO2 levels. We analyzed liver expression of genes involved in glucose homeostasis and found several that are expressed differentially in the mutant mice. These results point to the involvement of Fmr1 and Fxr2 in maintaining the normal metabolic state in mice. Contributors: Lumaban, Jeannette; Nelson, David 191 BAYLOR COLLEGE OF MEDICINE TYROSINE PHOSPHORYLATED IRAK4 MODULATION OF TLR ACTIVATION Jessica Leigh Lunsford Department of Pathology & Immunology Advisor: Jonathan Levitt, Ph.D.-Department of Pathology & Immunology Src homology region 2 domain-containing phosphatase-1 (SHP-1) is a tyrosine phosphatase which inhibits NF-κB signaling through interference in MyD88/IRAK4dependent toll-like receptor (TLR) signaling. However, the specific substrates of SHP-1 in this pathway are unknown. Our lab has previously identified IRAK4 as a binding partner of SHP-1, despite zero reports of tyrosine phosphorylated IRAK4 in the literature. Further, expression of a dominant negative form of SHP-1 leads to accumulation of tyrosine phosphorylated IRAK4 in dendritic cells. We hypothesized that SHP-1 interacts with IRAK4 to inhibit TLR-dependent NF-κB signaling. We have identified three tyrosine residues in the IRAK4 structure which are consistent with SHP1 binding motifs: Y48, Y371 and Y413. Since phenylalanine resembles a tyrosine which cannot be phosphorylated we mutated these three tyrosine sites to phenylalanines, singly and in double and triple mutant permutations. RAW264.7 cells, a murine macrophage like cell line, were transfected with various IRAK4 mutations, and activated overnight with LPS. Activation was assayed by IL-6 ELISA and NFkB activity. Compared to transfected WT IRAK4, all mutations reduce activation of RAW264.7 cells. Continuing experiments will explore causes of downregulation in the endogenous TLR pathway, the kinase responsible for tyrosine phosphorylation after TLR activation, and the involvment of SHP-1 in this pathway. Contributors: Konduri, Vanaja; Decker, William; Levitt, Jonathan 192 2014 GRADUATE STUDENT SYMPOSIUM IDENTIFY TRIC SUBUNITS IN APO-STATE IN CRYO-EM RECONSTRUCTION Boxue Ma Department of Biochemistry & Molecular Biology Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology Chaperonin TRiC is the essential protein-folding machine in eukaryotic cells. It is a hetero-oligomeric complex containing two rings sitting back to back, which serve as folding chambers that sequester substrate from the cellular environment. Each ring consists of eight homologous subunits, known as CCT1-CCT8. In vivo and in vitro studies have shown that the eight subunits have varied affinity to different substrates. Therefore, it is important to identify the subunits for understanding the mechanism of how TRiC interacts with substrates. Several studies have been carried out, however, due to the high structural similarity among the eight subunits, it is difficult to distinguish them directly even at near atomic resolution. Thus, we propose to perform cryo electron microscopy single particle analysis to reconstruct the structure of an engineered yeast TRiC which has a calmodulin binding peptide (CBP) fused into one of the eight subunits. The CBP tag will bind to calmodulin, which serves as a label on the specific subunit in cryo-EM reconstruction. Combining with the two-fold symmetry between the two CCT rings and the asymmetric feature within each ring, labeling and identifying subunit CCT2 or CCT6 will confirm the arrangement of all the eight subunits. Such labeling allows the recognition of the subunits of apo-state TRiC at medium resolution. Furthermore, the labeled TRiC will be a powerful tool for future structural study to reveal the substratebinding sites on TRiC. Contributors: Ma, Boxue; Gestaut, Dan; Chiu, Wah 193 BAYLOR COLLEGE OF MEDICINE ROLE OF ASL IN ALTERED CELLULAR ENERGETICS AND LIVER DYSFUNCTION Simran Madan Program in Translational Biology & Molecular Medicine Advisor: Brendan Lee, M.D./Ph.D.-Department of Molecular & Human Genetics Brett Graham, M.D./Ph.D.-Department of Molecular & Human Genetics Deficiency of urea cycle enzyme argininosuccinate lyase (ASL) is is characterized by hyperammonemia, vomiting, seizures and if left untreated, can lead to coma and death. Despite early intervention and management, some patients still develop long term complications such as hypertension, liver dysfunction, and neurocognitive deficits. In this study, we are characterizing the liver pathology in a hypomorphic mouse model of ASL. Plasma from ASL deficient mice show significantly elevated liver enzymes AST and ALT, which is an indication of liver damage. Electron microscopy on livers from ASL hypomorphic mice showed dysmorphic mitochondria and an unusually large number of lysosomes. Histological analysis show increased glycogen mobilization. Previously, we have shown that ASL is an important regulator for nitric oxide production. Proteomic analysis of livers from ASL hypomorphic mice compared to WT mice showed that loss of ASL results in loss of nitrosylation of several key metabolic enzymes in the citric acid cycle and the electron transport chain. Currently, we are investigating if this loss of nitrosylation causes altered mitochondrial energetics and mitochondrial dysfunction which may explain the liver dysfunction seen in ASL deficient mice and patients. Contributors: Madan, Simran; Burrage, Lindsay; Chen, Yuqing; Bertin, Terry; Graham, Brett; Lee, Brendan 194 2014 GRADUATE STUDENT SYMPOSIUM INHIBITION OF DNA REPLICATION INITIATION BY CHROMOSOME TETHERING IN BACTERIA David Russell Magnan Integrative Program in Molecular and Biomedical Sciences Advisor: David Bates, Ph.D.-Department of Molecular & Human Genetics All cells must initiate new rounds of DNA replication once and only once per cell division in order to maintain genomic stability. E. coli strictly regulates initiation timing and periodicity through multiple mechanism including staged assembly of replication initiation factors on the replication origin oriC and cell-cycle dependent changes to oriC structure. Additional data suggests that oriC position in the cell influences capacity of origins to fire. To date however no mechanistic link between cell division and replication initiation has been discovered in E. coli. In order to study these models we developed a system to disrupt position and structure of specific chromosomal loci. A TetR (DNA binding) fusion protein normally used to visualize loci in living cells was fused to a membrane-inserted chemotaxis protein, creating a physical linkage between the chromosome and the cell membrane at specific loci of our choosing. Tethering chromosomal loci to the cell membrane using this system inhibits cell division and slowly reduces cell viability. Tethering does not cause DNA damage or disrupt membrane integrity. Tethering does not inhibit replication elongation or nucleoid segregation but does strongly inhibit replication initiation. Surprisingly, tethering oriCdistal loci inhibit initiation to the same extent as tethering oriC-proximal loci, albeit in a delayed manner. Global chromosome structure is disrupted in tethered strains, suggesting we may have disrupted a long-sought mechanistic link between cell division and replication initiation. Contributors: Magnan, David; Joshi, Mohan; Bates, David 195 BAYLOR COLLEGE OF MEDICINE ROLE OF NLRP7 IN IMPRINTING AND ANOMALOUS REPRODUCTIVE OUTCOMES Sangeetha Kaveri Mahadevan Program in Translational Biology & Molecular Medicine Advisor: Ignatia Van Den Veyver, M.D.-Department of Obstetrics & Gynecology Carlos Bacino, M.D.-Department of Molecular & Human Genetics Maternal effect mutations in NLRP7 and less commonly, KHDC3L, cause Biparental Hydatidiform Moles (BiHM), a rare form of Gestational Trophoblastic Disease characterized by degenerate cystic villi, hyperproliferative trophoblast, abnormally high β-HCG levels and absent fetal development. Unlike their more common androgenetically derived counterparts, BiHM pregnancies are recurrent, biparentally inherited and tissues from these pregnancies exhibit loss of methylation at maternally imprinted loci. Because NLRP7 is not present in rodents or other commonly used animal models, we generated NLRP7 knockdown and overexpressing hESC lines which have enabled us to gain insight into NLRP7's function in an in vivo system that most closely resembles germ cells. Whole genome methylation and RNA-Seq studies on the NLRP7 knockdown hESC have revealed altered methylation and expression at several hundred genes suggesting loss of NLRP7 has a more genome wide influence on DNA methylation and expression that is not restricted to imprinted loci. Expression studies on these cells further revealed a cell cycle dependent increase and intracellular redistribution of both BiHM proteins NLRP7 and KHDC3L. Nuclear staining was observed during the mitotic phase of the cell cycle which is a novel observation given that NLRP proteins are known to be cytoplasmic in localization. This observation of nuclear staining also supports the genome wide alterations in DNA methylation and expression observed in NLRP7 knockdown cells. A yeast two hybrid screen and hypothesis driven candidate approach was employed to detect interacting partners of NLRP7 and this revealed positive interactions with NPM1 and CFP1. Given that the expression of both these CFP1 and NPM1 oscillates during the cell cycle, further investigations into the interaction of NLRP7 with these proteins could uncover the mechanisms, by which (i) NLRP7’s expression is regulated during the course of the cell cycle, (ii) NLRP7 impacts DNA methylation and expression at imprinted and nonimprinted loci. These studies are currently ongoing. Contributors: Mahadevan, Sangeetha; Wan, Ying-Wooi; Liu, Zhandong; Van den Veyver, Ignatia 196 2014 GRADUATE STUDENT SYMPOSIUM REGULATION OF E2F1 BY A DEUBIQUITINATING ENZYME, UCHL5, FOR DNA DAMAGE RESPONSE Christina Susan Mahanic Integrative Program in Molecular and Biomedical Sciences Advisor: Weei-Chin Lin, M.D./Ph.D.-Department of Medicine E2F1 is a transcription factor that plays a critical role in diverse cellular pathways such as cell cycle progression, apoptosis induction, and the DNA damage response. E2F1 has been shown to be necessary for inducing apoptosis in a chemotherapeutic response by transcriptionally activating target genes such as p73. The mechanism by which E2F1 differentially regulates transcription of proliferative genes and apoptotic genes remains unknown. Our co-IP/MS experiment identifying potential interacting proteins of E2F1 has identified a deubiquitinating enzyme (DUB), UCHL5, as a novel E2F1 interacting protein. Through further investigation, I have shown that E2F1 interacts with ectopically overexpressed UCHL5 in 293T cells with a slight induction after treatment with a chemotherapeutic drug, adriamcyin. In 293T cells, the accumulation of ubiquitinated E2F1 (K63-chain specific) is decreased in the presence of ectopic UCHL5. This suggests that UCHL5 is a potential DUB for E2F1. Through using a semi-endogenous ubiquitination assay, E2F1 is shown to be ubiquitinated through K63-specific linkages and when E2F1 and UbK63 are co-expressed, E2F1 transcriptional activity decreases. Conversely, a reporter assay has shown that E2F1 transcriptional activity is increased when UCHL5 is present, and Q-RT-PCR analysis has shown that p73, an E2F1 target gene, is upregulated in the presence of UCHL5. This functional role of UCHL5 has been verified using a stable knockdown cell line with the reporter assay and Q-RT-PCR. As a functional assay, a Caspase 3/7 reporter assay was used to show that UCHL5 knockdown cell lines have less Caspase 3/7 activity when compared to the scrambled control, suggesting UCHL5 has a role in apoptosis. Finally, I have shown through chromatin immunoprecipitation (ChIP) that UCHL5 is localized at the site of E2F1-specific pro-apoptotic promoters such as APAF1, caspase 3 and 7, and PARP1. Prior studies have shown UCHL5 is able to interact with the INO80 chromatin-remodeling complex, also placing UCHL5 at the site of transcription. I hypothesize that UCHL5 is able to interact with E2F1 on the chromatin, and deubiquitinate E2F1, thus enabling an upregulation of transcriptional activity of genes required for apoptosis. I propose that ubiquitination is an additional regulation of transcriptional activity of E2F1. Discovering a mechanism in which E2F1 transcriptional activity is increased could lead to a possible target therapy that induces apoptosis in cancer cells. In conclusion, I have found that a DUB, UCHL5, interacts with E2F1 and deubiquitinates E2F1 in vivo and knockdown of UCHL5 decreases an apoptotic response when DNA damage is present. Contributors: Mahanic, Christina; Budhavarapu, Varija; Lin, Weei-Chin 197 BAYLOR COLLEGE OF MEDICINE HDAC-6 INHIBITION IN APP/PS1 MICE SHOWS A RECOVERY IN SHORT-TERM FEAR-ASSOCIATED MEMORY AND AXONAL TRANSPORT DEFICITS Tabassum Majid Program in Translational Biology & Molecular Medicine Advisors:Robia Pautler, Ph.D.-Department of Molecular Physiology & Biophysics Rachelle Doody, M.D./Ph.D.-Department of Neurology Current therapy for Alzheimer’s disease (AD) focuses on delaying progression, illustrating the need for more effective therapeutic targets. A target of interest in Alzheimer’s disease is HDAC6. Of all the HDAC classes, this enzyme has been shown to have roles in the clearance of protein aggregates, interact with microtubule-stabilizing proteins, and recruit chaperones for axonal motor proteins. In addition, HDAC6 is elevated in Alzheimer’s patient brains. HDAC6 genetic depletion studies in pre-clinical AD models have demonstrated ex vivo axonal transport is recovered. However, HDAC6 inhibitors have been met with limited success in pre-clinical models due to a lack of blood-brain-barrier penetration by selective HDAC6 inhibitors. Our previous work has demonstrated that axonal transport deficits are detectable in vivo using manganeseenhanced MRI (MEMRI) in Alzheimer’s models prior to the onset of amyloid deposits. Due to the crucial role of microtubule-based axonal transport in Alzheimer’s disease and our ability to measure it using MEMRI, we investigated the potential of a specific, potent HDAC6 inhibitor (ACY-738) in a mouse model of Alzheimer’s disease. ACY-738 has been shown to have specificity to HDAC-6 inhibition, permeation of the blood brain barrier, and its rapid clearance from brain and plasma in mouse models. Using a chow-based treatment of ACY-738, we sought to determine the effects of HDAC6 inhibition on axonal transport, behavior, and pathology in the APP/PS1 mouse model of AD. Group 1 received a treatment of ACY-738 for 21 days and were evaluated at 3 months of age. After treatment, we measure therapeutic levels in both brain and plasma of 3 month-old mice. We also demonstrate a recovery in axonal transport deficits after 21 days of chow treatment. Group 2 was treated with ACY-738 and evaluation was completed at 6 months of age. Therapeutic levels of ACY-738 are present in both the plasma and brain of APP/PS1 mice. We demonstrate a recovery in short-term fearassociated learning and memory deficits and axonal transport deficits after 3 months of treatment with ACY-738. Our findings implicate the development of specific, targeted HDAC6 inhibitors for AD patients. The completion of this pre-clinical proof of concept study in the APP/PS1 mouse model of AD indicate a recovery in short-term learning and memory and axonal transport deficits. Further investigation into the effects of HDAC-6 inhibitors in AD will allow for the development of more effectively targeted therapeutic interventions to combat the deleterious effects of this devastating disease. Contributors: Majid, Tabassum, Griffin, Deric, Criss II, Zachary, Pautler, Robia G. 198 2014 GRADUATE STUDENT SYMPOSIUM THE HOMOLOG OF A BACTERIAL STRESS RESPONSE REGULATOR IN C. ELEGANS- AN UNEXPECTED ROLE IN COLD TOLERANCE Keng Hou Mak Integrative Program in Molecular and Biomedical Sciences Advisor: Jue Wang, Ph.D.-Department of Molecular & Human Genetics Meng Wang, Ph.D.-Department of Molecular & Human Genetics The ability to sense and signal stress is essential for an organism to survive. Intracellular nucleotides are an important group of signaling molecules that relay diverse environmental or metabolic information. Among them, the highly phosphorylated nucleotides guanosine tetra(penta)phosphate ((p)ppGpp) play a major role in stress signaling in prokaryotes and are required for adapting bacterial physiology to changing environments, such as nutrient starvation and pathogenesis. It has been a longstanding mystery whether (p)ppGpp has physiological functions in metazoans. Recently, a protein family of (p)ppGpp regulatory enzymes was identified in higher animals, with one member each in the genomes of human, Drosophila and C. elegans. In Drosophila, MESH1 mutants have altered starvation tolerance, and the protein product hydrolyzes (p)ppGpp in vitro. However, no (p)ppGpp was detected in vivo, and the functions of MESH1 on the tissue and cellular levels are still unknown. This knowledge will provide key insight into physiological functions beyond starvation response and guide future efforts to identify (p)ppGpp or other MESH1 substrates in vivo. C. elegans serves as a good model because of its power to uncover gene function on both cell and tissue levels. To first test the physiological role of mesh-1 (encoding the uncharacterized C. elegans MESH1), we examined the starvation tolerance of mesh-1 mutants. Surprisingly, mesh-1 mutants show no change in starvation tolerance and display no apparent growth and morphology defects. However, they have enhanced tolerance to pre-freezing coldness, which can be complemented by overexpressing the wildtype gene, demonstrating the role of mesh-1 in cold sensitivity. Tissue-specific RNAi knockdown indicates mesh-1 activity in either the epidermis or intestine is functionally required for the phenotype. Because (p)ppGpp regulatory enzymes function in chloroplasts in plants, I am currently testing the hypothesis that MESH-1 regulates mitochondrial functions associated with cold tolerance. Future studies include identifying MESH-1 substrate(s) in vivo. Taken together, our results suggest a tissue-specific role for MESH-1 in thermal adaptation and extend the function of MESH1 beyond starvation response. Contributors: Mak, Keng Hou; Wang, Meng; Wang, Jue 199 BAYLOR COLLEGE OF MEDICINE A HIGH-THROUGHPUT SCREEN TO IDENTIFY NOVEL RNA FOCI-ASSOCIATED PROTEINS IN MYOTONIC DYSTROPHY Kathleen Seger Manning Integrative Program in Molecular and Biomedical Sciences Advisor: Thomas Cooper, M.D.-Department of Pathology & Immunology Myotonic dystrophy (DM) is the most common form of muscular dystrophy in adults. DM is characterized by myotonia, muscle wasting, cardiac conduction abnormalities, and central nervous system dysfunction, among other symptoms. Myotonic dystrophy type I (DM1) is caused by an unstable CTG repeat expansion in the 3’ UTR of the muscle kinase gene DMPK. The expanded CTG repeat is transcribed into CUG expanded repeat (CUGexp) RNA transcripts, which are retained in the nucleus and aggregate into distinct foci. CUGexp RNA sequesters Muscleblind-like (MBNL) family splicing factors and alters the activity of CUGBP Elav-like family (CELF) RNA binding proteins, leading to defects in post-transcriptional gene regulation for multiple genes. However, while misregulation of MBNL or CELF proteins is each sufficient to induce a subset of DM1 symptoms in mouse models of disease, some effects of CUGexp RNA are independent of MBNL and CELF dysfunction. To identify novel mechanisms of pathogenesis in DM1, we propose a high-throughput immunofluorescence-based assay to objectively screen for proteins associated with CUGexp RNA foci. While these CUGexp RNA foci are a characteristic feature of DM1, their full protein composition is unknown. A library of monoclonal antibodies will be robotically screened in immortalized cell lines from DM1 patients to identify antibodies that co-localize with CUGexp RNA foci by immunofluorescence. Positive hits will be identified by mass spectrometry. A comprehensive understanding of the protein-protein and protein-RNA interactions associated with CUGexp RNA foci will improve therapeutic targeting and may reveal novel mechanisms of pathogenesis in DM1. This project will also provide a framework for future studies of foci and aggregate composition in other disease models. Contributors: FUNCTIONAL ANALYSIS OF VAPB AND ITS RELEVANCE TO ALS 200 2014 GRADUATE STUDENT SYMPOSIUM Dongxue Mao Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that affects both upper and lower motor neurons, leading to muscle atrophy and paralysis. There is no cure for ALS and the pathogenesis is still poorly understood. VapB is a highly conserved type II integral membrane protein associated with the Endoplasmic Reticulum (ER). It is implicated in lipid transport, muscle mitochondrial dynamics and ER stress. Mutations in VapB are identified through linkage analysis in families with ALS and spinal muscular atrophy. VapB mRNA or protein level is decreased in the spinal cord of sporadic ALS patients as well as in ALS mouse models, indicating there is correlation between VapB and ALS. Previous data suggest that the N-terminal MSP domain of VapB is cleaved and secreted and ectopically expressed VapBP56S aggregates in the ER, leading to the elevation of ER stress and the interference of MSP secretion in Drosophila melanogaster. In C. elegans, secreted MSP binds to the Ephrin receptor (Eph), Lar-like protein-tyrosine-phosphatase (CLR) and Roundabout (Robo) in the muscle. This interaction is required for proper cytoskeleton organization, which affects mitochondrial morphology and localization in the muscle. In SODG93A transgenic mice, a wellestablished animal model for ALS, we find that full length VapB protein level is elevated in muscle but the cleaved MSP is reduced. This indicates that insufficient MSP cleavage or secretion may lead to muscle atrophy and a feed-back mechanism is required to upregulate VapB protein synthesis to maintain the MSP pool. The MSP cleavage is present in most mouse tissue such as nervous system, heart and liver. Secreted MSP is also detected in human blood serum and Cerebrospinal Fluid (CSF). Addition of recombinant MSP induces tyrosine phosphorylation of RTKs, such as EGFR and FGFR in HEK293 cells. In summary, the neuronal or muscular degeneration in ALS patients or disease model correlates with loss of function of VapB or defect in MSP secretion. Therefore, we propose that MSP function as a hormone. It is a beneficial factor that prevents muscle atrophy by modulating RTKs. Contributors: Mao, Dongxue; Lin, Guang; Tepe, Burak; Bellen, Hugo 201 BAYLOR COLLEGE OF MEDICINE DEGENERATION AND PROTECTION OF AXONAL SUB DOMAINS AFTER OPTIC NERVE CRUSH Miguel Marin Department of Neuroscience Advisor: Matthew Rasband, Ph.D.-Department of Neuroscience Myelinated axons are divided into several distinct domains, which includes the axon initial segment (AIS), nodes of Ranvier, paranodes, and juxtaparanodes. The AIS serves as both a physical barrier between the axonal and somato-dendritic compartments of the neuron and as the site of action potential (AP) initiation. Nodes of Ranvier are responsible for the rapid and efficient propagation of APs along the axon. Disruption of the AIS or nodes of Ranvier by genetic and/or pharmacological manipulation has a dramatic impact on the central nervous system. With this in mind, we have designed a series of experiments, which will allow us to assess the efficacy of neuroprotective paradigms upon axons of the central nervous system after insult. Using the optic nerve crush injury model, we have established a timeline for degenerative events of the nodes of Ranvier of the optic nerve and AIS of retinal ganglion cells. We have established that loss of nodes of Ranvier begins 6 hours after injury and progresses both distal and proximal to the injury site. A total loss of nodes of Ranvier occurs 1 week after injury and persists 1-month post crush. Loss of AIS in retinal ganglion cells begins 24 hours after injury and persists 1-month post crush. We have assessed the neuroprotective efficacy of MDL-28170 - a calpain inhibitor that has been shown to protect from AIS degeneration after ischemic injury. MDL-28170 spares nodes of Ranvier and AIS from degeneration 24 hours post injury. Contributors: Miguel Marin1 & Matthew N. Rasband1 & 2 202 2014 GRADUATE STUDENT SYMPOSIUM IDENTIFICATION OF NOVEL ONCOGENIC DRIVERS USING A BREAST CANCER MODEL INDUCED BY CONDITIONAL INACTIVATIN OF P53 AND PTEN IN ADULT MICE Jarrod Don Martinez Department of Molecular & Cellular Biology Advisor: Jianming Xu, Ph.D.-Department of Molecular & Cellular Biology PTEN loss occurs approximately 33-48% across different molecular subtypes of breast cancer (BC) and has correlations with poor prognosis and disease related death. In conjunction, P53 alterations or deletions in BCs has been reported to be as high as 80%. To date few studies have examined the effects of P53 and PTEN dual deletions and have yielded important insights. Most importantly these studies show that there is an important selective pressure to deactivate or delete P53 when PTEN is lost. Our model system will use the RCAS-CRE/TVA system from which we will be able in infect mammary epithelial cells and subsequently cause the deletion of both P53 and PTEN alleles and the cleavage of the RFP to GFP reporter. Sequential genomic profiling of the cancers pathological progression from initiation to distant metastases will allow us to obtain a comprehensive view of the genomic changes that occur throughout the course of the disease. Despite the heterogeneity of mutations an genomic alterations that accompany cancer progression, we hypothesize that there are certain genomic changes that must occur sequentially during the progression of the disease that allows for BC cells to initiate progress, and metastasize to distant organs. To test this hypothesis we will use the following aims: Aim 1: To characterize and profile the malignant progression of the resulting PTEN and P53 null tumors from our novel TVA+ R/G+ PTENf/f P53f/f mouse model. Aim 2: To determine the genomic changes at DNA/RNA levels that occur after the deletion of PTEN and P53 in various stages of tumorigenesis and metastasis of the transformed mammary epithelial cells. Approximately 90 percent of BC related deaths are attributed to metastasis, making it the greatest threat to overall patient survival. The understanding of the molecular mechanisms regulating the various stages of tumorigenesis and metastasis is therefore essential in the development of high risk management and new therapies for patients. Contributors: Qin, Li Ph.D; Xu, Yixiang; Li, Yi Ph.D.; Xu, Jianming Ph.D. 203 BAYLOR COLLEGE OF MEDICINE EXTRACELLULAR MECHANISMS OF TFEB-MEDIATED PTAU CLEARANCE Heidi Martini-Stoica Program in Translational Biology & Molecular Medicine/M.D.-Ph.D. Program Advisor: Hui Zheng, Ph.D.-Department of Molecular & Human Genetics Joshua Shulman, M.D./Ph.D.-Department of Neurology Presently, tau-targeting therapies to treat Alzheimer’s disease (AD) are limited by efficacy and safety. To date, the majority of therapeutic trials have focused on Aβ because of the strong genetic evidence linking mutations in APP and presenilins to familial AD. However, tau-based neurofibrillary tangle (NFT) pathology correlates more closely with cognitive decline than Aβ plaques. Thus, the failure of Aβ directed AD therapies may in part be due to their inability to target AD’s NFT pathology. Thus, the long-term goal of this work is to gain a better understanding of the cellular mechanisms of NFT clearance in order to develop more effective therapies. The overall objective of this proposal is to dissect the multiple roles of transcription factor EB (TFEB), a critical regulator of lysosomal biogenesis, in clearing these pathological forms of tau. The central hypothesis is that neuronal exocytosis of tau and subsequent astroglial uptake are crucial components of TFEB-mediated clearance of aberrant tau in diseases of tauopathy, in addition to TFEB’s cell-autonomous effect in enhancing the autophagylysosomal pathway (ALP). This hypothesis has been formulated based on recent work demonstrating the therapeutic potential of TFEB in ameliorating tau pathology in a mouse model of tauopathy. In addition, TFEB transcriptionally activates lysosomal exocytosis genes to promote cellular clearance, as well as endocytosis genes to promote cellular uptake. These known targets of TFEB suggest an extracellular mechanism of aberrant tau clearance that may involve neurons discarding tau extracellularly through lysosomal exocytosis and astrocytes subsequently taking up tau through endocytosis. In the astrocytes, tau could then undergo clearance through the ALP, also enhanced by TFEB. In order to accomplish the overall objective for the current proposal, the central hypothesis will be tested by pursuing the following specific aims: 1) Determine how TFEB affects tau handling in neurons using in vitro and in vivo overexpression of TFEB in neurons while measuring extracellular release of tau. 2) Determine how TFEB affects the role of astrocytes in tauopathies by measuring TFEBmediated astrocyte uptake of tau in vitro and determining the therapeutic efficacy of astrocyte specific TFEB overexpression in a mouse model of tauopathy. The successful completion of these aims will define the extracellular mechanisms of TFEB’s action and provide valuable insight into the poorly defined role of astrocytes in tauopathies. Clarification of the extracellular mechanisms of TFEB-mediated pTau clearance is expected to identify novel therapeutic targets to guide the development of new therapeutic strategies in the treatment of AD. Contributors: Martini-Stoica, Heidi; Zheng, Hui 204 2014 GRADUATE STUDENT SYMPOSIUM DROSOPHILA DEFICIENT FOR PORIN PROVIDE A LINK BETWEEN FAT METABOLISM AND NEUROLOGICAL FUNCTION Ruchi Masand Department of Molecular & Human Genetics Advisor: Brett Graham, M.D./Ph.D.-Department of Molecular & Human Genetics The voltage-dependent anion channel (VDAC or porin) is an integral membrane protein present in the mitochondrial outer membrane (MOM). VDACs are not only the predominant determinant of MOM permeability but also integrate mitochondrial function and other cellular pathways by interacting with various mitochondrial and cytoplasmic proteins; however, details of these functions as well as the interacting pathways remain poorly understood. VDACs have also been implicated to play a pathogenic role in several human diseases including cancer, diabetes, Alzheimer’s and cardiac ischemiareperfusion injury. Flies mutant for porin (the predominant VDAC in Drosophila) demonstrate energy metabolism defects, neurologic dysfunction with abnormal mitochondrial distribution in motor neurons and male infertility. A pilot modifier screen identified multiple deletions that suppress male infertility in porin mutants, including one deletion that also suppressed neuronal dysfunction in porin mutants. Deletion mapping identified Lsp2, a hexamerin expressed in the fat body, as a suppressor of porin mutant CNS phenotype. Expression microarray analysis and qRT-PCR on adult porin deficient flies revealed extremely high levels of Larval Serum Proteins (LSPs) as well as Fat Body Proteins (FBPs), all of which are typically highly expressed in the larval Drosophila fat body, which is analogous to the adipose tissue and liver in mammals as the major site of energy metabolism and fat storage in the fly. In addition, we also observed significantly lower levels of triglycerides (TAG) in flies deficient for porin. Elevated levels of LSPs and FBPs have also been reported in Drosophila mutant for technical knockout (tko), previously reported as showing respiratory chain deficiency, developmental delay, and neurological abnormalities. A p-element insertion allele of Lsp2 was able to rescue the increased bang sensitivity as well as the secondary complex I deficiency phenotypes seen in porin mutants. Also, both CNS and fat body specific expression of porin in the mutant flies was able to rescue the increased bang sensitivity phenotype. Since tissue specific expression of porin in the fat body, the key metabolic tissue in the fly, is sufficient to rescue the CNS phenotype in porin mutants and some of the key genes expressed almost exclusively in the fat body are elevated in both porin and tko mutants, our data suggests a possible link between mitochondria, fat body (i.e, adipose tissue) metabolism and neurological function. Contributors: Masand, Ruchi; Graham, Brett H. 205 BAYLOR COLLEGE OF MEDICINE DEVELOPMENT OF A LARGE ANIMAL MODEL TO EVALUATE CAR T-CELL THERAPY: EX VIVO ACTIVATION, EXPANSION, AND GENETIC MODIFICATION OF CANINE T-CELLS Melinda Mata Department of Pathology & Immunology Advisor: Stephen Gottschalk, M.D.-Department of Pediatrics Adoptive transfer of T-cells expressing chimeric antigen receptors (CARs) has shown promising anti-tumor activity in early phase clinical studies, especially for hematological malignancies. However, several patients developed adverse events after CAR T-cell infusions, which were not predicted by preclinical studies since most murine models do not reliably mimic human disease. Dogs are excellent models of human disease since canine tumors exhibit similar genetic abnormalities, tumor location, and comparable metastatic spread in a natural tumor environment. We reasoned that developing an adoptive T-cell therapy approach for spontaneous tumors occurring in dogs, such as osteosarcoma (OS), would more closely reproduce the condition in human cancer and allow us to further evaluate CAR T-cell therapy prior to human clinical trials. To generate CAR-expressing canine T-cells we developed expansion and transduction protocols that allow for the generation of sufficient numbers of CARexpressing canine T-cells for future clinical studies in dogs within 2 weeks of ex vivo culture. Seven days post-activation of canine T-cells with irradiated K562-APC, PHA, and IL21, cell lines consisted predominately of CD3-positive T-cells (83 ± 10.7%) with a mixture of CD4- and CD8-positive subsets. Restimulation of T-cells in Grex tissue culture devices with the addition of IL2 resulted in 100-fold expansion of cells. Since canine OS tumor biopsies and cell lines express the tumor antigen HER2, we retrovirally transduced canine T-cells 3 days post-activation with GALV-pseudotyped retroviral particles encoding a HER2-specific CAR. Post-transduction up to 38% of canine T-cells expressed HER2-CARs and were able to recognize canine HER2, secrete IFNγ, and lyse HER2+ canine OS tumor cell lines in an antigen dependent manner. To reduce the potential immunogenicity of the CAR we evaluated a CAR with canine-derived transmembrane and signaling domains, and found no functional difference between human and canine CARs. Hence, we have successfully developed a strategy to generate CAR-expressing canine T-cells for future preclinical studies in dogs. Testing Tcell therapies in an immunocompetent, outbred animal model may improve our ability to predict their safety and efficacy prior to conducting studies in humans. Contributors: Mata, Melinda; Vera, Juan; Gerken, Claudia; Rooney, Cliona M.; Miller, Tasha; Pfent Catherine; Wang, Lisa L.; Wilson-Robles, Heather M.; Gottschalk, Stephen 206 2014 GRADUATE STUDENT SYMPOSIUM DNMT3A LOSS PREDISPOSES MURINE HEMATOPOIETIC STEM CELLS TO MALIGNANT TRANSFORMATION Allison Mayle Department of Molecular & Human Genetics Advisor: Margaret Goodell, Ph.D.-Department of Pediatrics DNA METHYLTRANSFERASE 3A (DNMT3A) is mutated in hematological malignancies affecting myeloid, mixed, and lymphoid lineages and is associated with poor prognosis. Past studies in mice revealed Dnmt3a-KO HSCs had increased selfrenewal, but no leukemia was observed. Here, all lethally irradiated mice transplanted with Dnmt3a-deleted HSCs died within one year, most developing hematopoietic malignancies across a spectrum as observed in patients. In some cases, Dnmt3a-KO HSCs acquired secondary mutations similar to those identified in patients, further validating this model. In this study, loss of Dnmt3a led to disturbed methylation patterns unique to different diseases, rather than to a uniform methylation profile, pointing to lineage-specific methylation aberrations promoted by Dnmt3a loss. Global hypomethylation was observed in all of the malignancies but lymphoid malignancies also exhibited hypermethylation specifically at promoter regions. This mouse model underscores the important role of Dnmt3a in normal hematopoietic development, and demonstrates that Dnmt3a loss-of-function confers a preleukemic phenotype on murine HSCs. In patients, preleukemic stem cells with DNMT3A mutations are clinically silent, and outcompeted by malignant clones during disease presentation. These cells are reported to survive chemotherapy, and may contribute to increased rates of relapse. Our mouse model may prove valuable for developing targeted strategies for eliminating preleukemic cells for prevention and treatment of hematological malignancies in the future. Contributors: Mayle, Allison; Yang, Liubin; Rodriguez, Ben; Zhou, Ting; Chang, Edmund; Curry, Choladda V; Challen, Grant A; Li, Wei; Wheeler, David; Rebel, Vivienne; Goodell, Margaret 207 BAYLOR COLLEGE OF MEDICINE A MECHANISM FOR REPAIR OF BROKEN REPLICATION FORKS Ryan Mayle Department of Molecular & Human Genetics Advisor: Grzegorz Ira, Ph.D.-Department of Molecular & Human Genetics A major source of a spontaneous double stand break (DSB) in replicating cells is a single strand nick which is encountered by a replication fork, creating a single ended DSB break, commonly called a broken fork. Break-induced replication (BIR) is a pathway of homologous recombination known to repair single ended DSBs, and has been hypothesized to be responsible for repairing broken replication forks. However, BIR has been studied at DSBs outside the context of a replication fork, thus is remains unclear whether BIR has similar mechanistic and enzymatic requirements as the repair of broken replication forks. The hallmarks of BIR in yeast are its long duration, taking hours to complete, high mutagenicity, and a requirement of proteins which stimulate a D-loop migration mode of repair synthesis, including Pol32, the non-essential subunit of Polδ, and the helicase Pif1. We used a site-specific nick-induced DSB repair assay to study the process of broken replication fork repair, and compare this to BIR. By studying the genetic requirements and mutagenicity of broken fork repair at two distinct genomic loci, we can determine how closely this pathway resembles traditional BIR. We have placed nick sites between two origins as well as between the first origin and the telomere. The latter situation presents the cell with only one option for repair; synthesis primed from the single end break. We find that the role of BIR in repair at both break locations is minimal, as Pif1 and Pol32 are not required. However, two HR endonucleases, Mus81 and Yen1, are required for repair. This is consistent with repair mediated by cleavage of the D-loop formed at the break, leading to re-establishment of a normal, high fidelity, fork. In contrast to BIR, where mutagenesis remains high throughout repair, broken fork repair results in no increase in mutations away from the break. When Mus81 in absent, mutagenesis is elevated away from the break, and has similar characteristics to BIR. Therefore, we propose that Mus81 regulates repair pathway choice at broken forks by cleaving the D-loop, preventing BIR, and promoting repair utilizing a high fidelity fork. Contributors: Mayle, Ryan; Ira, Grzegorz 208 2014 GRADUATE STUDENT SYMPOSIUM DYSREGULATION OF PURINERGIC SIGNALING IN HEPATOCELLULAR CARCINOMA Janielle P Maynard Program in Translational Biology & Molecular Medicine Advisor: Sundararajah Thevananther, Ph.D.-Department of Pediatrics John Goss, M.D.-Department of Surgery Hepatocellular carcinoma (HCC) is the third most lethal cancer worldwide, but molecular mechanisms of its pathogenesis are not well understood. Recent studies suggest that extracellular ATP-mediated activation of P2Y2 purinergic receptor induces hepatocyte proliferation in response to partial hepatectomy and ATP treatment alone was sufficient to induce hepatocyte proliferation in vitro. The purpose of this study was to characterize extracellular nucleotide effects on HCC cell proliferation and to examine the role of P2 purinergic signaling in the pathogenesis of HCC in patients and Mst1/2-/-, a mouse model of HCC. Hypothesis: Dysregulation of purinergic signaling facilitates aberrant cell proliferation underlying hepatocellular carcinogenesis. Methods. HCC human-derived Huh7 cells, maintained in serum free media for 24h, were treated with ATP S, or ADP (100µM) for different time intervals. SP600125 pretreatment was used to inhibit c-Jun N-terminal Kinase (JNK) signaling. Western blotting, qRT-PCR and 5Bromo-2'-deoxy-uridine (BrdU) incorporation analysis were done. Mst1/2-/- and WT mouse livers (1, 3, & 6 months) and HCC patient livers (n=27) were analyzed by qRTPCR for all 15 P2 purinergic receptor isoforms. Results. Extracellular nucleotide treatment alone was sufficient to induce cell cycle progression in Huh7 cells, evidenced by increased BrdU incorporation and increased cyclin D3, E, and A mRNA and protein expression. We observed downregulation of cyclin D1 mRNA, however, as previously reported in a subset of HCC with high tumor grade. JNK inhibition attenuated nucleotide-induced cyclin D3, E and A protein expression, but enhanced downregulation of cyclin D1. Mst1/2-/- mouse tumors (at 3-6 months) exhibit dysregulated expression of multiple P2 purinergic receptor isoforms as compared to WT. In HCC patients, multiple P2 purinergic receptor isoforms were elevated ≥2-fold in liver tumors as compared to uninvolved areas in up to 52% of patients. P2 purinergic receptor upregulation was more prevalent among HCC patients infected with hepatitis C virus (HCV) (75%) as compared to non-viral groups (20%) identifying a unique subset of viralinduced HCC overexpressing P2 receptors. Conclusions. Our results suggest that extracellular nucleotides are potent mitogens in Huh7 cells, inducing downregulation of cyclin D1 and upregulation of cyclin E, which are associated with poor prognosis in HCC patients. Our analysis of HCC patient and Mst1/2-/- mice livers has uncovered a likely role for purinergic signaling in the pathogenesis of HCC, highlighting P2 purinergic receptors as potential biomarkers and novel therapeutic targets for HCC. Contributors: Johnson, Randy L.; Lee, Ju-Seog; Lopez-Terrada, Dolores; Goss, John A; Thevananther, Sundararajah 209 BAYLOR COLLEGE OF MEDICINE INVASIVE UTI ASSOCIATED WITH LOWER DIVERSITY OF BLADDER FLORA IN CLINICAL TRIAL OF BACTERIAL INTERFERENCE WITH E. COLI HU2117 Tyler Jordan McCue Integrative Program in Molecular and Biomedical Sciences Advisor: Joseph Petrosino, Ph.D.-Department of Molecular Virology & Microbiology Catheter-associated urinary tract infections (CAUTI) are most common infections in nursing homes and rank in the top three of hospital-acquired infections. There is currently no effective strategy in place to prevent CAUTI in long-term catheterized patients and with a continuing increase in the elderly population this problem will only grow in the coming years. We hypothesized that colonizing the bladder with a benign strain of E. coli (HU2117) isolated from a subject with asymptomatic bacteriuria would be a safe and effective means of using bacterial interference to prevent CAUTI in older, catheterized adults. Urine was collected longitudinally from 4 patients that were inoculated with HU2117 via receiving an indwelling catheter that had been pre-coated with E. coli HU2117. Aliquots of samples were monitored for cultivatable organisms for 28 days or until the loss of E. coli HU2117 using standard microbiological methods. Each of the 4 subjects had a different outcome: infection developed without HU2117 colonization, infection developed during HU2117 colonization, no infection or HU2117 colonization, and no infection during HU2117 colonization. In order to better understand the dynamics of the microbial communities in these patients’ bladders we extracted DNA and performed 16S gene targeted sequencing to identify the bacteria present. Analysis of the microbial communities present in the subject samples revealed a significant decrease in the diversity of bladder flora in subjects that developed an infection. It was found that colonization with E. coli HU2117 did not prevent infection and did not lead to any significant changes in bladder flora diversity. These results suggest that microbial diversity may play a protective role against invasive infection of the catheterized bladder. Contributors: McCue, Tyler; Horwitz, Deborah; Mapes, Abigail C.; Ajami, Nadim J.; Petrosino, Joseph F.; Trautner, Barbara W. 210 2014 GRADUATE STUDENT SYMPOSIUM γδ T CELLS PROMOTE OBESITY-INDUCED INFLAMMATION AND INSULIN RESISTANCE IN MICE Pooja Chetan Mehta Department of Pathology & Immunology Advisor: C Smith, M.D.-Department of Pediatrics γδ T cells are resident in adipose tissue and increase during diet-induced obesity. Their possible contribution to the inflammatory response that accompanies diet-induced obesity was investigated in mice after a 5-10 week high milk fat diet (HFD). The HFD resulted in significant increases in CD44hi, CD62Llo and TNF-α+ γδ T cells in epididymal adipose tissue (eAT) of wildtype (WT) mice. Mice deficient in all γδ T cells (TCRδ-/-) or only Vγ4 and Vγ6 subsets (Vγ4/6-/-) were compared with wildtype mice with regard to pro-inflammatory cytokine production and macrophage accumulation in eAT. Obesity among these mouse strains did not differ, but obese TCRδ-/- and Vγ4/6-/mice had significantly reduced eAT expression of F4/80, a macrophage marker, and inflammatory mediators CCL2 and IL-6 compared to WT mice. Obese TCRδ-/- mice had significantly reduced CD11c+ and TNF-α+ macrophage accumulation in eAT after 5 and 10 weeks on the HFD, and obese Vγ4/6-/- mice had significantly increased CD206+ macrophages in eAT after 5 weeks on the diet and significantly reduced macrophages after 10 weeks. Obese TCRδ-/- mice had significant reductions in systemic insulin resistance and inflammation in liver and skeletal muscle after longer-term HFD feeding (10 and 24 weeks). In vitro studies revealed that isolated γδ T cells directly stimulated RAW264.7 macrophage TNF-α expression, but did not stimulate inflammatory mediator expression in 3T3-L1 adipocytes. These findings are consistent with a role for γδ T cells in the pro-inflammatory response that accompanies diet-induced obesity. Contributors: Mehta, Pooja ; Smith, C. Wayne 211 BAYLOR COLLEGE OF MEDICINE EVOLUTION OF ANTIBIOTIC RESISTANCE AMONG CLINICAL VARIANTS OF KPC β-LACTAMASE Shrenik Chetan Mehta Department of Pharmacology Advisor: Timothy Palzkill, Ph.D.-Department of Pharmacology β-lactam antibiotics are the most commonly prescribed class of drugs for treating bacterial infections. β-lactamase enzymes are the most important source of resistance to these drugs as they hydrolyze the antibiotic and render it inactive. The rapid dissemination and evolution of these enzymes has resulted in many classes of βlactamases capable of hydrolyzing a broad range of antibiotic substrates. The KPC group of β-lactamases is one of the most threatening groups of enzymes as they are capable of hydrolyzing the carbapenem class of β-lactam antibiotics. The carbapenems currently serve as our last line of defense against bacterial infections. Variants of KPC β-lactamases differing by one to two amino acids have been isolated from patients all over the world. Despite the identification of these enzyme variants from the patients, there is a lack of information about their substrate profiles and catalytic mechanism, which could help in deciding treatment regimens for such infections. In this study, we have characterized nine KPC variants that differ from KPC-2 (pseudo wild-type) by one to two amino acids. The results indicate that both single and double mutant enzymes hydrolyze the carbapenem substrates as proficiently as KPC-2. When tested for ceftazidime, a 3rd generation cephalosporin antibiotic, the bacterial cells expressing the single mutants showed a minimum inhibitory concentration (MIC) up to 6-fold higher than KPC-2 while the bacterial cells expressing the double mutants showed a 3 to 100 fold increase in MIC. The purified double mutant enzymes also exhibited a 7 to 75-fold increase in catalytic efficiency for hydrolyzing ceftazidime while the single mutants had a catalytic efficiency 7 to 11-fold higher than KPC-2. 3rd generation cephalosporins are the drug of choice for treating unknown bacterial infections in patients; thus, the evolution of KPC to better hydrolyze ceftazidime is likely the result of selective pressure due to antibiotic therapy. In addition, this improvement in ability to hydrolyze ceftazidime does not significantly alter their ability to hydrolyze carbapenems. Thus, mutations acquired by KPC-2 in natural isolates of bacteria significantly broaden its substrate profile leading to multi-drug resistance. Contributors: Mehta, Shrenik; Kacie Rice; Palzkill Timothy 212 2014 GRADUATE STUDENT SYMPOSIUM DEFICIENCY OF MECP2 IN GLUTAMATERGIC NEURONS LEADS TO DISTINCT FEATURES COMPARED TO GABAERGIC CONDITIONAL DELETION Xiangling Meng Department of Neuroscience Advisor: Huda Zoghbi, M.D.-Department of Pediatrics Background: Rett Syndrome (RTT) is a postnatal neurological disorder caused by loss of function mutations in the gene encoding methyl-CpG-binding protein 2 (MeCP2). Deleting Mecp2 only from brain tissue at embryonic day 12 leads to phenotypes identical to those of the null mutation, indicating that loss of MeCP2 from the CNS is responsible for the RTT phenotypes. Deletion of Mecp2 only from inhibitory GABAergic neurons recapitulates many RTT phenotypes including the stereotypies and altered social interaction, but does not replicate anxiety-like behaviors and tremor. The role that excitatory glutamatergic neurons play in the pathogenesis of RTT has not been explored in detail. Method: We conditionally deleted Mecp2 from glutamatergic neurons in the mouse brain using a vGlut2-Cre line, and characterized the mice by a comprehensive battery of behavioral tests as well as neurophysiological methods. Results: The glutamatergic conditional knockout mice (CKO) became obese, and developed impaired acoustic startle and motor deficits. Interestingly, unlike the GABAergic CKO, the glutamatergic CKO showed anxiety-like behaviors as early as 5 weeks of age, and developed severe tremor. Furthermore, they died early with half of them dead by 10 weeks. These phenotypes are identical to the disease progression pattern of the Mecp2 null mutation. The glutamatergic CKO mice also showed deficits in conditional fear memory and impairment of excitatory synaptic plasticity measured by hippocampal CA1 long-term potentiation. Conclusion: These data demonstrate that dysfunction of MeCP2 in excitatory glutamatergic neurons contributes to numerous neuropsychiatric phenotypes. Especially, it drives the onset of anxiety-like behaviors, tremor, and obesity in RTT, indicating an excitatory neuron-dependent mechanism underlying these phenotypes of Rett syndrome. Contributors: Meng, Xiangling; Lu, Hui; Wang, Wei; Chen, Hongmei; Zoghbi, Huda. 213 BAYLOR COLLEGE OF MEDICINE THE STRINGENT STRESS RESPONSE IS REQUIRED FOR MUTAGENIC REPAIR OF DNA BREAKS Phillip Jay Minnick Department of Biochemistry & Molecular Biology Advisor: Susan Rosenberg, Ph.D.-Department of Molecular & Human Genetics Mutagenesis mechanisms controlled by stress responses increase mutation rates during stress, potentially accelerating adaptation to stressors such as nutrient limitation, antibiotics, antifungal drugs and hypoxia, in bacteria, yeast and human cancer cells. Inhibiting these mechanisms could provide new ways to combat cancer and infectious disease. One stress-induced mutagenesis mechanism in Escherichia coli is mutagenic repair of DNA double-strand breaks (DSBs). Under stress, or if the general (RpoS) stress response is activated artificially, repair of DSBs switches from use of the high fidelity DNA Pol III to use of error-prone Pol IV causing base-substitution and indel (“point”) mutations, or use of Pol I causing copy-number variations. Mutagenic repair of DSBs localizes new mutations both in time (under stress), and in genomic space (near DSBs). Three stress responses promote mutagenic break repair in E. coli: the envelope-protein stress response promotes break formation; the DNA-damage response upregulates Pol IV (promoting point mutagenesis); and the general stress response licenses use of Pols IV, II and I in repair. Here we demonstrate that a fourth stress response must be activated for mutagenic break repair to occur: the stringent (starvation) response. We show that stringent-response-defective ∆relA or ∆dksA cells are defective for both point mutations and gene amplification. Moreover, the stringent response promotes mutagenesis independently of the other three stress responses. First, DksA is still required if DSBs are given artificially with I-SceI enzyme. Thus, the stringent response role is not in formation of DSBs (role of the envelope-protein-stress response). Second, the RelA/DksA role is not in DNA damage-response induction because it is not suppressed by expression of Pol IV(the sole role of the damage response). Third, mutations that allow general-stress-response-independent mutagenesis do not compensate for RelA/DksA, demonstrating a role independent of the general stress response. We conclude that the stringent response constitutes a fourth, independent, stress response input into mutagenic break repair. The stringent response presumably regulates the expression of a currently unknown protein(s), critical to mutagenic break repair. Our data imply that cells regulate mutagenesis very carefully, requiring four independent stress response inputs to unleash their mutagenesis “program”. Contributors: Frisch, Ryan; Gibson, Janet; Terwiliger, Austen; McCue, Tyler; Darrow, Michele; Herman, Christophe; Rosenberg, Susan 214 2014 GRADUATE STUDENT SYMPOSIUM THE EFFECT OF HEPATITIS B VIRUS HBX PROTEIN ON DDB1 FUNCTION Marissa Mie Kehaulani Minor Department of Molecular Virology & Microbiology Advisor: Betty Slagle, Ph.D.-Department of Molecular Virology & Microbiology Chronic Hepatitis B virus (HBV) affects over 400 million people worldwide and is a major risk factor for the development of hepatocellular carcinoma (HCC). Our laboratory studies the HBV regulatory protein HBx, which is required for virus replication in infection models. We and others have demonstrated that an interaction between HBx and cellular damaged DNA binding protein 1 (DDB1) is critical for virus replication. DDB1 functions as an adapter to the Cullin 4A E3 ubiquitin ligase complex. DDB1 recruits DDB1 Cullin Associated Binding Factors (DCAFs) to the Cullin 4A E3 ubiquitin ligase complex, and the DCAFs in turn recruit substrates to be ubiquitinated and degraded. Each of the ~90 human DCAFs share a 16 amino acid DDB1-binding WD40 motif. The HBx contains a sequence similar to the DDB1-binding WD40 motif and has been shown through crystal structure to interact directly with DDB1, suggesting that it is a viral DCAF. The DDB1 function affected by HBx binding is unknown and there are few biologically relevant systems to study this interaction. We will be using human liver chimeric mice (immunodeficient mice that have been engrafted with human hepatocytes) as an infection model in which to study the HBx-DDB1 interaction. We hypothesize that HBx binding to DDB1 may alter the DCAF profile, and that this will lead to changes in the array of cellular factors that are ubiquitinated and degraded, thereby creating a cellular environment that benefits virus replication. Aim 1: The first aim of this project will determine the DCAF profiles of normal, uninfected livers as well as HBV-infected livers and comparing the two to determine the impact of HBV replication. This will be done using human liver chimeric mice of the same hepatocyte donor. Immunoprecipitation/mass spectrometry will be used to identify DCAFs that are bound to DDB1 in both infected and uninfected hepatocytes. Aim 2: The second aim of this project will determine the impact of the HBx-DDB1 interaction on DDB1 functions such as the DNA damage response and innate immunity. We will analyze uninfected and HBV-infected liver tissue for the DNA damage marker γH2AX to determine if the DNA damage response is being activated during HBV infection in vivo. We will also be using a variety of methods to analyze how HBV may interact with DDB1 in order to dampen the immune response. Contributors: Minor, Marissa 215 BAYLOR COLLEGE OF MEDICINE THE STRUCTURE AND FUNCTION OF MICROTUBULE ASSOCIATED PROTEIN TAU IN ALZHEIMER’S DISEASE AND OTHER TAUOPATHIES Patrick Gerald Mitchell Integrative Program in Molecular and Biomedical Sciences Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology Microtubule associated protein tau (MAPT, or tau) is implicated as an associated factor in the pathology of Alzheimer’s Disease (AD), as well as several other neurodegenerative disorders, collectively referred to as tauopathies. The disease associated form of tau aggregates in paired-helical filaments (PHF), which interfere with tau’s ability to bind and stabilize microtubules, and cause higher-order intracellular tau aggregate formation. These aggregates, or neurofibrillary tangles (NFT’s), are a common feature of neurological diseases characterized by neuronal dysfunction and eventual cell death. While it is well supported that pathologically modified tau and tau aggregates can play a role in neurodegeneration, the specific neurotoxic species, and the mechanisms underlying their pathology, remain unclear. Although tau has been the subject of increasing interest in the Alzheimer’s Disease research community, a comprehensive structural study of tau aggregates has yet to be performed. Recent technological advances in cryo-electron microscopy (cryoEM) allow for the elucidation of structural features of tau fibrils at a resolution that has not previously been attainable using this technique. A detailed structural study can provide insight into the important differences between wild-type tau and aggregation prone mutant tau. In addition to high-resolution features of the tau fibril, the role of tau aggregation within the cell is also an important consideration in our understanding of tau pathology. Eukaryotic cells have been conventionally considered to exceed the maximum specimen thickness for investigation by cryo-EM. However, as freezing techniques and imaging technologies have improved, it has been demonstrated that cryo-EM is a useful technique for imaging of some eukaryotic cells, especially in thin cellular projections, such as neurites. As studies of larger specimens have become more common in the EM community, a hybrid technique known as correlative light and electron microscopy (CLEM) has emerged as a useful bridge between light microscopy, and the molecular resolution of EM. This technique will be useful to visualize the structural characteristics of tauopathies, as well as the mechanistic action of potential treatments. Contributors: Mitchell, Patrick; Li, Hongmei; Martini-Stoica, Heidi; Dai, Wei; Zheng, Hui; Chiu, Wah 216 2014 GRADUATE STUDENT SYMPOSIUM STRUCTURAL CHARACTERIZATION OF INFLUENZA A VIRUS NON-STRUCTURAL PROTEIN-1 (NS1) BINDING TO CPSF30 Sayantan Mitra Department of Biochemistry & Molecular Biology Advisor: B Prasad, Ph.D.-Department of Biochemistry & Molecular Biology Influenza viruses cause acute highly contagious respiratory disease and affect 500 million people annually worldwide. Currently the highly lethal H5N1 strain posing a serious threat for worldwide pandemic, along with recent emergence of influenza virus strains resistant to the available antiviral drugs make it necessary to identify potential new drug targets. One of the promising targets is the non-structural protein 1 (NS1) of influenza virus, which is important for virus replication, spread, and pathogenesis. It is a potent antagonist of interferon-mediated host antiviral response. It consists of an RNA binding domain (RBD) and an effector domain (ED) separated by a flexible linker. It interacts with the F2F3 domain of cleavage and polyadenylation specificity factor (CPSF30) to inhibit 3’ end processing of cellular pre-mRNA including IFN-β pre-mRNA, thus inhibiting host antiviral immune response. Although the structure of NS1 ED with F2F3 has been determined, the structural characteristics of the full-length (FL) NS1 suggest these structures might not be relevant. To determine the structure of H5N1 FLNS1 and F2F3 by X-ray crystallography, both the proteins have been purified individually and after forming complex, it has been used for initial crystallographic screening. Currently, optimization of the favorable conditions are been carried out. The binding studies of H5N1 FL-NS1 and H5N1 NS1-ED with F2F3 by Isothermal titration calorimetry (ITC) have also suggested FL-NS1 interacts with F2F3 more strongly than ED alone. This might be due to a different and/or additional set of interactions that allows for increased F2F3 binding affinity with theFL-NS1 than with ED alone. . In addition to providing a better mechanistic understanding of how FL-NS1 interacts with F2F3 and what are the characteristics of these interacting regions, they will also provide a rational basis for the design and development of new small molecule drugs that disrupt NS1 interaction with F2F3 and thus the ability of NS1 to antagonize the host antiviral response. Contributors: Mitra, Sayantan; Prasad, BVV 217 BAYLOR COLLEGE OF MEDICINE EPITHELIAL, STROMAL, AND SYSTEMIC PTCH1 ARE CRITICAL FOR MAMMARY GLAND DEVELOPMENT Teresa Monkkonen Department of Molecular & Cellular Biology Advisor: Michael Lewis, Ph.D.-Department of Molecular & Cellular Biology Hedgehog signaling mediates organogenesis, and stem cell function in mammals, and often is misregulated in cancers, including breast. Breast lesions show decreased expression of Patched 1 (Ptch1), an inhibitor of signaling, and increased expression of Smoothened (Smo), an activator of signaling. Previous studies using mice homozygous for a hypomorphic Ptch1 allele showed stunted, dysplastic ducts, while mice heterozygous for a null allele showed hyperproliferative, hyperplastic mammary glands with filled-in ducts. Transplantation experiments with these mutants showed a requirement for epithelial, stromal, and systemic Ptch1; but the true Ptch1 loss-offunction phenotype in each compartment is unknown. We hypothesize that Ptch1 mediated inhibition of canonical hedgehog signaling has discrete epithelial, stromal, and systemic functions to regulate mammary gland morphogenesis, histology, and proliferation. To test this hypothesis, we performed tissue compartment-specific deletion of Ptch1. Outgrowths from transplanted Adenovirus-Cre treated, conditional null Ptch1fl/fl mammary epithelial cells yielded hyperbranched, hyperproliferative mammary ducts. Stromal loss of Ptch1 via Fsp-Cre deletion in fibroblasts and immune cells produced different phenotypes. Heterozygotes for the null allele were hyperbranched and hyperproliferative. Homozygous null Fsp-Cre, Ptch1fl/fl animals showed stunted ductal outgrowths and loss of proliferation in mature ducts and terminal end buds. These animals recapitulated the filled-in duct phenotype of Ptch1 +/- animals, indicating this phenotype was due to a stromal function of Ptch1. Both stromal mutants had aberrant microlumen formation and multiple layers of luminal cells. Interestingly, Fsp-Cre, Ptch1fl/fl animals had increased ER expression, but complete loss of PR at 8 weeks. Whole gland transplantation rescued the stunted duct and loss of side branching phenotypes, but filled-in ducts and microlumens were still present. In summary, we have shown that epithelial Ptch1 is required to suppress branching and proliferation. Ptch1 in a systemic Fsp positive cell promotes branching and proliferation; and Ptch1 in the mammary stroma regulates ductal histomorphology. These data underscore the importance of tissue-tissue crosstalk in the mammary gland. Contributors: Monkkonen, Teresa; Landua, John; Visbal, Adriana; and Lewis, Michael T. 218 2014 GRADUATE STUDENT SYMPOSIUM LOW LEVEL PROLIFERATION DURING PHYSIOLOGICAL CARDIAC HYPERTROPHY IS MODULATED BY HIPPO SIGNALING Tanner Oliver Monroe Department of Molecular Physiology & Biophysics Advisor: James Martin, M.D./Ph.D.-Department of Molecular Physiology & Biophysics George Rodney, Ph.D.-Department of Molecular Physiology & Biophysics Traditionally, cardiac hypertrophy was thought to be solely attributed to growth of the resident cardiomyocytes (CM). However, recent reports indicate an increased level of cell cycle re-entry and proliferation of resident cardiomyocytes during physiological hypertrophy. The Hippo pathway is an evolutionarily-conserved negative regulator of organ growth. In the heart, it activates during late development, facilitating cell cycle exit, which restricts CM proliferation, thereby controlling the final CM number. Further growth of the organ is attributed to hypertrophy of the individual myocytes. There are three main causes of post-natal hypertrophy: exercise, pregnancy (physiological), and chronic pressure overload (pathological). We believe that during physiological hypertrophy, where an increase in CM division has been reported, a set of CMs within the heart re-enter the cell cycle due to the down-regulation of Hippo pathway kinases, in a crosstalk with the Wnt-Akt signaling axis. Our data, and that of publicly available data sets indicate that cell-cycle related genes controlled by the Hippo pathway nuclear effector, Yap, are up-regulated during physiological hypertrophy. In contrast, during pathological hypertrophy, up-regulation of cell cycle-related transcripts is absent. Steady-state CM renewal is thought to be critically important to maintenance of the health of the organ, with baseline CM replenishment approximately 1% of the cells per year, diminishing progressively during aging. If our findings are genuine, we believe we have found a natural way to increase CM renewal, as well as a previously unreported mechanism for post-developmental heart growth. Further, we will gain valuable knowledge about the differences that separate pathologic and physiological hypertrophy, with the aim of targeting these pathways therapeutically. Contributors: Monroe, Tanner; Zhang, Min; Heallen, Todd; Leach, John; Rodney, George; Martin, James 219 BAYLOR COLLEGE OF MEDICINE REACTIVE OXYGEN SPECIES ARE REQUIRED FOR ENVIRONMENTAL STRESSINDUCED MUTAGENESIS IN STARVING ESCHERICHIA COLI CELLS Jessica M Moore Department of Biochemistry & Molecular Biology Advisor: Susan Rosenberg, Ph.D.-Department of Molecular & Human Genetics Philip Hastings, Ph.D.-Department of Molecular & Human Genetics E. coli under stress such as starvation increase mutation rate transiently under the control of stress responses, until a mutation occurs that allows adaptation. The mechanism of stress response-controlled switch to mutagenic repair of DNA breaks is well characterized. lac frameshift-bearing cells starved on lactose medium acquire compensating frameshift (“point”) mutations, or amplifications of the leaky lac allele to 20-50 copies, which confers sufficient enzyme activity for growth. The mutagenesis requires (spontaneous) double-stranded DNA breaks and their repair. We show that reactive oxygen species, specifically persistent oxidative lesions in DNA that result from them, are required for mutation formation under stress in E. coli. Addition of exogenous reducing agents, use of constitutively activating alleles for the oxidative damage responses, and over-expression of katG to scavenge hydroxyl radicals dramatically reduce mutation formation. We show that reactive oxygen species are not required in stress-induced mutation to oxidize proteins, but to form oxidative lesions in DNA. These lesions are not required for the formation of DNA double-stranded breaks or to titrate out mismatch repair components, and the effect on mutation is lost if lesions are excised from DNA. Together, these data show a novel requirement for unrepaired oxidative base changes in DNA for the formation of environmental stress-induced mutants separately from the requirement for DNA double-stranded breaks and stress response regulation. We suggest that lesions in DNA pause the replicative DNA polymerase, allowing error-prone translesion polymerases to become active, leading to mutation formation. Contributors: Moore, JM; Mojica AK; Rosenberg SM; Hastings PJ 220 2014 GRADUATE STUDENT SYMPOSIUM HISTIDINE METABOLISM BY LACTOBACILLUS REUTERI Christina Narie Morra Integrative Program in Molecular and Biomedical Sciences Advisor: James Versalovic, M.D./Ph.D.-Department of Pathology & Immunology Amino acid metabolism by the gut microbiome is critical for human health. Commensal bacteria, such as Lactobacillus reuteri, produce biologically active compounds that are capable of modulating host immunity. The decarboxylation of Lhistidine to histamine by HdcA in L. reuteri reduces the production of the proinflammatory molecule, TNF. Currently is it unknown whether metabolism of histidine by L. reuteri affects its growth or whether L. reuteri produces other host-modulating products. Using the stable isotope, 13C615N3 L-histidine in bacterial media we have begun to elucidate histidine metabolism by L. reuteri ATCC 6475 and identify novel histidine metabolism pathways. Analysis of 72 hr growth curves of wild type and two HdcA L. reuteri mutants in defined (LDMIV), semi-defined (LDMIII) and rich (MRS) media revealed no significant changes in growth or viability. This indicates that histidine metabolism is not essential for L. reuteri growth and viability. To assess the mechanism of histidine metabolism by L. reuteri, the semi-defined, LDMIII medium was supplemented with the heavy isotope, 13C615N3 L-histidine and analyzed by mass spectroscopy. A preliminary mass spectrometry screen for metabolites determined that the addition of carnosine (β-alanyl-L-histidine) in the medium significantly increases histamine production. Together this data has demonstrated that histidine metabolism, although providing benefit to the host, is not required for L. reuteri growth and that the addition of carnosine improves histamine production in vitro. Future work will involve coculturing of L. reuteri with human ileal enteroids to determine the effects of L. reuteri and histamine metabolism on the human intestinal epithelium. Contributors: Morra, Christina; Versalovic, James 221 BAYLOR COLLEGE OF MEDICINE EMAN2.1 – A NEW GENERATION OF SOFTWARE FOR VALIDATED SINGLE PARTICLE ANALYSIS AND SINGLE PARTICLE TOMOGRAPHY Stephen Christopher Murray Program in Structural and Computational Biology and Molecular Biophysics Advisor: Steven Ludtke, Ph.D.-Department of Biochemistry & Molecular Biology Over the last three years, the Cryo-EM community has begun shifting focus from improving resolution, to validation of results. EMAN2.1 represents the results of major new developments in EMAN2’s single particle reconstruction and single particle tomography workflows. The new tools such as e2refine-easy, integrate “gold standard” resolution assessment into the refinement process along with a number of new optimizations, both speeding the refinement process, and eliminating the need for empirical filtration of reconstructions by users. EMAN2.1 also integrates support for tiltpair validation and “true resolution” testing to insure self-consistency among data and final 3-D maps. This is one of the few methods which can identify incorrect maps at low resolution. Another approach often applied to increase confidence in a 3-D structure is to reprocess the same data using multiple algorithms, preferably based on different mathematical methods. EMAN2.1 includes an interface for converting data and metadata into the appropriate format for reprocessing in Relion[2] or FreAlign[3], two alternative single particle reconstruction packages. Once these packages complete their refinements, the results can be imported back into EMAN2.1 for comparison and analysis. As an alternative, EMAN2.1 can perform the opposite process as well. A refinement completed originally in one of these other packages can be converted into an EMAN2.1 project, which can then be used to re-refine the data from scratch. Another important area of improvement is single particle tomography. Rather than the traditional approach of reconstructing large numbers of 2-D images of identical particles in random orientations, in single particle tomography, tomographic data is collected for fields of particles, producing a low resolution and incomplete, but 3-D reconstruction for each individual molecule. EMAN2.1 now incorporates tools for subtomogram extraction, and a variety of different approaches for alignement and averaging of particles. This approach is a powerful alternative to single particle analysis particularly in cases where the particles are flexible or heterogeneous in solution Contributors: Murray, Stephen; Galaz-Montoya, Jesus; Tang, Grant; Flanagan IV, John; Ludtke, Steve 222 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF PRENATAL EXPOSURES IN MODULATION OF NEURODEVELOPMENT USING THE SHANK3e4-9/+ MUTANT MOUSE MODEL Jaclyn Nicole Murry Department of Molecular & Human Genetics Advisor: Ignatia Van Den Veyver, M.D.-Department of Obstetrics & Gynecology Autism spectrum disorder (ASD) comprises a set of neurodevelopmental conditions manifesting as repetitive behaviors and social and communication impairments. Genes affecting synaptic function have been found to be causative in a subset of ASD patients. Early exposure to environmental factors either by themselves or in conjunction with genetic factors has also been implicated in ASD, and in combination may play a role in ASD penetrance. To minimize confounding factors, our lab uses mouse models to answer this question. The overall goal of my project is to investigate how gene and prenatal environment interact (GXE) to influence ASD-like behavior in a Shank3 mutant mouse model. I chose the Shank3 null mice based on their ASD-like behaviors. Furthermore, deletions in SHANK3 cause PhelanMcDermid Syndrome, whereas mutations cause isolated ASD. Our overarching hypothesis is that ASD-associated risk alleles combined with certain common prenatal exposures can modulate neurodevelopment, altering maternal care and resulting in offspring behavior disturbances. This combination might cause an individual to reach a “phenotypic threshold” resulting in increased ASD penetrance. I will address this hypothesis through two specific aims. SPECIFIC AIM 1: To assess the effects of prenatal CUMS exposure on maternal care in Shank3 mutant dams. Preliminary data from our lab indicates that mutant male offspring of HET Shank3 dams exposed to gestational chronic unpredictable mild stressors (CUMS) exhibit an atypical response when compared to non-stressed gender and genotype-matched littermates in the three-chamber social behavior test. Because studies indicate that gestational stress can alter postpartum maternal care and offspring development, I am examining the effects of gestational stress on maternal care in WT and mutant Shank3 dams. Proposed experiments will likely answer how stress combined with genetic susceptibility contributes to observed phenotypes. SPECIFIC AIM 2: To assess the effects of prenatal FLX exposure of Shank3 HET dams on offspring neurodevelopment. For the second aim, I am investigating if other prenatal stressor-genotype combinations will result in altered social behavior in offspring. I am focusing on Fluoxetine (FLX), a Selective Serotonin Reuptake Inhibitor, regularly prescribed during pregnancy, and because maternal intake of (FLX) in the first trimester is associated with increased incidence of ASD. Therefore, I will examine whether FLX exposure in Shank3-mutant dams affects penetrance of ASD features in offspring through behavioral assessments. Postbattery, offspring brains are being harvested and processed for brain morphology, histology, gene and protein expression. In doing so, we will be assessing whether these factors exhibit additive defects. Proposed experiments will likely answer whether prenatal drug exposure combined with genetic susceptibility can play a role in ASD penetrance. Contributors: Murry, Jaclyn; He, Fang; Engler, David; Balasa, Alfred; Van den Veyver, Ignatia 223 BAYLOR COLLEGE OF MEDICINE THE CONTRIBUTIONS OF ADHERENS JUNCTIONS IN LUMINAL BREAST CANCER BONE METASTASIS Aaron M Muscarella Integrative Program in Molecular and Biomedical Sciences Advisor: Xiang Zhang, Ph.D.-Department of Molecular & Cellular Biology Background: Most deaths from breast cancer are not due to the primary tumor but from metastasis to secondary sites, the most common of which is the bone. The Zhang lab recently founds that disseminated tumor cells (DTCs) that arrive in the bone and progress into multi-cell micrometastases are almost always in proximity to, and in direct contact with, Osteoblasts, pre-osteoblasts, and their precursor mesenchymal stem cells (MSCs). It was also shown through in vitro co-culturing and in vivo xenograft methods that the breast cancer cells receive proliferative advantages and activation of mTOR signaling from these osteogenic cells, and that this relationship is dependent on direct contact, calcium, and E-cadherin. This was especially curious as MSCs are not known to express E-cadherin, only N- and OB-cadherins, and heterotypic cadherin adherens junctions (AJs) are rarely described in the literature. Little has been established about the initial, pre-clinical colonization of the bone by single DTCs. If we are able to learn more about the molecular mechanisms of this niche, there is potential for developing therapeutic strategies against their progression into fully pathogenic bone metastases. Experimental design and methods: I performed coated plate assays, where tissue culture plates were coated with IgG control or E-, N-, or OB- cadherin fusion proteins. I identified candidate Receptor Tyrosine Kinases (RTKs) that could activate mTOR signaling in the micrometastatic lesions using a phospho-tyrosine array, and explored these candidates using qPCR, western blot, and inhibitor treatments. In addition, I prepared CRISPR knockout constructs and shRNA constructs for AJ components and these RTKs to use in functional assays. Results: Cancer cell lines with strong E-cadherin expression proliferated selectively more on N-cadherin coated surfaces, and cell lines without strong E-cadherin expression grew selectively better on OB-cadherin coated surfaces. Furthermore, it has been demonstrated that E-cadherinexpressing cells form heterotypic E:N-cadherin AJs both in vitro using a pull-down assay, as well as in vivo using immunohistochemistry and the blocking E- and Ncadherin. Western blot indicated that both ALK and TrkA are not expressed by our luminal models, eliminating them as candidate RTK’s, while Ros1 inhibitor Foretinib was effective at inhibiting growth in cells in contact with MSCs in vitro. Conclusion: Luminal, E-cadherin expressing breast cancer cells form heterotypic, E:N-cadherin adhesion junctions in the bone, and relationship activated mTOR signaling through Ros1 or some other receptor on the cancer cell surface. Contributors: Muscarella, Aaron; Wang, Hai; Yu, Cuijua; Zhang, Xiang H.F. 224 2014 GRADUATE STUDENT SYMPOSIUM CONFINED DNA THERMODYNAMICS: STRUCTURE, PRESSURE, ELASTICITY Christopher Gary Myers Program in Structural and Computational Biology and Molecular Biophysics Advisor: B. Pettitt, Ph.D.-Biochemistry We have shown that excluded volume, electrostatic forces and surface-induced correlations are sufficient to predict most of the major features of the current structural data of DNA packaged within viral capsids without assuming any elastic conformational ordering. Current models assume highly-ordered, even spooled conformations, based on interpretation of cryo-EM density maps. We have shown that surface-induced ordering of unconnected DNA polymer segments is the only necessary ingredient in creating ringed densities consistent with experimental density maps. This implies that the ensemble of possible conformations of polymeric DNA within the capsid consistent with cryo-EM data may be much larger than implied by the traditional interpretation that such rings indicate a highly-ordered spool conformation. This suggests a more disordered, entropically-driven view of phage packaging may be possible. We have also shown the electrostatics of the DNA contributes a large portion of the internal hydrostatic and osmotic pressures of a phage virion, suggesting that nonlinear elastic anomalies might be responsible reduction of overall elastic bending energies, allowing more disordered conformations to be free-energetically favorable. Currently MC path sampling techniques, phage genomic data, and sequencedependent DNA elasticity predictions are being employed to accurately model the elastic bending contributions. Biophysical prediction of the pressure and structures of DNA confined within phage capsids will aid prediction of DNA expression in vivo, as well as the design of phage sequence-delivery methods. Contributors: Myers CG1,2, Pettitt BM1,2 225 BAYLOR COLLEGE OF MEDICINE PTPN12 TUMOR SUPPRESSOR IN BREAST CANCER: FROM GENETIC SCREENS TO NOVEL INSIGHTS FOR THERAPEUTIC INTERVENTION IN BREAST CANCER Amritha Nair Department of Molecular & Human Genetics Advisor: Thomas Westbrook, Ph.D.-Department of Biochemistry & Molecular Biology Background:Tyrosine kinases are known oncogenic drivers of cancer. The role of the antagonizing enzymes namely the Tyrosine Phosphatases has been less characterized. Our lab identified PTPN12, a tyrosine phosphatase, to be a bonafide tumor suppressor in breast cancer. It is frequently inactivated in Triple negative breast cancer [TNBC] and negatively regulates several oncogenic TKs. Furthermore the tumorigenic potential of PTPN12- deficient breast cancer cells is severely impaired upon restoration of PTPN12 function. This suggests that loss of the tumor suppressor PTPN12 drives cooperative TK activation in many breast cancers and that identifying these PTPN12-regulated kinases and further perturbing them in combination might ameliorate these cancers. Experimental design and methods: Using multiple orthogonal genetic and biochemical approaches we chose to identify the PTPN12-regulated kinome. We tested the role of PTPN12-regulated kinases in TNBC proliferation and identified combinatorial TKi treatments that impair survival of these cancers in vitro. We further utilized comprehensive testing of xenografts and patient-derived tumorgrafts [PDXs] models to validate our pharmacological findings in vivo. Results: Using a confluence of techniques including BiFC, RTK arrays and IP we have identified a narrow number of oncogenic RTKs that are regulated by PTPN12, including PDGFRB, MET, NTRK1, NTRK2, ALK and others. We report that PTPN12 sensitive breast cancers respond in vitro to TK inhibition using a combination of the drugs Sunitinib & Crizotinib that together inhibit a broad spectrum of the PTPN12regulated kinome. Due to the frequency of PTPN12 loss in TNBC we have expanded our pre-clinical testing to rigorously test the efficacy of this combination on a panel of 16 PDXs. Our studies show that PDXs with low levels of PTPN12 [determined by IHC] seem to correlate with better response to the drug combination. Conclusion: This study clarifies a paradigm wherein cancers that lack conventional biomarkers for TK dependency like amplifications/mutations, could still be responsive to combination Tki therapy due to loss of a key regulator of tyrosine kinase signaling and activation like PTPN12. Contributors: Nair, Amritha1; Sun, Tingting2; Tyagi, Siddhartha2; Dominguez-Vidana, Rocio3; Dobrolecki, Lacey.E. 4 ; Petrovic, Ivana 4 ; Schmitt, Earlene 3; Osborne, C.Kent 4 ; Schiff, Rachel 4 ; Shaw, Chad.A. 1; Lewis, Michael.T. 4; Westbrook,Thomas.F. 1,2,3,4. 1Department of Molecular and Human Genetics, Baylor College of Medicine, TX. 2The Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX. 3Interdepartmental Graduate Program in Cell and Molecular Biology, Baylor College of Medicine, Houston, TX, 4Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX. 226 2014 GRADUATE STUDENT SYMPOSIUM EFFECTS OF ACTIVIN SIGNALING ON ADIPOSE TISSUE FUNCTION AND ENERGY METABOLISM Maria Namwanje Department of Molecular & Human Genetics Advisor: Chester Brown, M.D./Ph.D.-Department of Molecular & Human Genetics Obesity is a growing epidemic worldwide; including the United States, where 35% of adults are classified as obese. Several members of the TGF-beta superfamily play important roles in body composition, adiposity and energy metabolism, including protection from diet-induced obesity. Activins (A and (B are TGF-beta family members with demonstrated roles in adipocyte differentiation and function. Activin (A inhibits adipocyte differentiation of 3T3-L1 cells and human preadipocytes by decreasing the expression of C/EBP(, which results in increased preadipocyte proliferation. Increased expression of activin (B in mice and humans is associated with obesity. Additionally, activin (B decreases lipolysis and down-regulates expression of lipolytic genes. In this study, we seek to explore direct roles of activins in white adipose tissue. We utilize mouse models with an adipose-specific conditional knockout of Inhba (activin (A) and a global Inhbb (activin (B)-null mutation to understand the combinatory effects of activin deficiency in adipose tissue and metabolism. Loss of either Inhbba or Inhbb has a modest suppressive effect on diet-induced obesity. A combination of adipose-selective deletion of Inhbba on the Inhbb-null background (activin double mutant mice) leads to severe weight loss and reduced adiposity. Combined activin (A and activin (B deficiency up-regulates the gene expression of Cidea, Cpt1b, FoxO1 and Pgc1-( in brown adipose tissue, liver and skeletal muscle in activin double mutant mice when compared to wild type controls. Moreover, we found that activin (A and activin (B deficiency induces ‘britening/beiging’ predominantly in visceral white adipose tissue, where the white adipose tissue adopts a gene expression profile and morphological characteristics of brown adipose tissue. Together our results suggest that disruption of activin signaling reduces body growth and adipostity by up-regulating expression of genes involved in metabolism and mitochondrial function. The ‘britening’ of white adipose tissue suggests that combined loss of activin (A and activin (B probably alters the function of white adipose tissue from storage to energy expenditure in the activin double mutant mice. Together, these data highlight activins as suitable therapeutic targets, especially in visceral white adipose tissue where they might be useful in promoting energy expenditure. Contributors: Namwanje, Maria; Huang, Lihua; Bournat Juan, Brown, Chester 227 BAYLOR COLLEGE OF MEDICINE A TRIPLE SPECIES COCKTAIL FOR TREATMENT OF RECURRENT C. DIFFICILE INFECTION Andrea Kathleen Nash Department of Molecular Virology & Microbiology Advisor: Joseph Petrosino, Ph.D.-Department of Molecular Virology & Microbiology As the incidence of Clostridium difficile infection (CDI) increases, the use of highly successful fecal microbiota transplants continues to grow. Concerns with this procedure, though, including the need for a healthy stool donor, the potential for pathogen transmission, and the inability to standardize a treatment regimen, have lead to exploration of alternative approaches. One approach involves the transplant of a defined bacterial community, which avoids the concerns that come with using feces mentioned above. A man with chronic co-morbidities was admitted to the VA Hospital in Houston, TX because of C. difficile-associated diarrhea that began while receiving augmentin. Stools were C. difficile toxin positive by PCR and enteric pathogen negative. Vancomycin was given with minimal effect. After 7 days he received a Bacteroides transplant, which contained 109 cells of three different Bacteroides species resuspended in 200 mL of preservative-free saline (with 1% human albumin), infused into the proximal intestine via an ultraslim endoscope. He improved rapidly and within 24 hours was clinically well. He was discharged after 12 days with one formed bowel movement every other day and has remained without diarrhea for more than one year. Assessment of changes in the stool microbiota was done by 16S rRNA gene sequencing using DNA isolated from samples collected on the day of treatment (before treatment), one to eleven days post-treatment, and from a final sample collected at a day 40 follow-up visit. Treatment with the triple species Bacteroides infusion resulted in marked changes in the microbial communities found in the stool of the subject during the first 10 days of treatment. By day 40, a dramatic rise in Bacteroides was observed. Whole genome shotgun sequencing studies revealed that all of the three strains were present up to 40 days after treatment. Therefore, relapsing chronic CDI was successfully treated with Bacteroides replacement therapy, which avoided the additional risks of using human feces. In contrast to fecal microbiota transplants, only three bacterial strains were required for successful amelioration of disease. Additional clinical studies await receipt of a pending IND. Contributors: Nash, Andrea; Wong, Matthew C; Smith, Daniel; Ajami, Nadim J; Attumi, Tariq; Opekun, Antone; Metclaf, Ginger A; Muzny, Donna; Highlander, Sarah; Graham, David Y; Petrosino, Joseph F 228 2014 GRADUATE STUDENT SYMPOSIUM EXPLOITING CONSERVED DEPENDENCIES TO TARGET UNDRUGGABLE CANCER DRIVERS Nicholas Jay Neill Department of Molecular & Human Genetics Advisor: Thomas Westbrook, Ph.D.-Department of Biochemistry & Molecular Biology c-MYC (MYC) overexpression or hyperactivation is one of the most common drivers of human cancer; however, despite intensive study, the MYC oncogene remains recalcitrant to therapeutic inhibition. Therefore, the development of targeted therapies for MYC-driven cancers remains an important challenge facing the field of cancer therapeutics. Non-oncogene addiction, the dependence of cancers on support pathways that allow cell survival in the presence of oncogene-induced stress, is a concept that can be exploited to develop targeted therapies for undruggable cancer drivers such as MYC. Our laboratory has identified several such non-oncogene addictions in MYCdriven breast cancer, including the cellular sumoylation machinery and the spliceosome. Importantly, we have also discovered that genetic background strongly influences the dependencies of MYC-driven tumors. Given the diverse spectrum of tumor-derived mutations that exist within any particular patient population, it is crucial to consider the effect of genetic background on dependencies that are intended to be translated into clinical therapies. The goal of this work is to identify genetic backgrounds that enhance the dependency of MYC-driven cancers on known non-oncogene addictions of MYC and to discover novel dependencies that are maintained across multiple genetic backgrounds. To do this, we will perform pooled shRNA screens in human mammary epithelial cells that have been engineered to allow inducible MYC hyperactivation in four separate genetic backgrounds. The results of these screens will allow us to identify cellular pathways that MYC-driven cancers are dependent on for survival under many different conditions, so-called “conserved” dependencies. The identification of these types of dependencies is important because they represent therapeutic targets that are potentially relevant to a larger patient population than dependencies that are restricted to a specific genetic background. In addition, it will be critical to determine whether known dependencies of MYC-driven cancers, such as the sumoylation and splicing machinery, are conserved across multiple genetic backgrounds or restricted to a particular set of circumstances. Contributors: Neill, Nicholas; Hsu, Tiffany; Dominguez-Vidana, Rocio; Karlin, Kristen; Westbrook, Thomas 229 BAYLOR COLLEGE OF MEDICINE WHOLE-ORGANISM METABOLOMIC PROFILING OF ANTHRAX REVEALS A NOVEL ROLE FOR LIPID MEDIATORS IN DISEASE PROGRESSION Chinh Thi Quynh Nguyen Department of Molecular Virology & Microbiology Advisor: Anthony Maresso, Ph.D.-Department of Molecular Virology & Microbiology Whereas DNA provides the information to design life, and proteins the materials to construct it, the metabolome can be viewed as the physiology that powers it. As such, metabolomics, the field charged with the study of dynamic small molecule fluctuations in response to changing biology, is now being used to study the basis of disease. The use of metabolomics has been increasingly gaining interest in the field of infectious diseases. The metabolome of an organism reflects all metabolic fluctuations and can give powerful insight into characterizing infection as well as fuel the discovery of new biomarkers. Here, we describe the first metabolomic analysis of a systemic bacterial infection using Bacillus anthracis, the etiological agent of anthrax disease, as the model pathogen. Whole organ and blood analysis identified more than 200 different metabolites that changed in response to infection, with most drastic alterations in lipid metabolites and their associated biosynthesis pathways. Bioinformatic analysis revealed two prominent lipid pathways affected during infection. Products of the polyunsaturated fatty acid and lysolipid biosynthesis pathways, potent mediators of the innate immune response that rely on the activity of host phospholipase A2 (PLA2) enzymes, were reduced in infected mice compared to uninfected control. Metabolite changes were detected as early as 1 day post-infection, well before onset of disease or the spread of bacteria to organs. These findings suggests a novel mechanism in which B. anthracis actively suppresses innate immune response by downregulating the levels of proinflammatory lipid mediators produced by PLA2 enzymes, thus preventing the initiation of robust host defense mechanisms. Functional studies confirmed the importance of PLA2 in host survival as drug inhibition of PLA2 activity potentiated disease severity and increased mortality in mice infected with B. anthracis compared to animals given bacteria or drug alone. Collectively, this study provides a blueprint for using metabolomics as a diagnostic platform that grades the stage and type of infection, as well as an identifier of novel pathogen- and host-induced processes that contribute to bacterial pathogenesis. Contributors: Nguyen, Chinh Thi Quynh; Maresso, Anthony 230 2014 GRADUATE STUDENT SYMPOSIUM MTOR INHIBITION SUPPRESSES FULLY ESTABLISHED EPILEPSY AND ASSOCIATED NEUROPATHOLOGY IN A MOUSE MODEL OF CORTICAL DYSPLASIA Lena H Nguyen Department of Neuroscience Advisor: Anne Anderson, M.D.-Department of Pediatrics Rationale: Cortical dysplasia (CD) is a malformation of cortical development that is a prevalent cause of severe and intractable pediatric epilepsy. Hyperactivation of the mechanistic target of rapamycin (mTOR) pathway has been demonstrated in human CD as well as animal models of epilepsy. Although inhibition of mTOR signaling early in epileptogenesis suppresses epileptiform activity in the neuron subset-specific Pten knockout (NS-Pten KO) mouse model of CD, the effects of mTOR inhibition after epilepsy is fully established have not been investigated in this model. Here, we evaluated whether mTOR inhibition suppresses epileptiform activity and other neuropathological correlates in older NS-Pten KO mice with severe and well-established epilepsy. Methods: The progression of mTOR pathway dysregulation and hippocampal gliosis were evaluated using western blotting in 2, 4, 6, and 8-9 week-old NS-Pten KO mice. Antibodies against p-S6 (S240/244) and p-AKT (S473) were used as markers for mTORC1 and mTORC2 activation, respectively. GFAP and IBA1 were used as markers for astrocytes and microglia, respectively. NS-Pten KO mice were treated with the mTOR inhibitor rapamycin (10 mg/kg i.p., 5 days/week) starting at postnatal weeks 9 and monitored with video-electroencephalography (EEG) recordings for epileptiform activity. Western blotting was also performed to evaluate the effects of rapamycin on the expression of the glial markers. Results: In parallel to the previously reported progressive epilepsy phenotype, we found elevated protein levels of p-S6, p-AKT, GFAP, and IBA1 in NS-Pten KO mice that became increasingly different from age-matched WT mice with age (p<0.05; n=6-15). Treatment with rapamycin significantly suppressed epileptiform activity (p<0.001; n=5-6) and increased survival (p<0.05; n=7-20) in severely epileptic NS-Pten KO mice compared to naïve and vehicle-treated controls, suggesting that aberrant mTOR signaling may play a crucial role in the maintenance of epilepsy in this model. At the molecular level, rapamycin treatment was associated with a reduction in the levels of pS6 and p-AKT as well as GFAP and IBA1 (p<0.05; n=7-14). Conclusions: Late inhibition of mTOR suppresses established, severe epilepsy and associated hippocampal gliosis in NS-Pten KO mice. These findings reveal a wide window for successful therapeutic interventions with mTOR inhibition in the NS-Pten KO mouse model and further support mTOR inhibition as an effective treatment for latestage epilepsy associated with CD. Contributors: Brewster, Amy; Clark, Madeline; Regnier-Golanov, Angelique; Sunnen, C. Nicole; Patil, Vinit; Anderson, Anne 231 BAYLOR COLLEGE OF MEDICINE A HER2/SRC-3/E2F SIGNALING AXIS REGULATES PROLIFERATION AND IDENTIFIES THERAPEUTIC TARGETS IN BREAST CANCER CELLS Bryan C Nikolai Department of Molecular & Cellular Biology Advisor: Bert O'Malley, M.D.-Department of Molecular & Cellular Biology Approximately 20% of early-stage breast cancers display amplification or overexpression of the ErbB2/HER2 oncogene, conferring poor prognosis and resistance to endocrine therapy. Specific targeting of HER2+ tumors with trastuzumab or the receptor tyrosine kinase (RTK) inhibitor lapatinib drastically improves survival, yet tumor resistance and progression of metastatic disease can develop over time. While the mechanisms of cytosolic HER2 signaling are well studied, nuclear signaling components and gene regulatory networks that bestow mechanisms of therapeutic resistance and proliferation are less well understood. Here, we use biochemical and bioinformatics approaches to identify effectors and targets of HER2 transcriptional signaling in BT-474 cells. Phosphorylation and activity of the Steroid Receptor Coactivator-3 (SRC-3) is reduced upon HER2 knockdown, and recruitment of SRC-3 to regulatory elements of endogenous genes is diminished. Transcripts regulated by HER2 signaling are highly enriched with E2F binding sites and define a gene signature associated with proliferative breast tumor subtypes, cell cycle progression, and G1 to S phase transition. Interestingly, pathway and network analysis identifies a cyclin dependent kinase (CDK) signaling node that, when targeted using the CDK4/6 inhibitor PD 0332991, is cooperative with other signaling pathways and potential therapeutic inhibitors thereof. This work shows that HER2 signaling drives proliferation in breast cancer cells through regulation of E2F target genes, at least in part through phosphorylation of SRC-3. Moreover, these results have implications for logic-based discovery of pharmacological combinations in pre-clinical models of adjuvant treatment and therapeutic resistance. Contributors: Lanz, Rainer B; Creighton, Chad J; Hilsenbeck, Sue G; Lonard, David M; Smith, Carolyn L; and O’Malley, Bert W. 232 2014 GRADUATE STUDENT SYMPOSIUM SEQUENCE, STRUCTURE, FUNCTION IN NON-CODING RNAS Ilya Novikov Department of Biochemistry & Molecular Biology Advisor: Olivier Lichtarge, M.D./Ph.D.-Department of Molecular & Human Genetics Functional non-coding RNAs play a vital role in a variety of subcellular processes including RNA and DNA modification, maintenance of genome stability, and gene regulation. In this work we aim to gain better insight into the evolutionary relationship between sequence, structure, and function of these molecules. In particular, we hypothesize that functionally-important nucleotides in these molecules can be identified by analyzing the patterns of sequence and phylogenetic variation displayed by the molecule though its evolutionary history. We then combine this evolutionary information with structural analysis and discover that the more evolutionarily-important nucleotides tend to form well-defined, non-random clusters on the structure of the molecule. We also show that these clusters often constitute known functionally-relevant regions, such as metabolite and ion binding sites, protein interfaces, and catalytic pockets. This suggests that evolutionarily-important nucleotides in structured ncRNA molecules evolve in a manner that is detectable, and that functional regions in these ncRNA molecules can be predicted based on sequence and phylogenetic information. Contributors: Wilkins, Angela; Lichtarge, Olivier 233 BAYLOR COLLEGE OF MEDICINE CHARACTERIZATION OF THE NOVEL NPAS2 CKO MOUSE MODEL INDICATES ROLE IN REGULATING METABOLIC HOMEOSTASIS Derek Steven O’Neil Program in Translational Biology & Molecular Medicine Advisor: Kjersti Aagaard, M.D./Ph.D.-Department of Obstetrics & Gynecology William Gibbons, M.D.-Department of Obstetrics & Gynecology Objective: We have previously demonstrated that a maternal high fat diet is associated with the disruption of the circadian pathway in the fetal liver. Npas2, a circadian gene, is associated with metabolic regulation in fetal development. It is unknown how circadian rhythms are established in fetal development with the lack of light/dark cues through the central clock. We have developed a novel Npas2 conditional knock out (cKO) mouse model of the peripheral clock (liver) to investigate the role of the peripheral clock in establishing metabolic homeostasis. Study Design: Npas2 cKO were generated by targeting the deletion of exon 3 through the cre-lox system of conditional gene deletion. Mice with the Albumin Cre transgene and heterozygous for the loxp flanked (Fl) region of the Npas2 gene (Albcre;Fl/+) were mated to control (Fl/Fl) mice to generate Npas2 cKO mice (Albcre;Fl/Fl) and control mice (Albcre;Fl/+ ). Mice are genotyped between postnatal day 14-20 (p14-20) by PCR amplification of tail genomic DNA. 149 mice were weaned at p21 onto control (CD, Harlan Teklad TD.08485) or high fat diet (HFD, Harlan Teklad TD.88137) and weighed biweekly. Results: Loss of Npas2 in the liver does not alter weight gain of the mice. As expected, both the male cKO mice and the Albcre;Fl/+ control mice placed on a HFD gained significantly more weight than the mice placed on the CD by 5 weeks post weaning (p=.02 and .04 respectively). However, male Fl/Fl mice on both CD and HFD were not significantly different at 5 weeks (p=.29) and 15 weeks (p=.27) post weaning. We observed that among the mice placed on the HFD the Albcre;Fl/+ mice weighed more than the mice of the cKO (2.1±.90g) and Fl/Fl (1.99±.59g) groups (15 weeks post weaning). Conclusion: We successfully generated a novel mouse model to study to role of the peripheral clock, regulated by Npas2. The initial phenotype indicates that Npas2 in the peripheral clock (liver) is necessary for maintaining metabolic homeostasis in response to a high fat diet. Contributors: Derek O’Neil, Danielle Goodspeed, Laura Krannich, Kjersti Aagaard 234 2014 GRADUATE STUDENT SYMPOSIUM IDENTIFICATION OF GERMLINE MUTATIONS IN TEP1 AMONG FAMILIAL AND SPORADIC PEDIATRIC ACUTE MYELOGENOUS LEUKEMIA COHORTS Ninad Ramesh Oak Department of Molecular & Human Genetics Advisor: Sharon Plon, M.D./Ph.D.-Department of Pediatrics Identification of cancer susceptibility gene mutations can impact patient risk stratification, surveillance for second primary malignancies and risk in family members. Germline mutations in genes related to telomere maintenance are associated with bone marrow failure and hematologic malignancies. To explore genetic predisposition to pediatric cancers, we performed whole exome sequencing of constitutive DNA from childhood cancer patients including seven kindreds with familial leukemia/lymphoma in at least two first degree relatives. We filtered for rare variants (<1% in dbSNP or in another non-cancer database, Atherosclerosis Risk in Communities (ARIC)) that are shared between affected individuals within a family and were predicted to exert deleterious effects on the protein. We limited our analysis to truncating (nonsense and frameshift) mutations. In one of the families, we found 7 frameshift and 4 nonsense mutations. The most interesting candidate among these was the novel variant p.R314X in TEP1. Loss-of-function mutations in TEP1 are very rare in sequence databases including the NHLBI exome sequencing project. This rare mutation was shared by a parent-child pair affected by acute lymphocytic leukemia (ALL) and acute myeloid leukemia (AML) respectively. We further explored whether germline mutations in TEP1 are found in larger cohort of apparently sporadic AML patients. AmpliSeq™ sequencing of 43 genes related to telomere maintenance and DNA repair pathways was performed on DNA samples from 82 pediatric AML patients. This analysis revealed 10 rare missense germline mutations in TEP1. Four of these 10 mutations predicted to have damaging effect on protein function according to Combined Annotation Dependent Depletion (CADD) tool (CADD scaled score of 15 or more). The TEP1 familial p.R314X truncating mutation in addition to germline TEP1 missense mutations in sporadic AML patients warrant further functional studies. These studies will include assessing the role of these TEP1 mutations on the telomere length phenotype of cells from the patients carrying the mutation. The results of these studies are designed to determine whether TEP1 represents another member of the telomere maintenance genes to be associated with AML predisposition. This work was supported by research grant RP10189 from the Cancer Prevention and Research Institute of Texas and R01-CA138836 to Plon, SE and training grant T32 GM007526 to Powell, BC. Contributors: Oak, Ninad; Ritter, Deborah; Powell, Bradford; Cheung, Hannah; Gramatges, Monica; Wheeler, David; Plon, Sharon 235 BAYLOR COLLEGE OF MEDICINE CARDIAC SPECIFIC FKBP12 DEFICIENCY PRODUCES DILATED CARDIOMYOPATHY WITH MARKEDLY ENHANCED SUSCEPTIBILITY TO PERIPARTUM DEATH Joshua Michael Oakes Program in Cardiovascular Sciences Advisor: Susan Hamilton, Ph.D.-Department of Molecular Physiology & Biophysics Two immunophilins, FKBP12 and FKBP12.6, regulate the activity of cardiac Ca2+ release channels (RyR2). Mice with a cardiac-specific deficiency in FKBP12 display an early onset dilated cardiomyopathy. To define the mechanisms that drive this myopathy, we assessed changes in Ca2+ handling in isolated cardiomyocytes. Ca2+ spark frequency and amplitude, but not duration, are increased by FKBP12 deficiency. We found that this increase in SR leak is due to enhanced SR stores alongside an increase in cytoplasmic Ca2+ levels. The source of this global Ca2+ increase in unclear, but could be due to enhanced Ca2+ influx, reduced efflux, or lowered Ca2+ buffering capacity Female FKBP12 deficient mice also displayed dilated cardiomyopathy but following birth have an increased risk of dying due to heart failure. Performing echocardiography, we found that between E14.5 and birth FKBP12 deficient females transition from beneficial to deleterious hypertrophy. In summary, these results indicate that FKBP12 plays a vital role in cardiac function at both baseline and during pregnancy, and Ca2+ handling in cardiomyocytes. Contributors: Oakes, Joshua; Hanna, Amy; Dagnino-Acosta, Adan; Lee, Chang Seok; Hamilton, Susan 236 2014 GRADUATE STUDENT SYMPOSIUM DETERMINING CELL TYPE PROPORTIONS AND METHYLATION PROFILES OF CONSTITUTING CELL TYPES IN BREAST CANCER Vitor Onuchic Program in Structural and Computational Biology and Molecular Biophysics Advisor: Aleksandar Milosavljevic, Ph.D.-Department of Molecular & Human Genetics A known issue with DNA methylation profiling in complex tissues, such as breast, is the fact that such datasets are generated from a heterogeneous cell population, and that each of the constituting cell types has a distinct methylation profile. Since methylation profiling techniques only measure the average level of methylation for a cell population, changes in cell type proportions between different samples will lead to differences in methylation between those samples. Distinguishing between methylation differences that are due to changes in proportions of cell types and those that are due to changes in the methylation status of a particular region of the genome in a specific cell type is essential for improving the sensitivity and specificity of case-control studies focused on changes in methylation. Further, determining the proportions of different cell types in each complex tissue sample will also allow one to detect changes in proportions of particular cell types between different sets of samples. Examples of the importance of detecting such shifts in cell type compositions are that changes in proportions of cell types in blood have been shown to associate with aging, and higher levels of immune cells in breast cancer have been shown to associate with better prognosis. We selected a set of 1000 target regions that are known to be involved in breast cancer biology. Using a set of primers designed to specifically amplify these regions in bisulfite treated genomic DNA, we have applied microdroplet-based targeted sequencing approach to 64 bisulfite-converted samples including breast cancer cell lines, a range of ER positive and negative tumors, and a set of breast tumor samples in different stages of development coming from the same individual. This dataset will be used to test a few different approaches for estimating both cell type composition of breast cancer samples, as well as methylation profiles of individual constituent cell types of such samples. The same approaches will then be applied to the 450K and 27K array breast cancer datasets available in TCGA. Contributors: Onuchic, Vitor; Hartmaier, Ryan; Boone, David; Roth, Matt; Samuels, Michael; Lee, Adrian; Milosavljevic, Aleksandar; 237 BAYLOR COLLEGE OF MEDICINE CYP7B1 ENZYME DELETION IMPAIRS MALE REPRODUCTIVE BEHAVIORS IN MICE Mario G Oyola Department of Neuroscience Advisor: Shailaja Mani, Ph.D.-Department of Molecular & Cellular Biology Mariella De Biasi, Ph.D.-Department of Neuroscience In addition to androgenic properties mediated via androgen receptors, DHT also regulates estrogenic functions via an alternate pathway. These estrogenic functions of DHT are mediated by its metabolite 5α- androstane-3β, 17β-diol (3β-diol) binding to estrogen receptor β (ERβ). CYP7B1 enzyme, involved in conversion of 3β-diol to inactive 6α- or 7α-triols, could play an important role as a regulator of estrogenic functions mediated by 3β-diol. Using a mutant mouse carrying a null mutation for the CYP7B1 gene (CYP7B1KO), we examined the contribution of CYP7B1 on physiology and behavior. Male, gonadectomized CYP7B1KO and their wild type (WT) littermates were assessed for their behavioral phenotype, anxiety-related behavioral measures and hypothalamic pituitary adrenal (HPA) axis reactivity. No significant effects of genotype were evident in anxiety-like behaviors in open field (OFA), light-dark (LD) exploration, and elevated plus maze (EPM). Testosterone (T) significantly reduced open arm time on the EPM, while not affecting LD exploratory and OFA behaviors in CYP7B1KO and WT littermates. T also attenuated the enhanced corticosterone response, following EPM, in both genotypes. T was able to reinstate male-specific reproductive behaviors (number of mounts, intromission, and ejaculation, and their respective latencies) in the WT, but not in the CYP7B1KO mice. The defect in male reproductive behaviors appears due to their inability of CYP7B1KO to distinguish olfactory cues from an estrous female. CYP7B1KO mice also showed a reduction in androgen receptor (AR) mRNA expression in the olfactory bulb. Our findings suggest a novel role for the CYP7B1 enzyme in the regulation of male reproductive behaviors. Contributors: Oyola, Mario G; Zuloaga, Damian G; Carbone, David; Malysz, Anna M; AcevedoRodriguez, Alexandra; Handa, Robert J; Mani, Shaila K. 238 2014 GRADUATE STUDENT SYMPOSIUM NEUROENDOCRINE REGULATION OF LIPID METABOLISM AND STORAGE Ayse-Sena Ozseker Program in Developmental Biology Advisor: Meng Wang, Ph.D.-Department of Molecular & Human Genetics Lipid metabolism is under control of environmental and genetic factors. The nervous system receives environmental and sensory inputs, and consequently communicates with adipose tissue to control lipid metabolism. To date, the molecular mechanisms underlying the neuroendocrine regulation of lipid metabolism remain poorly understood. My project aims to dissect the regulation of lipid storage and metabolism by sensory perception using Caenorhabditis elegans as a model organism. Based on the preliminary studies, my hypothesis is that signaling in sensory neurons modulates the release of neuroendocrine factors, which transduce signals to systemically regulate lipid homeostasis. To find the neurons and neuropeptides that are involved in the regulation of lipid storage, I have examined lipid storage in 30 chemosensation mutants using stimulated Raman scattering (SRS) microscopy. SRS microscopy is a quantitative chemical imaging technology that does not require labeling lipids or fixing worms and assesses the levels of triacylglycerides, the main lipid storage form in the worm adipose tissue, the intestine. Our best hit from this screen is the daf-11 mutant that lack a guanylyl cyclase expressed in five different head sensory neurons all of which show defective neuronal signaling in the mutant worms. The intestine of C. elegans is not innervated and daf-11 mutants display normal food intake, suggesting that secreted factors from these sensory neurons regulate intestinal lipid storage in a cell non-autonomous manner. Consistent with this hypothesis, EGL-21, a carboxypeptidase essential for neuropeptide processing, is required for the lipid storage increase in daf-11 mutants. Additionally, inactivation of the worm homolog of FoxO transcription factor, daf16, suppresses the increased lipid storage phenotype of daf-11 mutants. DAF-16 is the major effector of insulin/insulin-like growth factor signaling (IIS). daf-2 mutants that lack the insulin receptor also have high intestinal lipid storage in a daf-16 dependent manner. To test whether IIS is downstream of daf-11, we knocked-down the insulin receptor daf-2 in daf-11 mutants by RNAi and found that this can further increase the lipid storage suggesting daf-11 regulates daf-16 in a parallel pathway to IIS. Currently, we are analyzing which neuropeptides are differentially expressed in daf-11 mutants in comparison to wild-type. After identifying such neuropeptides, we will determine their necessity and sufficiency for increased lipid storage. In parallel, we are studying the tissue specificity of daf-16 in lipid storage regulation. Contributors: Ozseker, Ayse Sena; Yu, Yong; Wang, Meng (PI) 239 BAYLOR COLLEGE OF MEDICINE EVALUATING ADENO-ASSOCIATED VIRUS SEROTYPES FOR HEPATOCYTESPECIFIC GENE TARGETING Francis Pankowicz Department of Molecular & Cellular Biology Advisor: Karl-Dimiter Bissig, M.D./Ph.D.-Department of Molecular & Cellular Biology While gene therapy holds great potential for treating a wide variety of diseases, vector delivery remains a significant obstacle to overcome. Viral vector delivery systems are among the most promising, but are hindered by varied tissue-specificity and immunogenic effects. Adeno-Associated Virus (AAV) is a non-integrating virus that can infect both dividing and non-dividing cells, and is currently being evaluated for gene therapy potential due to its diminished immune response in humans. Tissue-specificity for AAV is mediated by its capsid serotype, some of which have been studied and tested in a clinical trial setting. Our work focuses on identifying the most suitable AAV serotype for gene transfer in the liver. Using AAV with known serotypes as well as recently isolated novel serotypes, we have highlighted the dramatic differences between in vitro and in vivo transduction efficiency. Furthermore, by utilizing a human liver chimeric mouse, we have demonstrated the preferential infection of hepatocytes from a species-specific perspective. Finally, we have identified AAV9 as the serotype most suitable for transduction of human hepatocytes in vivo (human liver chimeric mouse). This novel use of human liver chimeric mice therefore uncovers major transduction variability between the AAV serotypes. Moreover, our data suggest that AAV9 may produce superior hepatic transduction in clinical trials than the currently used AAV8. Contributors: Pankowicz, Francis; Wang, Lili; Bell, Peter; Bissig- Choisat, Beatrice; Barzi Dieguez, Mercedes; Kruse, Robert; Legras, Xavier; Wieland, Stefan; Bissig, Karl- Dimiter 240 2014 GRADUATE STUDENT SYMPOSIUM CONTRIBUTION OF APICAL DENDRITES TO RECEPTIVE FIELD PROPERTIES IN LAYER 2/3 OF MOUSE V1 Jiyoung Park Program in Structural and Computational Biology and Molecular Biophysics Advisor: Stelios Smirnakis, M.D./Ph.D.-Department of Neurology Although slice recordings and model studies provide a lot of information about how neurons integrate their inputs to overall responses, it is still not clear how neurons in the brain integrate their synaptic inputs to derive their functional properties in vivo. Pyramidal neurons in layer 2/3 of mouse V1 receive input from LGN via layer 4 and feedback from higher visual areas. Here, we focus on the function of apical dendrites, which play an important role in communicating with higher visual areas. We ablate apical dendrites of layer 2/3 pyramidal neurons in area V1, in vivo, and monitor their responses to visual stimuli by two photon microscopy. We compare orientation selectivity, contrast sensitivity and other receptive field properties of L2/3 pyramidal neurons before and after selective apical dendritic ablation. This approach provides information on how different dendritic inputs contribute to neuronal receptive field properties of L2/3 pyramidal neurons in mouse V1. Contributors: Park, Jiyoung ; Smirnakis, Stelios M 241 BAYLOR COLLEGE OF MEDICINE ROLE OF STABILIZING MUTATIONS IN THE EVOLUTION OF ANTIBIOTIC RESISTANT ENZYMES Maha Praful Patel Program in Translational Biology & Molecular Medicine Advisor: Timothy Palzkill, Ph.D.-Department of Pharmacology Robert Atmar, M.D.-Department of Medicine The Centers for Disease Control and Prevention reports that 2 million people acquire antibiotic-resistant infections and 23,000 people die as a result each year in the United States. β-lactam antibiotics are the most commonly prescribed antibiotics for Gram-negative bacterial infections. The most common mode of resistance to β-lactam antibiotics in Gram-negative bacteria is the production of β-lactamases. In particular, the CTX-M family of β-lactamases is the most widespread and frequently isolated in clinical samples around the world. CTX-M β-lactamases continually evolve in their ability to hydrolyze extended spectrum β-lactam antibiotics under selective pressure through point mutations in and around their active site. Studying the effect that naturally occurring mutations have on the structure, function and specificity of the CTX-M family of β-lactamases will provide insight into the evolution of these enzymes. The CTX-M family of β-lactamases is divided into five subfamilies based on amino acid sequence homology. The P167S and D240G substitutions were identified individually in the active site of CTX-M enzymes in several subfamilies upon the introduction of ceftazidime, a third generation cephalosporin antibiotic, into the clinic. These substitutions allow CTX-M β-lactamases to effectively hydrolyze ceftazidime but also result in an overall loss in stability to the enzyme. Stability defects can be offset through the acquisition of secondary mutations. The A77V mutation has been identified in combination with the P167S and D240G substitutions in numerous clinical isolates. The prevalence of the A77V mutation among clinical isolates emphasizes its probable importance in the evolution of CTX-M β-lactamase enzymes. Here we characterize the A77V mutation in the CTX-M-14 model enzyme using kinetic analysis, competition assays and thermostability assays. We hypothesize that the A77V substitution acts as a stabilizing mutation to offset the defects caused by both the P167S and D240G substitutions, allowing the enzyme to retain stability while acquiring the ability to hydrolyze ceftazidime. Contributors: Patel, Meha; Fryszczyn, Bartlomiej; Palzkill, Timothy 242 2014 GRADUATE STUDENT SYMPOSIUM PRKRIR: A POTENTIAL NOVEL GENOMIC DRIVER OF ENDOCRINE THERAPY RESISTANCE IN BREAST CANCER Sasha McKai Pejerrey Department of Molecular & Cellular Biology Advisor: Suzanne Fuqua, Ph.D.-Department of Medicine Tamoxifen (Tam) has been used for many years to treat women with estrogenreceptor alpha (ER()-positive breast cancer. However, 40% of women who receive Tam eventually relapse with Tam-resistant tumors. Aromatase inhibitors (AIs) are another type of hormone treatment used in patients, but resistance can also develop to these drugs. The mechanisms behind endocrine therapy resistance remain largely undefined, severely limiting treatment options. We have discovered that a novel potential driver of endocrine therapy resistance, called PRKRIR (protein-kinase, interferon-inducible double stranded RNA dependent inhibitor, repressor of (P58 repressor)) was overexpressed in resistant tumors by comparing Tam-sensitive primary human breast tumors to Tam-resistant metastatic tumors. Preliminary data indicates that PRKRIR probably interacts with PAK1 (p21 protein (Cdc42/Rac)-activated kinase 1), another protein known to be involved in resistance. PRKRIR and PAK1 are both co-amplified and co-overexpressed in breast tumors. Additionally, initial studies suggest that PRKRIR overexpression (OE) in vitro promotes Tam resistant growth. I hypothesize that PRKRIR cooperates with PAK1 to promote endocrine therapy resistance in ER-positive breast cancer. Our overall objective is to define how PRKRIR OE promotes endocrine therapy resistance in breast tumors, and to determine if that resistance in turn promotes metastasis and altered cell signaling. Our goal is to restore hormone sensitivity in resistant tumors. We predict that PRKRIR is a novel driver of endocrine therapy resistance. By binding to and activating PAK1, we anticipate that PRKRIR confers growth advantage to cells under endocrine therapy treatment, causing them to become resistant. Elucidating the mechanisms by which cells evolve into resistant populations will introduce a novel biomarker and uncover new clinical targets for personalized therapy. Contributors: Gu, Guowei; Rechoum, Yassine; Beyer, Amanda R., Wang, Xiaosong; Fuqua, Suzanne A. W. 243 BAYLOR COLLEGE OF MEDICINE TRANSCRIPTOMIC ANALYSIS REVEALS POTENTIAL ROLES FOR ERK3 IN FETAL LUNG MATURATION VIA SURFACTANT PROTEIN B AND CORTICOTROPIN RELEASING HORMONE Braden Kyle Pew Department of Molecular & Cellular Biology Advisor: Kjersti Aagaard, M.D./Ph.D.-Department of Obstetrics & Gynecology Dennis Bier, M.D.-Department of Pediatrics The mechanism by which prenatal glucocorticoid treatment promotes fetal lung maturation remains poorly understood despite decades of clinical use. In an Erk3-/murine model of neonatal respiratory distress syndrome (RDS), we showed partial abrogation of the pulmonary immaturity phenotype through antenatal dexamethasone (dex) treatment. Using transcriptomic analysis, we identified key molecular pathways in lung maturation and sought to determine the interaction of glucocorticoid treatment with these genes. Dexamethasone (0.4 mg/kg) or saline was administered on E16.5 and 17.5, and lungs were harvested at E18.5. RNA-seq was performed and analyzed for differential expression, then tested by Ingenuity Pathway Analysis (IPA) to determine gene/dex interaction. CRH and SFTPB protein expression was determined by immunohistochemistry of fetal lungs (n=5) examined by two reviewers. Statistical analysis was performed using Student's t test. Of 596 differentially expressed genes, IPA revealed 36 genes that interact with dex, including several with roles in lung development, including corticotropin releasing hormone (Crh) and surfactant protein B (Sftpb). Antenatal dex was associated with significantly attenuated CRH levels at E18.5 in both wildtype (WT) and Erk3-/- lungs (0.56-fold and 0.67-fold, p<0.001). As in humans, lungs of WT mice demonstrated increased SFTPB production in response to dex (p=0.003). However, Erk3-/- mice exhibited decreased pulmonary SFTPB when treated with dexamethasone (p=0.012). Using transcriptomics in this neonatal RDS model, we identified molecular pathways altered in response to glucocorticoid treatment. Our findings suggest an Erk3 independent and dependent mediation of CRH and SFTPB, respectively, in their roles as modulators of fetal lung maturation. These discoveries broaden our understanding of the underlying mechanism of treatment for neonatal pulmonary immaturity in preterm parturition. Contributors: Pew, Braden; Harris, R. Alan; Sbrana, Elena; Cuevas Guaman, Milenka; Klinger, Sonia; Shope, Cindy; Wang, Hui; Chen, Rui; Meloche, Sylvain; Aagaard, Kjersti 244 2014 GRADUATE STUDENT SYMPOSIUM FOXO1 DRIVES ERK-MEDIATED ARTERIAL SPECIFICATION VIA TRANSCRIPTIONAL REGULATION OF SPROUTY2 Victor George Piazza Program in Cardiovascular Sciences Advisor: Mary Dickinson, Ph.D.-Department of Molecular Physiology & Biophysics Arteriovenous malformations that can arise during development affect an estimated 300,000 Americans and can have severe impacts on the central nervous system. However, much remains to be understood about how arteries and veins form, including the signaling pathways involved and the consequences of disrupting these pathways. It is known that specification of arterial and venous vessels from a primitive capillary plexus is a critical event that begins prior to remodeling of that plexus into mature arteries and veins. Using a mouse model of endothelial FoxO1 transcription factor deletion (Tie2-Cre; FoxO1flox/flox), we have found embryonic lethality associated with defective vessel remodeling. Additionally, a significant reduction was observed in expression of genes associated with arterial fate specification at the pre-blood flow stage (E8.25). Erk activity levels, which we have determined are temporally required for arterial specification at this stage, are reduced in Tie2-Cre; FoxO1flox/flox embryos. Additionally, decreased arterial marker expression was associated with a significant upregulation of the Sprouty2 (Spry2) gene, the product of which is a known Erk antagonist. We have now determined that the Spry2 genomic locus is a target of direct FoxO1 binding in yolk sac endothelial cells. Collectively, this suggests FoxO1 transcriptionally regulates Sprouty2 expression to mediate arterial specification in the mouse embryo. We hypothesize that FoxO1 mediates Erk signaling in the pre-flow mouse embryo by transcriptionally repressing Sprouty2, therefore modulating arterial specification. We are testing this hypothesis as follows: 1) By determining whether Sprouty2 over-expression in the pre-flow mouse embryo recapitulates the arterial phenotype seen in the Tie2-Cre; FoxO1flox/flox FoxO1 mutant and Erk-inhibited embryos, and 2) by determining whether Sprouty2 knockout or knockdown in the Tie2Cre; FoxO1flox/flox mutant is necessary and sufficient to rescue arterial marker expression and vascular remodeling. Parallel experiments in vitro will support the causal relationship between FoxO1 and Sprouty2 in the context of arterial specification. Through this work we will elucidate a novel mechanism governing arterial specification and subsequently embryonic vascular remodeling. The applicability of this knowledge extends to new approaches to both promote vessel growth, as in tissue engineering of vascular constructs, and inhibit neo-vessel formation that accompanies tumor growth. Contributors: Piazza, Victor G; Garcia, Monica D; Wong, Leeyean; Udan, Ryan S; Dickinson Mary E 245 BAYLOR COLLEGE OF MEDICINE RETT SYNDROME LIKE PHENOTYPES IN THE R255X MECP2 MUTANT MOUSE ARE RESCUED BY MECP2 TRANSGENE Meagan Rochelle Pitcher Program in Translational Biology & Molecular Medicine Advisor: Jeffrey Neul, M.D./Ph.D.-Department of Pediatrics Daniel Glaze, M.D.-Department of Pediatrics Over thirty percent of Rett Syndrome (RTT) cases are due to nonsense mutations in MECP2, where a change in nucleotide sequence leads to a premature stop codon in the mRNA transcript. One strategy to overcome disease-causing stop mutations is treatment with nonsense suppressing read-through compounds, such as gentamicin, which reduce the stringency and fidelity of ribosomes translating mRNA messages to allow expression of full length proteins from a mutated gene. To determine if this strategy may be useful in RTT we characterized a new mouse model of RTT that has a knock-in nonsense mutation (p.R255X) in the Mecp2 locus (Mecp2tm1.1Irsf/J). Mecp2 is a four exon gene that encodes two functional domains: the methyl binding domain from exons 3 and 4 and the transcription repression domain in exon 4. Because the R255X mutation is located in the transcription repression domain of Mecp2, it is possible that a dominant negative DNA binding truncation product could be produced from the disease allele. To determine if the truncated gene product acts as a dominant negative allele, we genetically introduced an extra copy of MECP2 via a MECP2 transgene. This allows us to determine whether adding a wild-type version of MeCP2 is sufficient to rescue phenotypic abnormalities in Mecp2tm1.1Irsf/J mice, or whether the truncated allele has a dominant negative effect insurmountable by a wild-type copy. Mecp2tm1.1Irsf/J mice have phenotypes nearly identical to complete null animals: decreased weight early in life, decreased heart rate late in life, abnormal breathing phenotypes, poor motor coordination, and decreased survival time. The addition of the MECP2 transgene to Mecp2tm1.1Irsf/J mice abolished the phenotypic abnormalities and resulted in near complete rescue. This provides a proof of concept that this mutation is amenable to read-through therapy. Future studies will include pharmacokinetic and efficacy preclinical trials in the R255X model using read-through compounds that are currently in clinical trials for peripheral indications. We hope to demonstrate that read-through therapy is a viable treatment option for neurological disease caused by nonsense mutations. Contributors: Pitcher, Meagan R; Herrera, Jose A; Fisher, Amanda R; Schanen, N. Carolyn; Neul, Jeffrey L 246 2014 GRADUATE STUDENT SYMPOSIUM ACTIVITY-DEPENDENT TRANSCRIPTIONAL AND CHROMATIN DYNAMICS IN MECP2 DISORDERS Amy E Pohodich Department of Neuroscience/M.D.-Ph.D. Program Advisor: Huda Zoghbi, M.D.-Department of Pediatrics Methyl-CpG-Binding Protein 2 (MeCP2) was first described as a transcriptional repressor that binds to methylated DNA and garnered much research interest after the discovery that mutations in MECP2 cause Rett syndrome (RTT). RTT is a postnatal neurodevelopmental disorder characterized by a period of seemingly normal development followed by a progressive loss of acquired speech, cognitive abilities, and motor coordination. Stereotyped hand movements replace purposeful hand use, and seizures and autonomic dysfunction develop within a few years of disease onset. The importance of having the correct level of MeCP2 in the brain is underscored by the fact that duplications and triplications spanning MECP2 produce syndromic intellectual disability, progressive motor dysfunction, and seizures. In RTT patients, the delayed onset of disease coincides with the timing of peak stimulus-driven synaptic reorganization required for appropriate circuit formation and neuronal maturation, which led to the hypothesis that MeCP2 is important for establishing, refining, and/or maintaining synapses. Consistent with this hypothesis are the numerous abnormalities observed in MeCP2-deficient mice, including changes in basal transmission, presynaptic function, and alterations in the balance between excitatory and inhibitory signaling. Currently, however, the precise mechanisms by which MeCP2 can contribute to neuronal plasticity are unclear. To gain insight into the role of MeCP2 during neuronal activity in the mature brain, this project utilizes deep brain stimulation (DBS) to elicit robust activation of the dentate gyrus (DG) in awake, freely moving mice. This paradigm will enable assessment of the genome-wide transcriptional requirements for MeCP2 upon activation, and these data will be paired with analysis of changes in both MeCP2 localization and protein interactions to yield insight into MeCP2 function in neurons. Contributors: Pohodich, Amy; Zoghbi, Huda 247 BAYLOR COLLEGE OF MEDICINE TRYPTOPHAN IMPORT AND COLONY GROWTH POST-DNA DAMAGE ARE CONTROLLED BY SEA3’S REGULATION OF TORC1 SIGNALING Erica Jean Polleys Integrative Program in Molecular and Biomedical Sciences Advisor: Alison Bertuch, M.D./Ph.D.-Department of Pediatrics The Saccharomyces cerevisiae Iml1 complex inhibits TORC1 signaling and SEACAT antagonizes the Iml1 complex. Whereas conditions in which the Iml1 complex inhibits TORC1 have been described, those in which SEACAT functions remain largely unknown. The SEACAT member Sea3 was linked to telomere maintenance and DNA repair via physical and genetic interactions reported in genome-wide studies. Therefore, we questioned whether Sea3 functioned through TORC1 to influence these pathways. Deletion of SEA3 delayed emergence of telomerase independent survivors that utilize break-induced replication (BIR) to maintain their telomeres. Similarly, sea3∆ mutants exhibited a delay in colony formation in a BIR assay strain following double strand break (DSB) induction as well as on the DNA damaging agent bleomycin. The delay in colony formation was not due to a delay in the repair of the DSB or termination of the DNA damage checkpoint, but due to tryptophan auxotrophy. High levels of tryptophan in yeast peptone dextrose media did not rescue the delay in colony formation, suggesting a defect in tryptophan import. Consistent with this, the tryptophan permease Tat2 prematurely declined in sea3∆ mutants compared to wildtype. Deletion of IML1 rescued the delay in colony formation, consistent with Sea3 functioning as a regulator of TORC1 signaling. Together, these findings highlight the importance of enforcement of TORC1 signaling and internal tryptophan in the recovery of growth post DNA damage. Contributors: Polleys, Erica; Bertuch, Alison 248 2014 GRADUATE STUDENT SYMPOSIUM OBESTATIN STIMULATES INSULIN SECRETION UNDER GLUCOSE-STIMULATED CONDITION VIA GHRELIN RECEPTOR Geetali Pradhan Program in Translational Biology & Molecular Medicine Advisor: Yuxiang Sun, M.D./Ph.D.-Department of Pediatrics Susan Samson, M.D.-Department of Medicine Introduction: Obestatin, a 23 amino acid peptide derived from the ghrelin gene, is expressed in various tissues including stomach and pancreas. Obestatin is known to reduce food intake and body weight, improve memory and regulate sleep, but has no effect on secretion of growth hormone and corticosterone. Obestatin is also known to increase mass and survival of pancreatic β cells but it’s effect on insulin secretion remains unclear. Methods: We studied the effect of obestatin on insulin secretion under glucosestimulated condition both in vitro and ex vivo using rat insulinoma INS-1 cells and mouse pancreatic islets. To determine whether the effect of obestatin on insulin secretion is mediated through the ghrelin receptor, Growth Hormone Secretagogue Receptor (GHS-R), we used islets from our GHS-R knockout mouse model (ex vivo). We also transiently knocked down GHS-R in INS-1 cells and pharmacologically blocked GHS-R using antagonists YIL 781 and JMV2959. Results: Treatment of INS1 cells and pancreatic islets with different doses of Obestatin, significantly increased glucose stimulated insulin secretion (GSIS) compared to no obestatin treatment. However, static incubation of pancreatic islets from GHS-R KO mice showed no response to obestatin treatment. Further, treatment of obestatin in GHS-R knockdown INS-1 cells also showed similar effect on insulin secretion under glucose-stimulated condition. Similarly, on blocking GHS-R in INS-1 cells pharmacologically using antagonists also reduced obestatin’s effect on GSIS. Our results indicate that obestatin has profound stimulator effect on insulin secretion in both INS-1 cells and mouse pancreatic islets and this effect is potentially mediated via GHSR in β cells. Conclusions: In conclusion, our studies indicate that obestatin is a potent insulin secretagogue under glucose-stimulated condition. This effect of obestatin is likely mediated via its receptor GHS-R in pancreatic islets. Obestatin’s stimulatory effect on insulin secretion and promoting effect on β cell survival together make obestatin a powerful therapeutic candidate for Type 2 diabetes. Contributors: Pradhan, Geetali; Lee, Jong Han; Sun, Yuxiang 249 BAYLOR COLLEGE OF MEDICINE LOSS OF ENTEROCYTE DERIVED ARGININOSUCCINATE LYASE RESULTS IN INCREASED INCIDENCE OF NECROTIZING ENTEROCOLITIS Muralidhar Hebbur Premkumar Clinical Scientist Training Program Advisor: Brendan Lee, M.D./Ph.D.-Department of Molecular & Human Genetics Background: Nitric oxide (NO) is a mediator in the pathogenesis of necrotizing enterocolitis (NEC). Argininosuccinate lyase (ASL) is the only enzyme in the body capable of generating arginine, the substrate for NO. Previously, we have shown that ASL is required for the assembly of the NO synthesis complex and, loss of ASL results in decreased NO production. Enterocytes are the principal sites expressing ASL during the neonatal period. Understanding the contribution of cell specific NO production to the development of NEC has been limited by redundancies of the nitric oxide synthase (NOS) isoforms and the temporal and spatial regulation of NO production. Hence, we have generated an enterocyte-specific Asl CKO mouse, a novel, and better model to study the role of NO in causation of NEC in a cell-specific manner. Objective: We hypothesize that the deficiency of enterocyte-derived NO secondary to loss of ASL contributes to the development of NEC. Design/Methods: Utilizing Cre-Lox technology, we generated an enterocyte-specific knockout of Asl (CKO). NEC was established in these mutant and control mice by subjecting premature mouse pups to exclusive formula feed, hypoxia, and hypothermia. The severity of NEC was graded based on histological changes. Characterization of the model was performed by intestinal histomorphometry, immunostaining, nitrosylation studies, cytokine expression and microarray analysis. In-vitro assays were performed on IEC-6 cells. Results: Significant loss of ASL in the enterocytes was demonstrated by RTPCR, western blot and immunostaining. The expression of eNOS, IL-6, and BAX were significantly elevated in the CKO. The incidence of NEC was significantly higher (p=0.003) in the CKO [29/46(63%)] as compared to controls [32/84 (38%)]. CKO mice demonstrated elevated levels of IL-6, and neutrophilic infiltration. CKO also demonstrated significantly increased apoptosis as assessed by TUNEL staining and levels of pro-apoptotic proteins (BAX, AIF). IEC-6 cells with knockdown for Asl (shAsl) showed decreased migration in response to LPS. Conclusions: Loss of ASL in the enterocytes resulted in increased incidence of NEC. This enterocyte-specific loss of ASL was associated with increased states of inflammation and apoptosis. In-vitro studies suggested that loss of Asl in presence of stress impairs the migration of enterocytes in light of stress. Thus, our studies suggest that enterocyte-derived ASL is protective against the development of NEC. Hence, manipulation of enterocyte ASL may be of translational value in prevention or treatment of NEC. Contributors: Muralidhar H. Premkumar1, Brendan Lee2. 1Division of Neonatology, Baylor College of Medicine, Houston 2Department of Molecular and Human Genetics, Baylor College of Medicine, Houston 250 2014 GRADUATE STUDENT SYMPOSIUM CRYSTAL STRUCTURES OF CGMP-DEPENDENT PROTEIN KINASE Iα (PKG Iα) LEUCINE ZIPPER REVEAL THE ROLE OF THE DISULFIDE BOND C42-C42 IN BINDING ITS ISOZYME SPECIFIC SUBSTRATE, RHOA. Liying Qin Department of Biochemistry & Molecular Biology Advisor: Choel Kim, Ph.D.-Department of Pharmacology Type Iα cGMP-dependent protein kinase (PKG Iα) regulates physiological processes such as vasorelaxation and smooth muscle tone, thus plays a crucial role in heart protection and blood pressure regulation. Because PKG Iα is a broad specificity kinase, its interaction with specific substrates is essential for proper substrate phosphorylation and signal transduction. As one of the functionally important substrates of PKG Iα, RhoA is recognized by the N-terminal leucine zipper (LZ) of the kinase. Phosphorylation of RhoA at serine188 by PKG Iα inhibits RhoA from activating Rho kinase, resulting in smooth muscle relaxation. While it is well established that PKG Iα can be activated by either cGMP or autophosphorylation, recent reports suggest that residue cysteine42 (C42) may serve as a redox sensor where its oxidation and the resultant interchain disulfide bond (C42-C42’) formation directly activates PKG Iα independent of cGMP. However, the underlying mechanism of this redox sensing remains unknown. We hypothesize that the formation of C42-C42 disulfide bond does not directly activate PKG Iα independent of cGMP, but indirectly by increasing the stability of the LZ domain, resulting in a higher affinity for RhoA. Since most of oxidizing regents suffer from partial oxidation and non-specific effects, we constructed two LZ mutants, C42S and C42L, representing the reduced and oxidized states of the wild type LZ, respectively. We then solved the crystal structures of both wild type and C42L LZ domains. The structure of the C42L LZ is highly similar to that of the wild type containing the C42-C42 disulfide with an RMSD of 0.49 angstrom. The C42L LZ also has a lower temperature factor than wild type, indicating a higher thermal stability. To further investigate the stability of the LZ domains, we are in the process of obtaining their melting temperatures using circular dichroism. Ongoing studies include a pull-down assay to test the interaction between wild type and mutant PKG Iα and RhoA and a kinase assay to investigate the phosphorylation of RhoA by PKG Iα in vitro. Contributors: Qin, Liying; Reger, Albert S.; Guo, Elaine; Yang, Matthew P.; Zwart, Peter; Kim, Choel 251 BAYLOR COLLEGE OF MEDICINE STRIATED MUSCLE PREFERENTIALLY EXPRESSED GENE (SPEG) IS NECESSARY FOR ADULT CARDIAC FUNCTION IN MICE Ann Pepper Quick Program in Cardiovascular Sciences Advisor: Xander Wehrens, M.D./Ph.D.-Department of Molecular Physiology & Biophysics BACKGROUND. Heart failure is one of the leading causes of hospitalization, with a median survival rate of only 5 years,1 yet there remains a lack of targeted therapeutic options. Calcium dysregulation through disruption of calcium release proteins including ryanodine receptor type 2 (RyR2) and junctophilin-2 (JPH2) has been implicated in heart failure. SPEG, a novel prospective regulator of calcium release was found by coimmunoprecipitation (co-IP) from mouse heart lysate for RyR2 and JPH2 followed by mass spectrometry. SPEG is a kinase necessary for cardiac development.2 However, it remains unknown whether SPEG is essential for adult cardiac function. Therefore, we obtained SPEG tamoxifen-inducible conditional knockout (cKO) mice in order to study the physiological role of SPEG in the adult mouse heart. HYPOTHESIS. SPEG maintains proper adult cardiac function by regulating RyR2 calcium release. METHODS. Transthoracic echocardiography was performed on anesthetized mice at baseline and 4 and 8 weeks post tamoxifen injection. Echocardiograms were analyzed using Vevo 2100 to determine cardiac function. At 8 weeks post tamoxifen injection, mice were euthanized and cardiomyocytes were isolated for either 1) calcium imaging with fluo-4AM 2) T-tubule staining with di-8-anneps, or 3) co-localization studies of JPH2 and RyR2 by immunofluorescence. Cells were imaged by confocal microscopy and analyzed using Image J. RESULTS. 8 weeks post tamoxifen injection, cKO mice developed heart failure with an average ejection fraction of 27.1%+/-17.7% compared to 66.3% in the control. SPEG cKO cells exhibited > 2-fold increased Ca spark frequency and deceased transient amplitude. T-tubule power was decreased by 50% in SPEG cKO cells and JPH2-RyR2 co-localization was decreased in SPEG cKO cardiomyocytes (R= 0.19) compared to controls (R=.42). CONCLUSIONS. SPEG is necessary for adult cardiac function in mice. Physiological and cellular studies confirm that SPEG cKO mice develop heart failure associated with calcium dysregulation, T-tubule disruption, and decreased JPH2-RyR2 co-localization. Future studies prior to the onset of heart failure are needed to determine whether the loss of SPEG is the cause or effect of disrupted calcium homeostasis. 1. Go AS et al. Circulation.129:e28-e292 2. Liu X et al. Circulation. 2009;119:261-268 Contributors: Quick, Ann P.1; Wang, Qiongling1; Reynolds, Julia O.1; Chiang, David Y.2; Beavers, David L.2; Wehrens, Xander H.T.1. 252 2014 GRADUATE STUDENT SYMPOSIUM FUNCTIONAL PREDICTION AND CHARACTERIZATION OF THE MALARIAL PROTEIN EXP1 Joel Patrick Quiros Integrative Program in Molecular and Biomedical Sciences Advisor: Olivier Lichtarge, M.D./Ph.D.-Department of Molecular & Human Genetics The malaria parasite Plasmodium falciparum causes the most deadly form of malaria in humans. According to the World Health Organization, an estimated 225 million cases of malaria infection were reported in 2009 with 781,000 resulting in death. Before the discovery of artemisinin, widespread use of the antimalarial chloroquine has lead to the emergence of drug resistant P. falciparum strains. In these resistant strains, one up-regulated and important, yet functionally unknown gene is the exported antigen 1 (EXP1), a human antigen and vaccine candidate that is exported to the parasital vacuole membrane and food vacuole. EXP1 is essential and may play an important role in malaria and in its human immune response. Here, we functionally predict and biochemically characterize EXP1 as a membrane glutathione S-transferase using a computational network method. EXP1 efficiently degrades cytotoxic hematin, is potently inhibited by artesunate, and is associated with artesunate metabolism and susceptibility in drug-pressured malaria parasites. These data implicate EXP1 in the mode of action of a frontline antimalarial drug. Contributors: Quiros, Joel*; Lisewski, Andreas*; Ng, Caroline; Adikesavan, Anbu; Miura, Kazutoyo; Putluri, Nagireddy; Eastman, Richard; Scanfeld, Daniel; Regenbogen, Sam; Altenhofen, Lindsey; Llinás, Manuel; Sreekumar, Arun; Long, Carole; Fidock, David; Lichtarge, Olivier 253 BAYLOR COLLEGE OF MEDICINE VOLTAGE-DEPENDENT ANION CHANNEL 2 (VDAC2)-BAK INTERACTION MODIFIES CELLULAR RESPONSE TO ER STRESS-INDUCED APOPTOSIS Adithya Raghavan Department of Molecular & Human Genetics Advisor: William Craigen, M.D./Ph.D.-Department of Molecular & Human Genetics Voltage-dependant anion channels (VDACs) are pore-forming ion-channel proteins located in the mitochondrial outer membrane (MOM). We are interested in the role of the mammalian VDAC isoform VDAC2 in apoptosis, since we have previously shown that VDAC2 directly interacts with and anchors the pro-apoptotic protein BAK to the MOM. Here, we report that in the absence of VDAC2, in addition to BAK being reduced in mitochondria, BAK levels in the endoplasmic reticulum (ER) are significantly increased. We further identified that upon prolonged treatment with ER stressors, there is a significantly reduced rate of ER stress-induced apoptosis in Vdac2-/- MEFs. A double knockout of Vdac2/Bak in MEFs restores ER-stress induced rate of cell death to wildtype (WT) levels, demonstrating a dominant negative role for ER-localized BAK in the process. In mice, while Vdac2-/- whole-body knockouts exhibit embryonic lethality, heart-specific conditional knock-outs develop cardiac fibrosis and cardiomyopathy. A double knockout of Bak and heart-specific Vdac2 rescues the aforementioned phenotypes. In order to further identify the potential mechanisms, we performed a microarray expression analysis between Wt and Vdac2-/- MEFs. We observed a consistent downregulation in the expression levels of key players in the ER stress response pathway in Vdac2-/- MEFs, confirmed by quantitative RT-PCR and immunoblotting. A double knockout of Vdac2/Bak increases expression of ER stress genes to levels comparable to Wt MEFs. Preliminary results from Vdac2-/- MEFs indicate impaired function of ER-stress response protein IRE1alpha, a known interactor of BAK in the ER. We propose that in the absence of VDAC2 there is loss of BAK in the MOM, accumulation of BAK in the ER, interactions of ER-localized BAK with ER stress response proteins, resulting in blunted transcription of ER stress response genes and a reduced rate of ER-stress induced apoptosis. Contributors: Raghavan, Adithya; Sheiko, Tatiana; Graham, Brett H.; Craigen, William C. 254 2014 GRADUATE STUDENT SYMPOSIUM A FORCE ASSAY TO MONITOR THE MOTILITY OF CANCER CELLS Vivek Rajasekharan Department of Molecular & Cellular Biology Advisor: Frederick Pereira, Ph.D.-Department of Molecular & Cellular Biology During local invasion in metastasis, cancer cells form migratory structures like filopodia, lamellipodia and invadopodia via actin remodeling, enabling them to produce protrusive forces required to breach the basement membrane and/or invade surrounding stroma. These forces are produced by actin networks through the growth of individual filaments against the cell membrane. There are two types of actin networks, bundled and branched. One of the migratory structures is a filopodium – a finger like projection that protrudes in all directions beyond the cell's edge; filopodia are filled with parallel rods of actin filaments bundled by cross-linking proteins. The specific role of filopodia in relation to carcinoma is not understood. We developed a force assay to measure the change in the axial membrane force that results from activity of actin polymerization and depolymerization at the tip of a filopodium-like structure formed from a cancer cell. We see force fluctuations, and we hypothesize that along with pulls (inverse sawtooths) due to actin polymerization, pushes (sawtooths) due to actin depolymerization, are also observed at the tip. We determine the force (F) and its direction (θ) and extract the stationary states (dF/dt=0), their dwell-times, the change in force between adjacent stationary states and the time it takes to go from one state to another, for all the transient stationary states in the system. Analysis of this information reveals characteristics of the actin biochemical network. We form the structure from HN31, head and neck squamous cell carcinoma cells, because they exhibit an H-Ras mutation that results in constitutively active RhoGTPases which activate the actin remodeling pathway. We will compare the characteristics of the networks for untreated cells, with cells treated with drugs that inhibit motor proteins like actin, microtubules and myosin, and by H-Ras knockdown as a control, to assess the role of actin remodeling in force generation. Contributors: Rajasekharan, Vivek; Sreenivasan, Varun; Yuan, Tao; Patel, Ameeta A; Myers, Jeffrey N; Pereira, Fred A; Farrell, Brenda; 255 BAYLOR COLLEGE OF MEDICINE THE ROLE OF LYSOSOMAL SIGNALING IN METABOLIC MORBIDITY ASSOCIATED WITH NEURODEGENERATION Prasanna Ramachandran Department of Molecular & Human Genetics/M.D.-Ph.D. Program Advisor: Meng Wang, Ph.D.-Department of Molecular & Human Genetics Neurodegenerative disorders are debilitating conditions characterized by the progressive degeneration or death of neurons. Although degeneration is limited to cells of the nervous system, manifestations of the disease often afflict distant peripheral tissues, and non-neuronal pathology contributes significantly to the morbidity and mortality of these disorders. The molecular mechanisms underlying these seemingly disparate findings remain unknown. Our research focuses on the neuronal regulation of peripheral metabolic processes, using the evolutionarily conserved Tubby protein as a model. Mutations in the Tub gene, which is expressed exclusively in neurons, result not only in retinal degeneration and neurosensory hearing loss, but also in morbid adult onset peripheral obesity and insulin resistance. Loss of function of the Caenorhabditis elegans homolog of Tub, tub-1, also results in analogous phenotypes of neurosensory deficit and lipid accumulation. In preliminary studies, we identified that neuronal deficiency of tub-1 gene leads to abnormal lysosomal biogenesis in the fat storage tissue. Interestingly, lysosomal dysfunction has been linked to neurodegeneration and metabolic morbidity. I hypothesize that tub-1 modulates lipid metabolism through a neuroendocrine regulatory axis of lysosomal function. To elucidate this novel regulatory axis, we will first identify the specific subset of neurons involved in modulating peripheral lysosomal content, using single-neuron ablation as well as optogenetic activation. We will also delineate the neuroendocrine signaling pathway that mediates the cell-nonautonomous effect on lysosomal function, through genetic screens, proteomic analysis and transcriptome profiling. Finally, we will identify lysosomal metabolic changes that are responsible for the neurodegenerative and lipid storage phenotypes in tub-1 mutants, using high- throughput metabolomic profiling of lysosomes. Contributors: 256 2014 GRADUATE STUDENT SYMPOSIUM INVESTIGATING THE FUNCTIONAL IMPACT OF AMINO ACID SUBSTITUTIONS ON THE HIGHLY PROFICIENT OROTIDINE DECARBOXYLASE BY RANDOMIZATION MUTAGENESIS Sam Julian Regenbogen Department of Pharmacology Advisor: Olivier Lichtarge, M.D./Ph.D.-Department of Molecular & Human Genetics Discerning the relationship between protein structure and function is one of the major goals of modern biology, and many methods have been devised to both predict and observe this relationship. We are using two methods to investigate the structure-function relationship of the E. coli enzyme orotidine-5’-monophosphate decarboxylase (ODCase): a computational method to predict the impact of specific mutations on protein function, and large-scale randomization to experimentally assay the functional effects of mutations. ODCase is an essential enzyme in the pyrimidine biosynthesis pathway and is found in all domains of life; it catalyzes the conversion of orotidine-5’-monophosphate (OMP) into uridine-5’-monophosphate (UMP). It is one of the most proficient enzymes known, catalyzing the decarboxylation of OMP at a rate 1017-fold faster than the uncatalyzed reaction. This has made ODCase an attractive target for structure-function studies, and yet, despite catalytic residues and overall structure that are virtually invariant among species, its mechanism is still not fully understood. We have used a computational method of predicting the functional effects of amino acid substitutions in ODCase and are currently undertaking an experimental method of investigating the real outcomes of the same mutations. We are designing mutagenic primers for each of the 245 codons in the pyrF gene (which encodes for E. coli ODCase), which replace the three nucleotides of each codon with NNS – N meaning A,T,G, or C; S meaning C or G. By performing site-directed mutagenesis with each of these NNS primer pairs, we will produce, for each codon, a library containing all possible permutations (all 20 amino acids have at least one possible codon represented by NNS). These libraries will then be transformed into an E. coli strain lacking pyrF, and selected on minimal media, which will only allow the growth of mutants that can synthesize UMP. These selected libraries will then be pooled and submitted for deep sequencing, allowing us to identify all single-residue substitutions that still maintain ODCase function. For each position in the ODCase protein sequence, we will be able to determine the relative effect on ODCase function of all 20 possible amino acids using their relative frequencies in the pooled sequence data. This information will provide a strong test for the predictions made computationally, and will provide further insight to the mechanistic requirements of this highly proficient enzyme. Contributors: Atri, Benu; Katsonis, Panagiotis; Palzkill, Timothy; Lichtarge, Olivier 257 BAYLOR COLLEGE OF MEDICINE NETWORK MODELING OF TRANSCRIPTIONAL RESPONSE TO INFLUENZA VACCINATION Alexander Renwick Program in Structural and Computational Biology and Molecular Biophysics Advisor: Chad Shaw, Ph.D.-Department of Molecular & Human Genetics Although genome wide gene expression data has become common in biomedical science, fully exploiting the multivariate character of the resource has remained a challenge. The data are frequently used as a massively parallel screening tool to identify individual gene candidates. When more systems level analyses are performed on data they are often introduced sequentially, where, for example, a list of differentially expressed genes is generated and then interrogated for pathway enrichment. These approaches fail to exploit the dependence structure in gene expression that can help to detect differential expression. Computational tools for more sophisticated analyses have recently been developed. One of these is the Gaussian Lasso, which applies the tools of sparse modeling to infer an undirected Bayesian Network. Such a network can be decomposed into expression modules, each of which can be analyzed as an ensemble. We have applied this method to the transcriptome response to influenza vaccine in human subjects. A systems-level approach is especially appropriate for this study because of the complexity of the process of immune response. We compare result of the network-based analysis to that of a univariate, gene-by-gene analysis. Our analysis verifies that interferon signaling system is important in the first day after vaccination. Our results highlight the similarities and differences as well as the additional power of the multivariate content analysis approach. Contributors: Renwick, Alexander; Belmont, John; Shaw, Chad 258 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF ESTROGEN IN METASTASIS Natalie Michelle Reyes Integrative Program in Molecular and Biomedical Sciences Advisor: Suzanne Fuqua, Ph.D.-Department of Medicine Background: Although 75% of breast tumors are ER(-postive, there are no ERpositive preclinical models which reliably metastasize to study the effects of hormones on invasion and metastasis. We have studied the role of estrogen in metastasis in an ER(-positive MCF-7 sub-line (MCF-7 Met) which has spontaneously acquired the ability to metastasize in vivo. Experimental design and methods: Microarray analysis was performed comparing MCF-7 Met to parental, non-metastatic MCF-7 cells. The differential gene expression was compared to genes differentially expressed when cells expressed K303R ER alpha. We performed soft agar growth assays, mammosphere assays, signal transduction assays and ERE luciferase assays. We injected MCF-7 Met and MCF-7 cells into athymic nude mice to study in vivo growth properties. Results: We found that the MCF-7 Met line shared 66% gene expression similarity with cells expressing K303R ER alpha. We also found that upon hormone treatment the MCF-7 Met cells had higher levels of pS118 and pS305 ER alpha, which was observed in cells expressing K303R ER alpha. The K303R ER alpha model also had an increased in anchorage independent growth under estrogen treatment and we observed similar results in the MCF-7 met line. The MCF-7 Met line was able to form more spheres during a mammosphere assay when compared to MCF-7. In preliminary results we also observed estrogen hypersentivity in the MCF-7 Met line during ERE luciferase assay, which is another phenotype that was observed in cells expressing the K303R ER alpha mutation. 94% of the mice injected with the MCF-7 Met line metastasized to distant sites regardless of hormonal treatment but their was no difference in primary tumor growth when treated with Tam or estrogen withdrawal. Conclusion: I have determined that many of the genes differentially expressed in MCF-7 Met cells are related to estrogen signaling and tamoxifen resistance using bioinformatic approaches. This suggests that the processes of invasion and metastasis may be influenced by the hormonal mileau. I have also determined that the MCF-7 Met line may share a similar phenotype to cells expressing the K303R ER alpha mutation. Although MCF-7 Met cells remain hormone-dependent in vivo, they have acquired the ability to metastasize, potentially suggesting that hormone resistance is not a prerequisite for ER-positive breast cancer cells to metastazise to distant sites. Contributors: Corona, Arnoldo; Covington, Kyle; Gu, Guowei 259 BAYLOR COLLEGE OF MEDICINE THE ONTOGENY OF CEREBROVASCULAR CRITICAL CLOSING PRESSURE IN HUMAN PREMATURE INFANTS Christopher J Rhee Clinical Scientist Training Program Advisor: Kenneth Brady, M.D.-Department of Anesthesiology Background Premature infants are at risk of vascular neurologic insults. Hypotension and hypertension are considered injurious, but neither condition is defined with consensus. Critical closing pressure (CrCP) is the arterial blood pressure (ABP) at which cerebral blood flow ceases. CrCP may serve to define subject-specific low or high ABP. Objective To quantify CrCP as a function of gestational age (GA). Methods Premature infants (n=179) with GA from 23–31 weeks had recordings of ABP and middle cerebral artery flow velocity twice daily for 3 days and then daily for the next 4 days during the first week of life. CrCP was calculated using an impedance-model derivation with Doppler-based estimations of cerebrovascular resistance and compliance. The association between GA and CrCP was determined in a multivariate analysis adjusting for hour of life (HOL), use of vasopressors, 5-minute Apgar score, and arterial carbon dioxide tension. Results The median [interquartile range] CrCP for the cohort was 22 mm Hg [19-25]. CrCP increased significantly with GA (r=0.6; slope = 1.4 mm Hg/week gestation), an association that persisted with multivariate analysis (p<0.0001). Higher GA was associated with increased CrCP. Conclusions CrCP increases significantly at the end of the second and beginning of the third trimester at a rate of 1.4 mm Hg per week of gestation. The low CrCP observed in very premature infants may explain their ability to tolerate low ABP without global cerebral infarct or hemorrhage Contributors: Rhee, Christopher; Fraser III, Charles; Kibler, Kathleen; Easley, Ronald; Czosnyka, Marek; Varsos, Georgios; Smielewski, Peter; Rusin, Craig; Brady, Ken; Kaiser, Jeffrey 260 2014 GRADUATE STUDENT SYMPOSIUM HYPERMETHYLATION DIMINISHES MSH5 EXPRESSION IN NOA MEN Alex David Ridgeway Department of Molecular & Cellular Biology Advisor: Dolores Lamb, Ph.D.-Department of Urology Non-obstructive azoospermia (NOA) is a severe form of male infertility that results in a complete absence of sperm in the ejaculate. While the molecular mechanisms underlying NOA remain incompletely understood, pathways crucial for spermatogenesis are believed to play an important role. MSH5 is a meiosis-associated gene implicated in the repair of double-stranded breaks and in the resolution of the Holliday junction - both key events during the meiotic division of spermatogenesis. MSH5 deficiencies in mice result in an infertile, NOA-like phenotype. Aberrant DNA methylation is a potential epigenetic cause for deficient MSH5 expression in humans. As such, we sought to identify and investigate the role of altered DNA methylation in men with NOA. We examined the DNA methylation profile of NOA men (n=31) and fertile controls (n=20) using the Illumina HumanMethylation450 array and bisulfite sequencing. A cohort of 6/31 NOA men were identified with significantly increased DNA methylation at 5 specific CpG-sites within exon 2 of MSH5. This cohort also exhibited decreased MSH5 mRNA and MSH5 protein levels. Removal of the aberrant hypermethylation, in vitro, using the demethylating agent 5-aza-2-deoxycytidine (5-Aza) resulted in an increase of MSH5 expression. Discontinuing 5-Aza treatment led to the reemergence of aberrant MSH5 methylation, suggesting a potential driving cis element/trans factor. In opposite effect, the targeted methylation of the identified CpG-sites using guide RNA’s, resulted in a reduction of MSH5 expression in a control cell line. Reduced expression of MSH5 also led to the loss of cellular proliferation and the ability to repair double stranded DNA breaks in response to treatment with the radiomimetic drug, neocarzinostatin. Taken together, our data shows that the hypermethylation of CpG sites within exon 2 of MSH5 deleteriously affects its expression. The presence of this hypermethylation in NOA patients and the resulting loss of MSH5 expression may be a significant contributor to their infertility. Contributors: 261 BAYLOR COLLEGE OF MEDICINE THE ROLE OF VITAMIN D METABOLISM IN PROSTATE AND PROSTATE CANCER CELLS Justin Michael Roberts Department of Molecular & Cellular Biology Advisor: Nancy Weigel, Ph.D.-Department of Molecular & Cellular Biology Epidemiological data suggests vitamin D may play a role in prostate cancer (PCa) prevention and data from our lab and others have demonstrated the active metabolite 1,25-dihydroxyvitamin D3 (1,25D) inhibits the growth of PCa both in vivo and in vitro. The main circulating form of vitamin D is 25-hydroxyvitamin D3 (25-OHD); it is converted to 1,25D, by 1α-hydroxylase (CYP27B1) in the kidney and other tissues like prostate. Normal prostate epithelial cells express CYP27B1 and are capable of converting 25-OHD into active 1,25D allowing pro-differentiating and growth regulatory functions to occur. Conversely, some PCa cell lines exhibit very low CYP27B1 activity resulting in low intracellular 1,25D synthesis insufficient to observe growth inhibitory effects in these cells. Preliminary data from our lab using RWPE-1 cells, an immortal non-transformed prostate cell line, confirms CYP27B1 expression and growth inhibition after 25-OHD treatment. In contrast, the VCaP PCa cell line expresses minimal CYP27B1 and is not growth inhibited after treatment with 25-OHD suggesting CYP27B1 is necessary for 25-OHD mediated growth inhibition. Loss of CYP27B1 expression has been suggested to be an early event in PCa progression that confers a growth advantage by reducing the ability of prostate cells to respond to circulating 25-OHD. This may also cooperate with the early formation of TMPRSS2:ETS fusions in PCa development. The enzyme 24-hydroxylase (CYP24) tightly regulates 1,25D by metabolizing it to a less active form. Our lab has shown ERG and 1,25D can cooperate to hyper-induce CYP24 in VCaP cells. Comparatively, VCaP cells, which contain the TMPRSS2:ERG fusion, express higher CYP24 levels than LNCaP PCa cells lacking this fusion. Preliminary data shows a time dependent decrease in 1,25D mediated gene induction in VCaP cells suggesting metabolism of 1,25D by CYP24. Clinical trials using 1,25D (calcitriol) have thus far been unsuccessful. Two reasons for this may be inactivation by CYP24 and hypercalcemia at high doses of calcitriol. We are testing nonsecosteroidal VDR agonists, which should be resistant to metabolism by CYP24 and have a larger safety range for hypercalcemia than calcitriol. Early data shows these VDR agonists inhibit the growth of both LNCaP and VCaP PCa cells in vitro and are resistant to metabolism by CYP24. Future studies will include assessing the ability of these VDR agonists to inhibit tumor growth in vivo. The loss of CYP27B1 activity coupled with increased CYP24 expression provides a model of early escape from vitamin D regulation by developing PCa cells. We aim to gain insight into the roles of CYP27B1 and CYP24 in cancer progression and how sensitivity to vitamin D metabolites is altered through deregulation of these enzymes in PCa. Contributors: Roberts, Justin; Kim, Jung-Sun; Weigel, Nancy 262 2014 GRADUATE STUDENT SYMPOSIUM SUMO CONJUGATING ENZYME UBC9 IS REQUIRED IN THE OOCYTE FOR FEMALE FERTILITY Amanda Rodriguez Department of Molecular & Cellular Biology Advisor: Stephanie Pangas, Ph.D.-Department of Pathology & Immunology Sumoylation is a dynamic post-translational modification process in which a SUMO (small-ubiquitin related modifier) moiety is conjugated to specific proteins involved in transcriptional regulation, protein transport, chromosome segregation and signal transduction via an enzymatic cascade and affects their protein subcellular localization, interactions, stability and activity. Although research has suggested that the SUMO pathway may play a crucial role during meiotic maturation in mice, the role of sumoylation in the oocyte has not been well characterized. A central component of the sumoylation cascade is the SUMO- conjugating enzyme UBC9, which is highly conserved across species and is essential for several developmental processes such as cell cycle regulation, chromosome segregation and cell viability. However, the connection between UBC9 and its potential function during oocyte development remains largely unknown. The overall goal of our project is to investigate the role of Ubc9 during oocyte development and to identify novel SUMO-target proteins in oocytes. We hypothesize that Ubc9 plays an essential role during oocyte development and loss of Ubc9 leads to ovarian defects and infertility. As conventional knockout Ubc9 leads to embryonic lethality, we generated conditional knockout mice with oocyte- specific deletion of Ubc9 (OoUbc9 cKO) by using the growth differentiation factor 9 (Gdf-9) iCre driver. Our preliminary fertility analysis revealed that targeted deletion of Ubc9 in oocytes leads to sterility in female mice. To investigate how the loss of UBC9 causes sterility, folliculogenesis, oocyte maturation and postnatal ovarian development will be assessed. In addition, we will identify target proteins sumoylated by UBC9 in oocytes and characterize their functions in fertility regulation. By identifying the causes of infertility in OoUbc9 cKO mice we will gain insight to the role of sumoylation in oocyte development and ovarian function. Contributors: Rodriguez, Amanda; Tripurani Swamy; Pangas, Stephanie 263 BAYLOR COLLEGE OF MEDICINE NEO HDL CHOLESTERYL ESTER TRANSFER ACTIVITY TO SERUM LIPOPROTEINS Perla Janet Rodriguez Integrative Program in Molecular and Biomedical Sciences Advisor: Henry Pownall, Ph.D.-Department of Medicine Serum opacity factor (SOF), a protein produced by S. pyogenes causes clouding of human plasma. In vitro, an active truncated recombinant (r) SOF catalyzes the disproportionation of plasma high density lipoproteins (HDL), its sole target, giving lipidfree apo A-I, a small neo HDL that is apo A-II-rich and cholesterol poor, and a cholesteryl ester rich microemulsion (CERM). In vitro tests showed that the neo HDL supports several reverse cholesterol transport (RCT) steps and has better atheroprotective qualities than the HDL from which it was derived: neo HDL is better as a substrate for lecithin:cholesterol acyltransferase (LCAT), as an acceptor of macrophage cholesterol efflux, and as an anti-inflammatory lipoprotein. Mice injected a low dose (4 g) rSOF had reduced plasma cholesterol ~50% in three hours. Size exclusion chromatography reveals that the CERM is prominent in the plasma profile in vivo. However, lipid-free apo A-I and neo HDL in vivo occur at <1% of their in vitro levels. The in vivo fate of neo HDL is not yet known. Our current hypothesis is that neo HDL fuses to HDL and is subsequently taken up by the HDL receptor SR-BI. To determine the catabolism of neo HDL, its nontransferable cholesteryl ester (CE) was labeled with tritium. Neo HDL-CE [3H] was incubated with human plasma and each isolated lipoprotein, with or without lipoprotein deficient serum (LPDS) which contains lipoprotein modifying enzymes cholesteryl ester transfer protein (CETP), phospholipid transfer protein (PLTP), and LCAT. Over time, CE transferred to all lipoproteins in the presence of LPDS. Neo HDL-CE transferred to HDL in the absence of LPDS but not to the other lipoproteins. In mouse plasma, the CE transferred to HDL at a much faster rate. These results suggest a potential fusion between neo HDL and HDL. Torcetrapib, a CETP inhibitor, was added to similar experiments which resulted in partial inhibition of neo HDL-CE transfer to all lipoproteins suggesting its involvement in neo HDL catabolism. Neo HDL-CE [3H] uptake studies were performed using three CHO cell lines, two of which express low endogenous levels of SR-BI and one overexpressing murine SRBI. CE uptake from neo HDL only occurred when SR-BI was over expressed. CE uptake experiments will be repeated in human hepatocytes using radiolabeled forms of Neo HDL. Together these results suggest that neo HDL may fuse to HDL and that neo HDL may potentially interact with the SR-BI receptor. To further investigate the role of plasma factors in neo HDL catabolism inhibitors targeting LCAT and PLTP will be used similarly. Future studies include injecting mice with radiolabeled neo HDL to effectively track its destination in vivo. Contributors: Rodriguez, Perla; Gillard, Baiba; Rosales, Corina; Pownall, Henry 264 2014 GRADUATE STUDENT SYMPOSIUM CONTRIBUTION OF EUKARYOTIC CHAPERONIN TRiC/CCT TO BIOGENESIS AND FOLDING OF A LEUKEMIC ONCOGENIC TRANSCRIPTION FACTOR, AML1-ETO Soung Hun Roh Department of Biochemistry & Molecular Biology Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology David Tweardy, M.D.-Department of Medicine Acute Myeloid Leukemia (AML) is the most severe type with 40% frequency in total leukemia and characterized by non-random chromosomal translocation giving rise to oncogenic fusion genes. One of the most frequent oncogenic fusion protein is AML1ETO (12%) resulting from the translocation at (8;21)(q22;q22). AML1-ETO is hard to be managed by conventional drug development strategies since it does not mediate its function by ligand-binding or enzyme activity, where are eligible drug-targeting sites by small molecules. In order to control this undruggable oncoprotein, a novel approach of altering the oncoprotein proteostasis network through type II eukaryotic chaperonin, TCP-1 Ring Complex (TRiC/CCT), has been suggested. Our study focuses on the functional relationship between TRiC and AML1-ETO and exploring the discrete interaction mechanism between oncoproteins and chaperonin. In this study, Co-IP results showed AML1-ETO is a TRiC client and its folding intermediate directly associates to TRiC through the DNA binding domain (AML_1~175). Chemical crosslinking and cryo-EM study also revealed that multiple TRiC subunits (CCT4, 6) bind cooperatively to AML1-ETO. More interestingly, total expression level of AML1-ETO is dramatically decreased in vitro translational system when endogenous TRiC was immune-depleted, which suggests not only TRiC is required for AML1-ETO’s biogenesis but also translational AML1-ETO-TRiC interaction is a potential drug target to control Acute Myeloid Leukemia. Contributors: Soung-Hun, Roh; Tweardy, David J; Chiu, Wah 265 BAYLOR COLLEGE OF MEDICINE RETINITIS PIGMENTOSA AND NONSENSE-MEDIATED DECAY IN MOUSE PHOTORECEPTORS Ramon Roman-Sanchez Integrative Program in Molecular and Biomedical Sciences Advisor: John Wilson, Ph.D.-Department of Biochemistry & Molecular Biology Retinitis pigmentosa (RP) is an inherited degenerative disease of the retina that results in rod and cone photoreceptor death. Initially as rods degenerate, subjects lose peripheral and night vision; this is followed by cone photoreceptor degeneration leaving the patients completely blind in some instances. RP affects 1 in every 4000 individuals worldwide and around 30-40% of these are the result of autosomal-dominant (ADRP) mutations. Rhodopsin (Rho) is a G protein-coupled receptor of the rods and it initiates the phototransduction cascade upon photon capture. Mutations in Rho, which account for about 25% of all ADRP cases, affect many processes, including Rho posttranslational modifications, transport, folding and signaling capabilities. More than 150 mutations that result in RP have been identified within Rho. Seven of these are nonsense mutations of which five cause dominant RP (dRP) and the two cause recessive RP (rRP). One possible explanation for this difference is that rRP mutants undergo nonsense-mediated mRNA decay (NMD) whereas dRP do not, thereby producing a toxic Rho truncation. The NMD pathway is the cell’s quality control mechanism for the detection and degradation of aberrant mRNA transcripts. If an mRNA molecule contains a premature termination codon (PTC), defined as a stop codon ~50nt upstream of an exon-exon junction, then degradation will ensue by NMD. Our lab has found that the transcript of Rho-Q64X, which causes dRP in humans, is present a similar levels to wild type Rho in a mouse model. The same observation was made in a mouse model with a duplicated exon 2 that results in a PTC at the exon-exon junction. One study in cells detected degradation by NMD of the Rho Q249X nonsense mutant that causes rRP, but in vivo studies are absent. These observations raise the question of whether Rho is insensitive to NMD in photoreceptors. I am studying Rho nonsense transcripts and NMD in the context of the retina so as to have a better understanding of RP, its mechanisms and aid in the development of gene therapy strategies based on suppression and replacement of the mutant Rho. Contributors: Roman-Sanchez, Ramon; Wilson, John H 266 2014 GRADUATE STUDENT SYMPOSIUM EFFICIENT APICAL TRANSDUCTION OF POLARIZED HUMAN AIRWAY EPITHELIUM BY ADENOVIRUS SEROTYPE 3 Amanda Nicole Rosewell Shaw Department of Molecular & Human Genetics Advisor: Philip Ng, Ph.D.-Department of Molecular & Human Genetics Viral-based Cystic Fibrosis (CF) gene therapy has not, to date, been successful because the receptors for the most widely used vectors are not accessible via the apical surface of the airway epithelium. Indeed, the major receptor for adenovirus serotype 5 (Ad5), CAR (coxackie-adenovirus receptor), is sequestered to the basolateral surface by tight junctions. Ad5 can efficiently transduce the airway epithelium only if the tight junctions are transiently disrupted by agents such as LPC or EGTA. We have found that administration of these agents into nonhuman primate lungs cause temporary but significant increases in respiratory resistance and decreases in compliance measurements. Thus, opening tight junctions may be unsuitable for CF gene therapy considering the compromised pulmonary status of CF patients with advanced disease. Consequently, identification of vectors that transduce via an apical surface receptor is needed to obviate the need for opening tight junctions. Recently, it was reported that adenovirus serotype 3 (Ad3) could mediate apical infection of various polarized epithelial cells using Desmoglein-2 as its receptor. This observation suggested that Ad3 might be useful for apical infection of airway epithelial cells for CF gene therapy. To evaluate this potential, we compared the transduction efficiency of Ad5 versus Ad3 in polarized human bronchiolar epithelium (HBE). We found that Ad3 transduces polarized HBE at least 10-fold more efficiently than Ad5. Indeed, transduction of polarized HBE by Ad3 alone was comparable to transduction by Ad5 only if the Ad5 infection was preceded by opening tight junctions with EGTA. These results suggest that vectors based on Ad3 may be useful for CF gene therapy because highly efficient transduction of the airway epithelium may be achieved without the need to open tight junctions. We have constructed a novel Helper-Dependent Ad3 vector by creating a replication incompetent Ad3 helper virus. Contributors: Rosewell Shaw, Amanda; Palmer, Donna; Ing, Jordan; Grove, Nathan; Ng, Philip 267 BAYLOR COLLEGE OF MEDICINE HIGH-THROUGHPUT METHODS FOR PROBING THE HUMAN VIROME Matthew Clayton Ross Department of Molecular Virology & Microbiology Advisor: Joseph Petrosino, Ph.D.-Department of Molecular Virology & Microbiology Comprising an estimated 1031 total particles, viruses are the most diverse and numerous biological entities on Earth. However, estimates of the number of viruses inhabiting the human body, the human virome, are shots in the dark at best. But we can be certain we have explored little of the virome by sequence, and even less by function. Fortunately, the field of viral metagenomics has evolved rapidly over the past few years owing mainly to breakthroughs in next generation sequencing. We now have the ability to probe viral communities from virtually any source, without the need for culture. Due to high interindividual variation, studies of the human microbiome, including the virome, require large numbers of samples to accomplish significant statistical power. Large-scale projects often require special considerations from a sample-processing standpoint. For example, concentration of large volumes of primary sample is not feasible for projects containing tens-of-thousands of samples, and often, specimens collected in the clinic as part of routine visits are very low biomass. Additionally, high-level multiplexing is a requirement to keep large projects within reasonable timelines and budgets. Our aim was to develop a high-throughput method to probe the human virome suitable for projects with thousands or tens-of-thousands of clinical samples, that is highly-multiplexable, without sacrificing significant sensitivity compared to whole genome shotgun (WGS) sequencing. Here we report a viral metagenomics sample preparation method that is highly sensitive, highly scalable and delivers significant cost cutting at the stage of sequencing library preparation. Through a combination of semirandom primer amplification and molecular barcoding, we are able to pool tens to hundreds of samples prior to sequencing, allowing for a single library to be created for a pool of samples instead of one library per sample. We demonstrate sensitivity through the use of a contrived mock viral community, and show that we do not introduce significant bias through pooling. Ultimately, this technique affords the ability to interrogate the human virome in a manner highly conducive to large scale microbiome projects. Contributors: Ross, Matthew; Wong, Matthew; Holder, Michael; Metcalf, Ginger; Gibbs, Richard; Petrosino, Joseph 268 2014 GRADUATE STUDENT SYMPOSIUM DICTYOSTELIUM DISCOIDUEM SOCIALITY DURING GROWTH WITH GRAMNEGATIVE BACTERIA Michelle Jenny Rubin Integrative Program in Molecular and Biomedical Sciences Advisor: Gad Shaulsky, Ph.D.-Department of Molecular & Human Genetics Dictyostelium discoideum is a eukaryotic amoeba that consumes bacteria during growth. Upon starvation, Dictyostelium cells cooperate with one another while developing into multicellular structures in which some of the cells sacrifice themselves while supporting the sporulation of the others. Currently, sociality is defined as intercellular interactions during Dictyostelium development, but we propose that social interactions occur during vegetative growth as well. We have generated many Dictyostelium mutants that do not grow on Gram-negative bacteria and we tested the cell-autonomous properties of the phenotype. Interestingly, when we mixed just 10% wild type Dictyostelium cells with one of the mutants, we observed that the mutant grew on Gram-negative bacteria. Additionally, when we plated the mutant at different cell densities on Gram-negative bacteria, the cells were able to grow at high cell densities. Additionally, we found that Dictyostelium cells secret a soluble factor, most likely a polypeptide that kills bacteria. Therefore, we hypothesize that Dictyostelium growth on bacteria is a social process that is mediated by cooperative predation. We will determine whether social interactions take place during growth on Gram-negative bacteria and we will use biochemical and genetic tools to determine the identity and the roles of the secreted factors in the process. Contributors: Rubin, Michelle; Shaulsky, Gad 269 BAYLOR COLLEGE OF MEDICINE DATA-DRIVEN MODELING OF MAMMARY DUCTAL ELONGATION Ingrid Sophie Runquist Department of Molecular & Cellular Biology Advisor: Michael Lewis, Ph.D.-Department of Molecular & Cellular Biology The Terminal End Bud (TEB) at the growing tip of mammary ducts is one of the fastest growing structures in mammals. TEBs drive ductal elongation during puberty and regress once development of the ductal tree is complete. Successful modeling of this structure may yield insight into breast development as well as cancer progression. Previous mathematical models have focused on branching morphogenesis, but there are currently no models that address ductal elongation itself. Our model exploits the constrained geometry of the TEB in vivo which provided the framework for an initial mathematical model. Parameters in this model were then informed with measureable data (morphological characteristics, proliferation rate, cell cycle duration, and apoptosis). These data were then used to calculate a value representing the movement of cells from the TEB into the mature duct (termed the flux) and this calculated flux was then used to predict a linear elongation rate. The prediction was compared to an experimentally measured TEB displacement rate and then underwent rounds of revision and validation. Our initial measurements of proliferation, apoptosis, and cell sizes, predicted a linear displacement rate of 1.39 mm per day, which was faster than our experimentally measured displacement rate of 0.54mm per day. We then refined our model by incorporating changes in the direction of growth due to bifurcation, a cost function for bifurcation (which describes duplication of the TEB), as well as an additional flux term to account for a migration of cap cells into the body cell layer. Iteration of the revised mathematical model yielded an estimate significantly closer to the measured displacement rate, thus indicating that the most relevant biological parameters have been accounted for. In addition, our data overturned a long held belief that cap cells contribute to the body cell lineage. We are now poised for in silico experiments that may yield predictions consistent with cancer phenotypes, as well as predictions that recapitulate known mutation phenotypes. Contributors: Chauviere, Arnaud; Landua, John; Sreekumar,Amulya; Cristini, Vittorio; Rosen, Jeffrey; Lewis, Michael 270 2014 GRADUATE STUDENT SYMPOSIUM OVERCOMING CHALLENGES IN SHORT READ DATA ANALYSIS FOR MICRORNA AND SH-RNA NEXT GENERATION SEQUENCING Eric Benjamin Rutledge Program in Structural and Computational Biology and Molecular Biophysics/M.D.-Ph.D. Program Advisor: Zhandong Liu, Ph.D.-Department of Pediatrics As next-generation sequencing (NGS) technology continues to improve, our ability to explore new parts of the genome increases. In order to study complex, genomic elements, new bioinformatic approaches must be developed to accurately and reliably analyze fast-growing NGS databases. Here we focus on profiling small, non-coding RNA elements microRNA and shorthairpin RNA (shRNA) from high-throughput RNA sequencing. Both microRNA and shRNA are short, non-coding RNAs that mediate translational suppression of target mRNAs through imperfect sequence homology. Profiling microRNA expression levels is of widespread interest because of their pivotal roles in all biological processes. Synthetic microRNAs can be introduced via shRNA infection and play a pivotal role in developing drugs and understanding pathological disease pathways from RNA interference (RNAi) screens. The biology of microRNAs and shRNAs is challenging to expression profiling with NGS microRNA-sequencing because it requires experimental modifications that must be accounted for in downstream data analysis. We hypothesize that minimizing contamination from adapter sequences will enhance the mappabiltiy of short reads generated from high-throughput microRNA sequencing and improve accuracy in measuring changes in miRNA expression levels. Developing a pipeline to reliably can increase the accuracy of microRNA expression profiling and accurately measure miRNA expression level changes from microRNA-seq data. We have identified sources of variability by comparing the performance of algorithms with different approaches to adapter sequence removal, mapping, and annotation using a control set with known levels of spiked-on microRNA. We intend to standardize a pipeline and demonstrate applicability to biologically complex conditions by applying it to TCGA Ovarian cancer datasets and evaluating correlation with the wellestablished Agilent microarray expression profiling. Furthermore, we intend to improve short read data analysis by developing a pipeline to increase accuracy in identifying targets in RNAi screens using local alignment mapping algorithms. Previously, the intractable computational time required for local alignment hindered its use short read data analysis. We have utilized the accuracy and precision of the Smith-Waterman algorithm to develop a robust pipeline in for shRNA expression profiling. Contributors: Rutledge, Eric 271 BAYLOR COLLEGE OF MEDICINE MOUSE RETINAL GANGLION CELLS ALTER THEIR SPATIOTEMPORAL PROPERTIES TO ENCODE VISION IN DIM LIGHT SETTINGS Jasdeep Singh Sabharwal Department of Neuroscience/M.D.-Ph.D. Program Advisor: Samuel Wu, Ph.D.-Department of Ophthalmology The retina must encode visual activity in dim starlight as well as bright sunlight. In order to accomplish this feat the retinal circuitry is organized into two parallel pathways originating from rods or cones for dim or bright light conditions, respectively. Each of these pathways has unique properties that are passed onto the retinal ganglion cells (RGC), the output cells of the retina. By using a multielectrode array (MEA) to record from many RGCs and probing with stimuli at different light intensities we find that RGCs slow their temporal tuning and widen their spatial tuning in dim light. These changes allow the RGC to encode light over longer time and wider space which is critical in low light settings because fewer photons are present. Identifying how RGC modify their properties will shed light on normal visual encoding and also improve our understanding of diseases that damage RGCs and cause deficit in dim light settings, like glaucoma. Contributors: Sabharwal, Jasdeep; Cowan, Cameron; Wu, Sam 272 2014 GRADUATE STUDENT SYMPOSIUM COLLAGEN METABOLISM BIOMARKERS AND HEALTH RELATED QUALITY OF LIFE IN PULMONARY ARTERIAL HYPERTENSION Zeenat Safdar Clinical Scientist Training Program Advisor: Mark Entman, M.D.-Department of Medicine Objectives. The goal of this study was to investigate the relationship between peripherally measurable collagen metabolism biomarkers and health related quality of life (HRQoL) in PAH patients. Methods. We prospectively enrolled 68 stable idiopathic, anorexigen-associated, and hereditary PAH subjects and 37 healthy controls with similar distributions of age and gender. Serum samples were analyzed for N-terminal propeptide of type III procollagen (PIIINP), c-terminal telopeptide of collagen type I (CITP), matrix metalloproteinase 9 (MMP-9) and tissue inhibitor of metalloproteinase 1 (TIMP-1). The Minnesota Living with Heart Failure (MLWHF), EQ-5D-3L (EQ-5D) and Short Form (SF36) general health survey were administered at the time of blood draw. Univariate and multivariable linear regression models were used to assess associations between variables. Results. Mean age of PAH patients and controls were similar (45±15 and 49±14 years respectively, mean±SD, p=0.18). The six-minute walk distance, Borg dyspnea score, WHO functional class, presence of edema, brain naturiectic peptide were all significantly different between controls and PAH patients (p<0.01 for all). CITP, PIIINP, MMP9, and TIMP1 levels, and all HRQoL domains were significantly different between controls and PAH patients (p<0.001 for each). Several of the CAMPHOR scores strongly linearly associated with PIIINP. PIIINP levels were significantly associated with MLWHF physical (coef=1.63, and p=0.02), SF-36 physical (coef=-2.62, p=0.007), and EQ-5D aggregate (coef=0.21, p=0.03) scores also correlated well with PIIINP. An ROC curve analysis was conducted for six-minute walk test distance with a threshold of 330 meters, using PIIINP as the predictor variable. Our result show that for each unit increase in PIIINP the odds of a patient obtaining a distance of ≥330 decreases by 38% (OR=0.62; 95% CI=0.43, 0.90; p=0.011). The area under the curve was 0.8142 and the PIIINP cutoff that gave the highest sensitivity and specificity was 4.63 ug/ml. Conclusions. Of all biomarkers we tested, PIIINP is the best predictor of disease severity, and is strongly related to HRQoL scores in PAH patients. These relationships suggest PIIINP as a promising tool for PAH clinicians to determine or confirm the level of disease severity. Contributors: Safdar, Zeenat; Tamez Emilio; Frost, Adaani; Guffey, Danielle; Minard, Charles; Entman Mark 273 BAYLOR COLLEGE OF MEDICINE INVESTIGATING THE PHARMACOGENETIC PROPERTIES CONFERRED BY THE VARIANTS OF NT5C1A, A 5’ NUCLEOTIDASE Jason Saliba Department of Molecular & Human Genetics Advisor: Sharon Plon, M.D./Ph.D.-Department of Pediatrics We use whole exome sequencing, bioinformatics analysis, and functional assays to discover and assess potential cancer susceptibility mutations in families segregating risk for pediatric lymphocytic leukemia and lymphoma. In one kindred with four individuals diagnosed during childhood with lymphocytic leukemia or lymphoma transmitted in an autosomal dominant pattern of inheritance, exome sequencing on the constitutional DNA of three of the affected individuals and an intra-familial control was performed. Exclusion of common variants seen in population databases and variants in the control, along with systematic bioinformatics, narrowed our list to eight heterozygous missense variants shared among all three patients predicted to be functionally important. Within this list, the L254P variant occurs at a highly conserved residue of Human Cytosolic 5’ Nucleotidase 1A (NT5C1A), which is involved in the adenosine metabolic pathway. NT5C1A’s primary function is to catalyze the production of adenosine by the dephosphorylation of AMP. In addition, NT5C1A can dephosphorylate chemotherapeutic nucleoside analogs used in the treatment of leukemia. Prior studies have shown overexpression of wild type NT5C1A in HEK293 cells improved cell survival after treatment with purine and pyrimidine analogs. To test the functional effect of the L254P variant, HEK293 cell lines that stably overexpress wild type or L254P NT5C1A were created. These cells underwent cytotoxicity assays after treatment with various nucleoside analogs. We demonstrated that cells overexpressing wildtype, but not L254P mutant NT5C1A are at least ten fold more resistant to nucleoside analog chemotherapy agents indicating L254P interferes with enzymatic activity without evidence for change in substrate specificity. In our search for a cancer susceptibility gene, we found a gene with potentially impactful pharmacogenetic effects. Recently, another 5’ nucleotidase, NT5C2, was shown to contain somatic mutations that activated the protein and ultimately resulted in tumor resistance to nucleoside analogs in ALL relapse patients. Oncomine contains data showing NT5C1A is overexpressed in a variety of tumor types. According to the NHLBI exome variant server, very few common mutations of NT5C1A occur within or near the protein’s three functional domains. We hypothesized that other variants seen within NT5C1A could lead to altered drug metabolism. We created multiple stable cell lines expressing 8 different NT5C1A variants reported in the NHLBI exome variant server by using Gateway® technology and a lenitviral vector delivery system. These overexpression cell lines are being assessed by cytotoxicity assays for their response to nucleoside analogs. This is the first systematic analysis of naturally occurring variants of any 5’ nucleotidase with regard to their ability to process chemotherapy drugs. NT5C1A could be another 5’ nucleotidase whose expression and sequence should be assessed in tumors in order to determine impact on chemotherapeutic drug responses. Contributors: Saliba, Jason; Zabriskie, Ryan; Powell, Bradford; Hicks, Stephanie; Kimmel, Marek; Cheung, Hannah; Ritter, Deborah; Muzny, Donna M; Reid, Jeffrey G; Wheeler, David A; Gibbs, Richard A; Plon, Sharon E 274 2014 GRADUATE STUDENT SYMPOSIUM NEXT-GENERATION SEQUENCING AND NOVEL VARIANT DETERMINATION IN A COHORT OF 92 FAMILIAL EXUDATIVE VITREORETINOPATHY PATIENTS Jason Scott Salvo Program in Structural and Computational Biology and Molecular Biophysics Advisor: Rui Chen, Ph.D.-Department of Molecular & Human Genetics Familial exudative vitreoretinopathy (FEVR) is a developmental disease that can cause visual impairment and retinal detachment at a young age. Four genes involved in the Wnt signaling pathway were previously linked to this disease: NDP, FDZ4, LRP5, and TSPAN12. Identification of novel disease causing alleles, allows for a deeper understanding of the disease, better molecular diagnosis, and improved treatment. Sequencing libraries from 92 FEVR patients were generated using a custom capture panel to enrich for 163 known retinal disease causing genes in humans. Samples were processed using next generation sequencing (NGS) techniques followed by data analysis to identify and classify single nucleotide variants and small insertions and deletions. Sanger validation and segregation testing were used to verify suspected variants. Of the cohort of 92, 45 patients were potentially solved (48.9%). Solved cases resulted from the determination of 49 unique mutations, 41 of which are novel. 13 of the novel variants discovered were highly likely to cause FEVR due to the nature of these variants (frameshifting indels, splicing mutations, and nonsense variants types). This is the largest study of a FEVR cohort utilizing NGS that we are aware of. We were able to determine probable disease causing variants in a large number of FEVR patients, the majority of which were novel. Knowledge of these variants will help to further characterize and diagnose FEVR. Contributors: Salvo, Jason; Xu, Mingchu; Lyubasyuk, Vera; Wang, Hui; Wang, Feng; Nguyen, Duy; Wang, Keqing; Luo, Hongrong; Wen, Cindy; Shi, Catherine; Lin, Dannin; Zhang, Kang; Chen, Rui 275 BAYLOR COLLEGE OF MEDICINE RECRUITMENT OF CD34D FIBROBLASTS IN TUMOR-ASSOCIATED REACTIVE STROMA: THE REACTIVE MICROVASCULATURE HYPOTHESIS Rebeca San Martin Department of Molecular & Cellular Biology Advisor: David Rowley, Ph.D.-Department of Molecular & Cellular Biology Reactive stroma co-evolves with cancer, exhibiting tumor-promoting properties. It is also evident at sites of wound repair and fibrosis, playing a key role in tissue homeostasis. The specific cell types of origin and the spatial/temporal patterns of reactive stroma initiation are poorly understood. In this study, we evaluated human tumor tissue arrays by using multiple labeled, quantitative, spectral deconvolution microscopy. We report here a novel CD34/vimentin dual-positive reactive fibroblast that is observed in the cancer microenvironment of human breast, colon, lung, pancreas, thyroid, prostate, and astrocytoma. Recruitment of these cells occurred in xenograft tumors and Matrigel plugs in vivo and was also observed in stromal nodules associated with human benign prostatic hyperplasia. Because spatial and temporal data suggested the microvasculature as a common site of origin for these cells, we analyzed microvasculature fragments in organ culture. Interestingly, fibroblasts with identical phenotypic properties and markers expanded radially from microvasculature explants. We propose the concept of reactive microvasculature for the evolution of reactive stroma at sites of epithelial disruption common in both benign and malignant disorders. Data suggest that the reactive stroma response is conserved among tissues, in normal repair, and in different human cancers. A more clear understanding of the nature and origin of reactive stroma is needed to identify novel therapeutic targets in cancer and fibrosis. (Am J Pathol 2014, 184: 1860e1870) Contributors: Rebeca San Martin, David A. Barron,* Jennifer A. Tuxhorn, Steven J. Ressler, Simon W. Hayward, Xiaoyun Shen,Rodolfo Laucirica, Thomas M. Wheeler, Carolina Gutierrez, Gustavo E. Ayala, Michael Ittmann, and David R. Rowley 276 2014 GRADUATE STUDENT SYMPOSIUM UNCOVERING THE TARGETOME OF A DEVELOPMENTAL MASTER REGULATOR IN DICTYOSTELIUM Balaji Santhanam Program in Structural and Computational Biology and Molecular Biophysics Advisor: Gad Shaulsky, Ph.D.-Department of Molecular & Human Genetics Multicellular development is achieved by the coordinated action of many transcriptional regulators and their associated regulatory modules. The master regulator GtaC, a ‘GATA’-family transcription factor is essential for morphological progression in the social amoeba Dictyostelium discoideum. Despite its obvious importance, little is known about the network modules regulated by GtaC. We characterized its function during development using genetic perturbation experiments and quantitatively measured the physiological states of these different strains through transcriptional profiling. Our results indicate that the strain carrying a mutated DNA-binding GtaC phenocopies gtaC-null, both morphologically and transcriptionally. We assayed for the developmental genomic occupancy of both the wild-type and mutant forms of GtaC and our results indicate that the mutant form has decreased propensity to bind DNA. Further, our results allude to positional but not temporal concurrence of their binding events. Finally, we adopted an integrative approach to combine both binding and transcriptome data to uncover the developmental targetome of this master transcription factor. Our findings shed novel insight in to GtaC-controlled components underlying the gene regulatory network orchestrating Dictyostelium development. Contributors: Katoh-Kurasawa, Mariko; Cai, Huaqing; Devreotes, Peter N; Kuspa, Adam; Shaulsky, Gad 277 BAYLOR COLLEGE OF MEDICINE MIR-130B TARGETS ARHGAP1 INCREASING CDC42 ACTIVITY AND METASTATIC POTENTIAL IN EWING SARCOMA CELLS Laura Louise Satterfield Integrative Program in Molecular and Biomedical Sciences Advisor: Jason Yustein, M.D./Ph.D.-Department of Pediatrics Osteosarcoma (OS) and Ewing’s sarcoma (EWS) are the most common bone tumors in the pediatric population. While significant progress has been made with towards increasing patient outcomes, our success in eradicating these two malignances is significantly less than most other pediatric malignancies. One reason for continued treatment failure is due to high risk disease states such as the presence of metastatic lesions. Metastasis is of the utmost clinical relevance as it accounts for more than 90% of solid tumor deaths. Improvements in treatment regimens have been stagnant over the past 20-30 years most likely due to our lack of understanding their molecular pathogenesis. Overall survival rates are approximately 65-70% for localized disease and less than 20% for aggressive, disseminated states. Thus, the clinical need to understand high risk disease, such as metastasis is significant in order to develop novel therapeutics. miRNAs are highly conserved short 18-22 single stranded non-coding RNAs that have the ability to repress target translation and affect key cell signaling pathways often deregulated in cancer. Recently, miRNAs have been implicated in numerous cancers; however the functional role of miRNAs in sarcomas is limited. In order to identify novel alterations in miRNAs, we have utilized our analysis of microRNA expression from localized and metastatic tumors derived from novel genetically engineered mouse models of osteosarcoma. Among the miRNAs most significantly dysregulated, miR-130b was shown to be significantly upregulated in the metastatic lesions. Furthermore, it has been shown that miR-130b is overexpressed in OS and EWS patient samples, and this event is significantly associated with poor patient outcome in the latter sarcoma. Furthermore, we have verified miR-130b overexpression in both mouse and human sarcoma cell lines and in metastatic EWS tumor samples obtained from Texas Children’s Hospital. Functional analysis shows overexpression of miR-130b increases migration and invasion in vitro In order to identify novel targets of miR-130b contributing to metastasis, we performed gene expression analysis. We identified Cdc42GAP as a novel target of miR-130b. Cdc42GAP is a negative regulator of Cdc42 which hydrolyzes the active GTPase. ELISA analysis of Cdc42 activity reveals that Cdc42 activity is increased with overexpression of miR-130b.Further microarray and pathway analysis suggest a mechanism by which activation of Cdc42 leads to positive regulation of the MAPK cascade. Subsequent western blot analysis reveals that overexpression of miR-130b leads to the activation of SAPK/JNK and subsequent phosphorylation of c-JUN suggesting activation of AP-1 transcription factor. Taken together, these findings suggest that overexpression of miR-130b promotes sarcoma cell migration and invasion through targeting of Cdc42GAP and modulation of the Cdc42 and SAPK pathways. Contributors: Satterfield Laura, Kurenbekova Lyazat, Donehower Lawrence, Yustein Jason 278 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF CONSTITUTIVE ANDROSTANE RECEPTOR AND Β-CATENIN IN HEPATOCELLULAR CARCINOMA Jessica Diane Scott Integrative Program in Molecular and Biomedical Sciences Advisor: David Moore, Ph.D.-Department of Molecular & Cellular Biology Hepatocellular carcinoma is a devastating liver cancer that is on the rise in the developed world. Constitutive Androstane Receptor (CAR) is a nuclear receptor which is located primarily in the liver and mediates xenobiotic response. CAR is also essential to mouse liver tumor development induced by phenobarbital, a CAR agonist. Recently, our lab has demonstrated that the administration of the CAR inverse agonist androstanol causes reduction in tumor size and number. In 80% of tumors induced by phenobarbital and the genotoxic carcinogen diethylnitrosamine (DEN), β-catenin is mutated, while tumors induced by DEN alone do not contain β-catenin mutations. This suggests that βcatenin and CAR may cooperate to induce liver hyperproliferation and tumor formation. To test this hypothesis, we activated CAR and β-catenin simultaneously in mice. Doing so results in hepatomegaly in the short term, and tumor development in the long term. Activating either component alone results in transient, relatively mild liver growth. In CAR-activated liver cells, high polyploidy serves as a proliferation checkpoint. In livers with CAR and β-catenin dual activation, ploidy is abnormally high and the normal senescence response is bypassed, allowing cells to proliferate unchecked. The PI3K/Akt pathway has been implicated in insulin-mediated liver polyploidy and is a commonly disrupted pathway in many types of cancer, including hepatocellular carcinoma. I propose that CAR and β-catenin act together through the PI3K/Akt pathway to overcome the polyploidy block to proliferation. Further studies will focus on elucidating this mechanism. Contributors: Scott, Jessica; Dong, Bingning; Moore, David 279 BAYLOR COLLEGE OF MEDICINE ELECTRICAL PROPERTIES OF THE VERTEBRATE PHOTORECEPTOR NETWORK Robert Lionel Seilheimer Program in Structural and Computational Biology and Molecular Biophysics/M.D.-Ph.D. Program Advisor: Samuel Wu, Ph.D.-Department of Ophthalmology Photoreceptor cells in the retina (rods and cones) convert light into electrical impulses that are ultimately sent to the brain for interpretation and integration, allowing us to see. These cells not only synapse with second-order cells in the retina to transmit visual information downstream to the brain, but also connect laterally with each other. While the physiology of individual photoreceptors has been well studied, the effect of electrical coupling on the action of photoreceptors is poorly understood. To investigate this coupling, we have recorded electrical responses from connected photoreceptors using dual cell patch techniques. These experiments demonstrate symmetric, linear coupling between pairs of photoreceptors. However, these methods are not sufficient to understand how photoreceptors interact on a large scale because of the extensive coupling in the network. Thus, we present a computational model of photoreceptor interaction that incorporates the biophysical properties of individual cells as well as the connections between cells. With this model, we will investigate how individual ion currents and electrical coupling affects the function of photoreceptors in a network. Contributors: Seilheimer, Robert; Gao, Fan; Wu, Samuel 280 2014 GRADUATE STUDENT SYMPOSIUM NOTCH SIGNALING AND STEM CELL-PROGENY INTERPLAY Fatih Semerci Program in Developmental Biology Advisor: Mirjana Maletic-Savatic, M.D./Ph.D.-Department of Pediatrics Formation of new neurons in the adult hippocampus represents an adaptive response of the brain to our environment and/or internal needs. The balance between underproduction and overproduction of newborn neurons is achieved by whole-cell means of altering functional circuitry based on the demand. This whole-cell plasticity suggests that some form of feedback signaling from the progeny to the primary neural stem cell (NSC) may exist. A known mechanism for cell-cell communication involves the Notch pathway. Thus, we hypothesized that amplifying neuroprogenitors (ANPs), which are in direct contact with the primary NSCs, may participate in regulating the NSC quiescence vs. active proliferation. Indeed, the key components of the Notch pathway are expressed in the neurogenic niche in a cell type specific manner. Notch receptor is expressed in the NSCs, while Notch ligands, Jag1and Delta1, are expressed in ANPs and granule cells, respectively. Moreover, utilizing a comprehensive bioinformatics approach, we have discovered that Lunatic Fringe (Lfng), a key modifier of Notch receptor, is selectively expressed in NSC. Further characterization of the Lfng-eGFP expressing cells confirmed that they are NSCs, as they gave rise to neurogenic progeny, decreased in number with increased age, and responded to the electrical stimuli known to increase NSC proliferation. Functionally, removal of Lfng resulted in increased NSC proliferation followed by decrease in NSC number and neuron production, suggesting the necessity of Lfng for proper NSC function. On the other hand, constitutive heterozygote deletion of Jag1 resulted in increased NSC cell cycle duration with no change in their absolute numbers. These results strongly implicate Notch signaling in the control of NSCs and their progeny, and suggest a potential communication and feedback mechanism between the NSCs and their progeny to fine tune adult neurogenesis. Our data are very significant as we present Lfng-eGFP as a new mouse model that permits comprehensive and specific studies of NSC properties and propose a novel mechanism that may operate in the hippocampal neurogenic niche. Contributors: Semerci F,1 Choi WT, 1,2 Thakkar A, 1 Encinas JM, 1,3 Maletic-Savatic M1* 281 BAYLOR COLLEGE OF MEDICINE PROBING INTEGRIN SIGNALING IN NEURONAL MAINTENANCE IN FLIES Mumine Senturk Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics Integrins form strong adhesive junctions between tissue layers, a process that is required in the wing to keep the dorsal and ventral cells attached to each other. A loss of adhesion between these two epithelial layers causes wing blisters. Our lab generated a collection of Drosophila X chromosome mutants to identify essential genes involved in wing development. To identify new players in the integrin signaling pathway, we screened these EMS induced mutations for wing blisters in mutant clones using a UbxFLP. We identified 30 new alleles of known integrin pathway genes and 25 additional mutants that exhibit similar wing blistering phenotypes. We mapped many of these mutants and one corresponds to ubiquilin (ubqn), the Drosophila homologue of UBQLN. Ubqn functions in the ubiquitin-proteasome system-mediated degradation by recruiting ubiquitinated proteins to the proteasome. Mutations in UBQLN2 have been shown to cause dominant X-linked ALS and mutations in UBQLN1 have been linked to Alzheimer’s disease (AD) by genetic linkage analysis and family-based association studies. We found that loss of ubqn in fly eye causes a neurodegeneration, glial death, and an accumulation of small mitochondria upon aging. Moreover, young retina of the ubqn mutants displays an ER expansion which is one of the distinguished features of several mutants that cause ALS. Interestingly, several components of the integrin signaling pathway have recently been associated in GWAS for AD. The link between Ubqn, integrin signaling, and neuronal maintenance is intriguing and I am therefore exploring this link using Drosophila as a model system. Contributors: Mumine Senturk, Shinya Yamamoto, Manish Jaiswal, Lita Duraine, and Hugo Bellen 282 2014 GRADUATE STUDENT SYMPOSIUM CHOLESTEROL HOMEOSTASIS IS REQUIRED TO MAINTAIN COCHLEAR PERIVASCULAR INTEGRITY AND AUDITORY FUNCTION Michelle L. Seymour Department of Molecular & Cellular Biology Advisor: Frederick Pereira, Ph.D.-Department of Molecular & Cellular Biology Animal models and human studies support a link between altered cholesterol homeostasis and sensorineural hearing loss. However, mechanistic understanding of how circulating lipids modulate cochlear cellular activities is lacking. The rising prevalence of obesity and hypercholesterolemia demands a clearer model of how systemic cholesterol homeostasis influences cochlear cholesterol homeostasis and hearing function. We seek to define the relationships between systemic and cochlear cholesterol homeostasis during hearing development and adulthood, as well as how elevations of systemic cholesterol affect cochlear and hearing function. Serum and cochlear cholesterol levels change significantly during normal development of cochlear and auditory function. Interestingly, the sensory epithelium (SE) contains higher levels of cholesterol than the vascular compartment. Low-density lipoprotein receptor knock-out (LDLR KO) mice display elevated hearing thresholds, indicative of hearing loss, concomitant with elevations in serum cholesterol by 1 month of age. However, cochlear cholesterol levels do not increase in LDLR KO mice until 812 months of age. Vascular organization within the stria vascularis (SV), the metabolic engine of the cochlea, is also altered in LDLR KO mice. Perivascular-resident macrophage-like melanocytes (PVM/Ms) contribute to maintenance of the structural integrity of the cochlear blood-labyrinth barrier (cBLB) in the SV vessels and are decreased in LDLR KO mice. Current studies focus on quantifying changes in SV vasculature organization and directly assessing the permeability of the cBLB during acute and chronic hypercholesterolemia. In summary, cochlear cholesterol homeostasis is influenced by serum cholesterol levels during development, but is relatively isolated from serum fluctuations after cBLB maturation. Mechanisms may also exist for concentrating cholesterol in the SE or for endogenous synthesis of cholesterol within the organ of Corti. LDLR KO mice have impaired auditory function that presents simultaneously with hypercholesterolemia. Loss of PVM/Ms and alterations in SV vascular organization occur prior to elevations in cochlear cholesterol, suggesting that cholesterol-induced hearing loss may initially result from strial damage and compromise of cBLB integrity. Contributors: Seymour, Michelle L.; Pereira, Fred A. 283 BAYLOR COLLEGE OF MEDICINE THE ANDROGEN RECEPTOR SPLICE VARIANT, AR-V7, REPROGRAMS METABOLISM TOWARDS A PHENOTYPE CONSISTENT WITH AGGRESSIVE PROSTATE CANCER Ayesha Shafi Department of Molecular & Cellular Biology Advisor: Nancy Weigel, Ph.D.-Department of Molecular & Cellular Biology Prostate cancer (PCa) is the second leading cause of cancer-related death in American men. Androgen ablation therapy is the most common therapy for advanced PCa. Within two years tumors become resistant to therapy and develop castrationresistant prostate cancer (CRPC). There is no effective treatment for CRPC, which is androgen-depletion resistant, but androgen receptor (AR)-dependent. AR is a hormoneactivated transcription factor that mediates androgen action. Recent studies have shown that constitutively active AR splice variants that lack hormone-binding domains are expressed in CRPC. Reports of the actions and contributions of variants to CRPC relative to full-length AR are conflicting. To address this question, we have generated derivatives of androgen-responsive LNCaP and VCaP cell lines with inducible expression of AR-V7, a variant containing exons 1, 2, and 3 of AR and a small amount of unique sequence from exon 3b. AR-V7 stimulates expression of some AR target genes, induces cell growth, and increases cell migration. We performed RNA-Seq to compare the transcriptomes of AR and AR-V7. RNA-Seq analysis reveals a common subset of genes regulated by both receptors and more intriguingly genes uniquely regulated either by AR or by AR-V7 suggesting isoform specific actions. Pathway analysis showed significant regulation of metabolic pathways. Cancer cells frequently become more dependent on the glycolytic pathway than are normal cells. Using liquid chromatography-mass spectrometry (LC-MS), we examined the effect of AR or AR-V7 activation on the levels of several metabolites involved in energy metabolism and proliferation. The changes in metabolite levels suggest that AR-V7 does not induce lipid accumulation, while it does preferentially enhance glycolysis and increase rates of glutamine metabolism (i.e. glutaminolysis). Consistent with this, a Seahorse metabolic flux assay showed that AR-V7 preferentially increases extracellular acidification rates (ECAR), a measure of glycolytic activity. In addition, AR-V7 induces changes in the kreb cycle metabolites including decreased citrate, which is consistent with aggressive PCa. We have confirmed that AR-V7 regulates genes that regulate these metabolites not only in our inducible LNCaP-V7, but in the VCaP-V7 cell model. The previously reported metabolic profile in 22Rv1 cells, which endogenously express full-length AR and AR-V7, is generally consistent with our findings. This study suggests that AR-V7 does not simply substitute for AR, but exhibits gains of function that may include the ability to grow more efficiently in an oxygen poor environment. Contributors: Shafi, Ayesha; Arnold, James; Putluri, Vasanta; Krause, William C; Zheng, Xia; Li, Wei; Putluri, Nagireddy; Sreekumar, Arun; Weigel, Nancy L 284 2014 GRADUATE STUDENT SYMPOSIUM DISSECTION OF FEEDBACK CIRCUIT FROM EXTRASTRIATE AREA LM TO V1 Shan Shen Department of Neuroscience Advisor: Andreas Tolias, Ph.D.-Department of Neuroscience The mammalian visual system is composed of multiple hierarchically organized cortical areas that extract progressively more complex features of the visual scene as information from the retina is fed forward from one cortical area to the next. These multiple visual areas are also extensively connected via feedback pathways, which allows the information extracted by higher areas in the visual pathway to influence more primitive visual responses in earlier areas such as primary visual cortex (V1). Compared to feedforward pathways, we know very little about the functions of feedback projections. Lateral-medial area (LM) project to V1 with abundant axon terminal, but the retinotopical organization and function of these feedback projections are largely unknown. Using optogenetics and eletrophysiological methods, we were able to study the wiring diagram of LM to V1 connections. Anatomically, we found that the LM to V1 feedback projections mainly target the retinotopically related area. Functionally, we found that L2/3 parvalbumin (PV) neurons and somatostatin (SST) neurons receive strongest excitatory input from feedback projections, while pyramidal cells receive both excitatory and inhibitory input. The balance of excitation and inhibition on pyramidal cells creates a sharp temporal excitatory window. Moreover, the feedback projections sharpen the spike timing of pyramidal cells in V1. These results suggest that the modulation of feedback pathway from LM to V1 is to induce temporally precise spiking. Contributors: Shen, Shan; Jiang, Xiaolong; Reimer, Jacob; Tolias, Andreas 285 BAYLOR COLLEGE OF MEDICINE KRÜPPEL-LIKE FACTOR 4 (KLF4) SUPPRESSES T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA BY INHIBITING EXPRESSION OF MAP2K7 AND EXPANSION OF LEUKEMIA INITIATING CELLS. Ye Shen Integrative Program in Molecular and Biomedical Sciences Advisor: H Lacorazza, Ph.D.-Department of Pathology & Immunology Acute lymphoblastic leukemia is the most common hematological malignancy in children. Even though more than 50% of T-cell ALL patients, a subset of ALL, exhibits activating mutations of NOTCH1, the use of gamma-secretase inhibitors has not been successful in patients due to limited response and toxicity. Therefore, identification of genetic factors that cooperate with T-ALL leukemogenesis is needed for the development of alternative therapies. KLF4 is a transcription factor that functions as a tumor suppressor or an oncogene depending on cellular context. Our data showed significant reduction of KLF4 transcripts in lymphoblasts from T-ALL patients compared to blood and bone marrow cells from healthy individuals. From these findings we hypothesized that KLF4 has tumor suppressor function in T-ALL leukemogenesis. To test our hypothesis, we transduced 5-FU treated bone marrow (BM) cells from control (Klf4fl/fl) and Klf4 null (Klf4fl/fl; Vav-iCre) mice with retrovirus carrying a NOTCH1 activating mutant (L1601P∆P) and then transplanted these BM cells into irradiated recipient mice. In contrast to controls, mice transplanted with transduced Klf4-null BM cells developed T-ALL with significantly higher penetrance and shorter latency. On the cellular level, loss of KLF4 led to increased proliferation of leukemia cells as assessed by in vivo BrdU incorporation, which correlated with decreased levels of p21 protein. Limited dilution transplantation of primary leukemia cells into secondary recipients showed a 9-fold increase of leukemia initiating cells (LIC) frequency in Klf4 null leukemia cells compared to controls. To elucidate molecular mechanism we performed microarray and ChIP-Seq in control and Klf4 null CD4+CD8+ leukemia cells. Combined analyses revealed 202 genes as KLF4 direct targets, of which 11 genes are also deregulated in human T-ALL cells by comparing with published microarray datasets. One of the top upregulated genes is Map2k7, which encodes a kinase upstream of the JNK pathway. Immunoblots in leukemia cells confirmed increased expression of MAP2K7 protein and enhanced phosphorylation of its downstream targets JNK and ATF2. JNK inhibitor SP600125 showed dose-dependent cytotoxicity in all human T-ALL cell lines tested regardless of their NOTCH1 status. Overall our results showed that KLF4 functions as a tumor suppressor in T-ALL by regulating proliferation of leukemia cells and frequency of LIC. Additional study elucidated that KLF4 suppresses the JNK pathway via direct transcriptional regulation of MAP2K7. Moreover, the vulnerability of human T-ALL cell lines to JNK inhibition provides a novel target for future therapy in T-ALL patients.Contributors: Ye Shen, Koramit Suppipat, Chun Shik Park, Takeshi Yamada, Toni-Ann Mistretta, and H. Daniel Lacorazza 286 2014 GRADUATE STUDENT SYMPOSIUM INTEGRATED ANALYSIS OF TRANSCRIPTION FACTORS IDENTIFIES SOX11 AS CRITICAL FOR BASAL-LIKE BREAST CANCER GROWTH Jonathan Shepherd Department of Molecular & Cellular Biology Advisor: Powel Brown, M.D./Ph.D.-Department of Molecular & Cellular Biology Intrinsic differences in gene expression between basal-like breast cancer (BLBC) and other breast cancer subtypes imply that transcriptional regulators are differentially activated in breast cancer subtypes and may be promising therapeutic targets. We hypothesized that genomic comparisons between BLBC and non-BLBC will identify transcription factors (TFs) critical for BLBC growth. We identified TFs using an integrative analysis comparing mRNA expression, frequency of TF response elements in differentially expressed genes, and DNAbinding activity of nuclear proteins in BLBC and non-BLBC. We then tested whether inhibition of these specific TFs suppresses the growth of BLBC. Analysis of mRNA expression identified 132 TFs significantly more highly expressed in TNBC tumors compared to non-TNBC samples across 15 breast tumor datasets. Examining promoter sequences of 117 BLBC genes, we identified 94 TF motifs over-represented among BLBC promoters versus ~1500 non-BLBC gene promoters. Binding of DNA motifs by nuclear protein was measured by protein/DNA Arrays (Affymetrix, Santa Clara, CA) using nuclear protein collected from BLBC and non-BLBC cells. This analysis identified 11 TF motifs which were more highly bound by BLBC nuclear lysate. To integrate the results of the three individual screens, we selected the set of TFs identified by at least 2/3 assays. This resulted in a set of 33 candidate BLBC TFs. We then performed a secondary growth screen using 2-dimensional growth of BLBC and non-BLBC cell lines transfected with siRNA to each candidate TF to determine which TFs are critical for BLBC growth. We found that inhibition of SOX11 reduced growth selectively in the BLBC cell lines tested, while growth in the non-BLBC was not affected. Further investigation of SOX11 demonstrated that SOX11 is also critical for other BLBC phenotypes. SOX11 inhibition resulted in reduced migration and invasion, and reduced expression of several genes which are characteristically high in BLBC. Further investigation of the SOX11 will improve our understanding BLBC and may provide a novel target for the treatment and prevention of BLBC. Contributors: Shepherd, Jonathan; Uray, Ivan; Mazumdar, Abhijit; Tsimelzon, Anna; Hilsenbeck, Susan G; Brown, Powel H 287 BAYLOR COLLEGE OF MEDICINE VISUAL WORKING MEMORY OF MULTI-FEATURE ITEMS WITH ORIENTATION AND COLOR Hongsup Shin Department of Neuroscience Advisor: Whee Ma, Ph.D.-Department of Neuroscience Michael Beauchamp, Ph.D.-Department of Neuroscience Visual working memory of items with multiple features has been one of the main interests in cognitive psychology and neuroscience. When more than one feature exists in an item, it brings several issues which do not exist in memorizing a single-feature item. First, memory resource can be shared among features or not. Second, if present, irrelevant feature can be encoded or not. And if irrelevant feature is encoded, it can either affect decision-making or not. These three factors give us six different models. In the current study we used a same experimental paradigm to examine the difference among the models to find the best candidate to explain human working memory behavior. We found that memory resource is not shared by orientation and color, and if present, irrelevant feature is automatically encoded, but can be ruled out during the decision process. Contributors: Shin, Hongsup; Ma, Wei Ji 288 2014 GRADUATE STUDENT SYMPOSIUM ARYL HYDROCARBON RECEPTOR CROSS TALKS WITH REL B TO MODULATE HYPEROXIC INJURY IN NEONATAL HUMAN PULMONARY MICROVASCULAR ENDOTHELIAL CELLS (HPMEC) Binoy Shivanna Clinical Scientist Training Program Advisor: Bhagavatula Moorthy, Ph.D.-Department of Pediatrics Background: Newborn aryl hydrocarbon receptor (AhR) dysfunctional mice are more susceptible to hyperoxia-induced lung inflammation and alveolar simplification. However, the molecular mechanism(s) by which AhR mitigates hyperoxic injury is poorly understood. The NFkB family member, Rel B, is known to exert anti-inflammatory effects in the lungs of adult mice. Interestingly, AhR is known to cross talk with Rel B to modulate inflammation in mouse fibroblasts exposed to cigarette smoke. Whether AhR interacts with Rel B to modulate hyperoxic injury in newborn human lung cells is unknown. Therefore, we tested the hypothesis that AhR attenuates hyperoxia-induced oxidative stress, inflammation, and cell death in HPMEC via Rel B. Objective: To determine the mechanism(s) by which AhR mitigates hyperoxic injury in HPMEC. Methods: AhR gene in HPMEC was knocked down by small interfering RNA (siRNA) transfection and the results were validated by western blotting and real-time RT-PCR analysis. HPMEC transfected with siRNA or treated with an AhR agonist, indole-3-carbinol, were exposed to room air (95% air and 5% CO2 at 370C) or hyperoxia (95% O2 and 5% CO2 at 370C) for upto 48 h. Following exposure, the cells were harvested to determine viability (3-(4, 5-dimethylthiazol-2-yl)-2, 5diphenyltetrazolium bromide (MTT) assay), necrosis and apoptosis (Annexin V staining and flow cytometry), reactive oxygen species (ROS) generation (2’-7’dichorodihyrofluorecein diacetate staining and flow cytometry), inflammation (milliplex cytokine/chemokine assay), cleaved PARP and nuclear Rel B expression (western blotting), and AhR activation (nuclear AhR protein expression by western blotting). Results: Hyperoxia decreased AhR expression and activation in HPMEC. AhR deficient HPMEC had significantly increased hyperoxia-induced ROS generation, cleavage of PARP, and cell death compared to AhR sufficient HPMEC. Additionally, AhR deficient HPMEC culture supernatants had significantly increased macrophage inflammatory protein 1α and 1β, indicating a heightened inflammatory state. Interestingly, loss of AhR was associated with a significantly decreased nuclear Rel B protein expression. AhR agonist, indole-3-carbinol, increased nuclear Rel B expression, decreased hyperoxia-induced ROS generation, and transiently improved the viability of HPMEC. Conclusions: AhR protects HPMEC against hyperoxia-induced oxidative stress, inflammation, and cell death via Rel B. Our data suggest that strategies directed towards increasing AhR activation would be effective in the prevention and treatment of BPD in preterm infants.Contributors: Shivanna Binoy, Shaojie Zhang, Chu Chun, Jiang Weiwu, Wang Lihua, Welty Stephen, and Moorthy Bhagavatula 289 BAYLOR COLLEGE OF MEDICINE RESISTING ADENOSINE IMMUNOSUPPRESION TO IMPROVE ACTIVITY OF TUMOR-SPECIFIC T-CELLS Thomas Shum Program in Translational Biology & Molecular Medicine/M.D.-Ph.D. Program Advisor: Cliona Rooney, Ph.D.-Department of Pediatrics Stephen Gottschalk, M.D.-Department of Pediatrics Adoptively transferred chimeric antigen receptor (CAR) T-cells can specifically home to tumors, proliferate extensively and attack tumor cells with highly effective cytotoxic mechanisms, while producing significantly less adverse effects compared to conventional chemotherapy. However, the lack of sustained T-cell activity after adoptive transfer remains a major challenge for solid tumor immunotherapy. One mechanism of this phenomenon is from T-cell anergy after encountering inhibitory cytokines in tumor microenvironments, with adenosine emerging as a key immunosuppressant. Physiologically, adenosine receptors predominantly activate the cyclic AMP/protein kinase A (PKA) axis to inhibit T-cell activation, serving as a feedback mechanism to prevent T-cells from damaging bystander tissue during injury and inflammation. However, adenosine’s normally protective signaling axis becomes an instrument of tumor growth when adenosine concentrations are elevated in hypoxic tumor microenvironments. While adenosine has been shown to be immunosuppresive for lymphocytes applicable for adoptive immunotherapy strategies such as tumor-infiltrating Tlymphocytes (TILs), natural killer cells, natural killer T-cells, and tumor-specific T-cells, it has yet to be demonstrated that adenosine is immunosuppressive for CAR T-cells. Using the stable adenosine analog, 2-chloro-adenosine (CADO), we have established that CADO inhibits cytokine production and proliferation of OKT3/CD28 activated T-cells (ATCs) and in ATCs expressing a GD2-specific CAR, in-vitro. Based on our preliminary data, we hypothesize that genetic modification of CAR T-cells to resist the adenosine axis of immunosuppression can boost CAR T-cell efficacy against tumor cells in hypoxic co-cultures in-vitro and against established solid tumors in-vivo. Contributors: Shum, Thomas; Omer, Bilal; Tashiro, Haruko; Gottschalk, Stephen; Rooney, Cliona. 290 2014 GRADUATE STUDENT SYMPOSIUM A CRITICAL DEVELOPMENTAL WINDOW FOR 17B-ESTRADIOL ANTIEPILEPTOGENIC EFFECT IN A GENETIC MOUSE MODEL OF X-LINKED INFANTILE SPASMS Meagan Siehr Department of Molecular & Human Genetics Advisor: Jeffrey Noebels, M.D./Ph.D.-Department of Neurology X-linked Infantile Spasms Syndrome (ISSX) is a catastrophic childhood epilepsy disorder characterized by motor spasms that typically arise in the first year of life. Children diagnosed with ISSX often develop seizures later in life that are difficult to clinically manage. Currently, there are few effective drugs available to treat infantile spasms as well as prevent epilepsy later in life and improve developmental outcomes [1]. The most common genetic cause of ISSX is a triplet repeat expansion in the Aristaless-related homeobox (ARX) gene. Our lab generated a mouse model of this mutation (Arx(GCG)10+7) that recapitulates many of the phenotypic features seen in patients with ISSX including epilepsy and motor spasms in early life. In several brain regions, the Arx(GCG)10+7 shows significantly reduced numbers of GABAergic interneurons, a neuronal type that produces the main inhibitory signals of the CNS important for maintaining excitation/inhibition balance in the brain [2]. Recently, our lab found that 17β-Estradiol (E2) (40ng/g/day) administered to neonatal Arx(GCG)10+7 mice from P3-P10 reduced spasms and seizures in this model. Interestingly, E2 treatment in neonatal Arx(GCG)10+7 mice also increased numbers of GABAergic interneurons. However, this effect appeared to be age-dependent, since treatment of adult mice from P33P40 with E2 had no effect on these phenotypes [3]. These results indicate that only early administration of E2 may have an antiepileptogenic effect in the Arx(GCG)10+7 model and there may be a critical, developmental window for effective E2 treatment. In this work, we aimed to define the temporal boundaries of the developmental window for E2 treatment by delaying treatment initiation from P3 to P7, and we have found that delaying E2 treatment initiation until P7, at both 40ng/g/day and 80ng/g/day, renders treatment ineffective and does not reduce seizures and interictal spikes Arx(GCG)10+7 mice. This suggests a critical, developmental window for E2 antiepileptogenic effect in a model of ISSX. Understanding the biological mechanism underlying this developmental window is crucial for understanding how E2 treatment can be utilized in patients with ISSX. Swann, J.W. and S.L. Moshe, On the Basic Mechanisms of Infantile Spasms. Jasper's Basic Mechanisms of the Epilepsies, 2012. Price, M., et al., A triplet repeat expansion genetic mouse model of infantile spasms syndrome, Arx(GCG)10+7, with interneuronopathy, spasms in infancy, persistent seizures, and adult cognitive and behavioral impairment. J. Neurosci, 2009. 29(27): p. 8752-8763. Olivetti, P., A. Maheshwari, and J. Noebels, Neonatal estradiol stimulation prevents epilepsy in Arx model of x-linked infantile spasms syndrome. Science translational medicine, 2014. 6(220). Contributors: Siehr, Meagan; Lucero, Rocco; Noebels, Jeffrey 291 BAYLOR COLLEGE OF MEDICINE A CATALOG OF CIS-REGULATORY VARIANTS IN HUMAN PLATELETS Lukas Simon Program in Structural and Computational Biology and Molecular Biophysics Advisor: Chad Shaw, Ph.D.-Department of Molecular & Human Genetics Blood platelets are essential for normal hemostasis and play an important role in inflammation, tissue repair and regeneration, endothelial cell stability and cancer metastasis. Reduced platelet function is a common cause of pathologic bleeding, whereas increased platelet numbers and reactivity are believed to contribute to pathologic thrombosis, such as myocardial infarction and stroke. Human platelets express and translate mRNAs, and contain a diverse repertoire of long and short noncoding RNAs. Thus, platelets present a unique opportunity to correlate transcriptomic information with cell function and other biologic parameters. The Platelet RNA and eXpression-1 study was designed to investigate significant heritable inter-individual variation in platelet functional properties. This study profiled platelet mRNA expression and genotype in a multi-ethnic cohort of 154 healthy humans. To broaden our understanding of the genetic basis underlying human platelet gene expression, we conducted expression quantitative trait locus (eQTL) analysis. Among 5911 genes expressed in platelets, we identified 439 genes with at least one significant cis-eQTLs (P<10e-6). Using data from the Genotype-Tissue Expression project we discovered that 108 of these genes also contained an eQTL in at least one different tissue and that the majority cis-eQTL genes are platelet-specific, including platelet genes PCTP, PDK1 and VAMP8. When analyzing data from a collection of genome wide association studies (GWAS), we observed that platelet genes containing cis-eQTLs are more likely to be disease associated compared to platelet genes without cis-eQTLs (P=4.7e-6). Plateletspecific cis-eQTL genes were significantly enriched among genes annotated to the MESH term “Cardiovascular Diseases” (P<0.05). Among GWAS traits significantly enriched for platelet-specific eQTL genes are platelet relevant terms such as “Apolipoproteins B”, “Carotid Arteries”, “Intracranial Aneurysm” and “Platelet Count” (P<0.05). The comprehensive catalog of all platelet cis-eQTLs can be a useful resource for future studies, such as in the prioritization and interpretation of GWAS implicated genes and polymorphisms. Contributors: Simon, Lukas; Chen, Edward; Edelstein, Leonard; Bray, Paul; Shaw, Chad 292 2014 GRADUATE STUDENT SYMPOSIUM DETERMINATION OF NK CELL LYTIC GRANULE MOTILITY AT THE IMMUNOLOGICAL SYNAPSE Papiya Sinha Department of Pathology & Immunology Advisor: Jordan Orange, M.D.-Department of Pediatrics Natural killer (NK) cells are cytotoxic lymphocytes of the innate immune system that kill virally infected and malignant cells. They contain specialized secretory lysosomes called lytic granules, which mediate target cell death by the secretion of perforin and granzymes onto a diseased cell for host defense. This process of cytotoxicity depends upon the ability of NK cells to transport their granules to the immunologic synapse (IS) with the help of microtubules. This is followed by granules approaching the NK cell plasma membrane by the attachment of actin-based motor protein myosin IIA present on granules to the F-actin network at the IS. Then, granule docking and fusion with the cell membrane and subsequent release of granule contents takes place. Until recently, it was suggested that immediately after granule-MTOC polarization to the IS, granules docked/ fused with the plasma membrane. However, using advanced high-resolution 2D-Total Internal Reflection Fluorescence microscopy (TIRFm), we recently defined an unexpected multidirectional movement of granules at the IS prior to docking and fusion. We first characterized the highly dynamic lytic granule motility at the IS through previously unapplied complex quantitative and statistical analyses. Initial results showed that granules are sub-diffusive (constrained motility due to the crowded environment) at the IS. Furthermore, depolymerization of microtubules using nocodazole inhibitor revealed lesser granule displacement, while inhibition of F-actin with latrunculin A treatment displayed more granule displacement at the IS. Inhibition of both microtubules and F-actin together led to decreased granule motility. Surprisingly, inhibition of myosin IIA with ML-9 and blebbistatin inhibitors led to lower granule displacement within the first few minutes post-treatment, followed by increased motility thereafter. In order to compare the sub-diffusive granule motility at the IS with an object that is freely diffusing, we calculated the free diffusion coefficient of granules. Since free diffusion motility is dependent on the viscosity of the local environment, it was important to determine the viscosity of NK cells which is currently unknown We microinjected NK cells with fluorescent beads of similar size to granules, tracked their motility at the IS using TIRFm, and determined the synaptic viscosity on NK cells for the first time. This study is important because it underscores a new and unforeseen step that facilitates NK cell killing. In addition to filling the gap in knowledge of granule motility at the IS, these results identified previously unknown requirements for effective cytotoxicity. Our results broaden the understanding of NK cell biology with the use of advanced microscopy and multifaceted quantitative analysis, which will be beneficial for therapeutic intervention of NK cell function to treat immunological disorders. Contributors: Sinha, Papiya; Tsao, David; Diehl Michael; Kolomeisky Anatoly; Orange, Jordan. 293 BAYLOR COLLEGE OF MEDICINE MAINTAINING TRANSCRIPTION FIDELITY IMPEDES DNA DOUBLE-STRAND BREAK REPAIR IN ESCHERICHIA COLI Priya Sivaramakrishnan Department of Molecular & Human Genetics Advisor: Christophe Herman, Ph.D.-Department of Molecular & Human Genetics For accurate function and viability, all organisms possess pathways that protect their nucleic acids. One of the most devastating forms of DNA damage is a doublestrand DNA break (DSB) caused by both endogenous and exogenous carcinogens. Improper processing of these breaks can lead to chromosomal rearrangements that trigger genomic instability. Transcription elongation can affect DSB repair as both these phenomena occur on the same DNA template. When an elongating RNA polymerase (RNAP) encounters a DSB, the cell has to choose between aborting transcription to fix the break, and attempting to continue transcription in order to preserve RNA fidelity. Although it would seem most likely that the cell would prefer to repair the break and safeguard the genome while compromising transcription, the fact that transcription errors occur more frequently than DNA replication errors and that mRNA mutations can have heritable consequences suggest that there may be situations when maintaining RNA fidelity outweighs protection of the genome. In Escherichia Coli, transcription factors that directly bind RNAP modulate its processivity. These include the transcription elongation factor DksA and the transcript cleavage factor GreA (Eukaryotic TFIIS). GreA rescues stalled RNAP complexes, ensuring the production of full-length transcripts and hence preserves RNA fidelity. We have found that loss of GreA results in greatly increased survival when cells are exposed to DSB inducing agents. Further, these mutants also show enhanced viability in an assay that is dependent on the ability to prevent or fix DSBs created by the I-SceI endonuclease. This result in conjunction with analysis of breaks through pulsed-field gel electrophoresis suggests that loss of GreA causes improved repair of DSBs. This ameliorated repair is through the canonical homologous recombination pathway. The absence of DksA on the other hand, displays an opposite phenotype, rendering cells highly sensitive to induced DSBs. Overexpression of DksA can rescue wild-type survival on exposure to DSBs, suggesting that a competition between GreA and DksA for RNAP binding ultimately affects repair outcome. We hypothesize that a transcribing RNAP can run into repair proteins that are responsible for processing DSBs to promote homologous recombination. In wild-type cells, RNAP acts as a physical blockade to DSB processing. In the absence of GreA, RNAP is a less formidable obstacle for these proteins, allowing for enhanced DSB repair. This idea is supported by our in vitro experiments showing that RNAP dissociates at a faster rate without GreA, when proteins are bound to the end of a DSB. Thus, these transcription factors modulate DSB repair and may play a role in the cellular decision between maintaining genome integrity and preserving transcription fidelity. Contributors: Sivaramakrishnan, Priya; Liu, Zhaokun; Halliday, Jennifer A.; Shee, Chandan; Artsimovitch, Irena; Rosenberg, Susan M.; and Herman, Christophe 294 2014 GRADUATE STUDENT SYMPOSIUM SYSTEMATIC ANALYSIS OF NON-PROTEIN CODING SEQUENCE VARIATION REVEALS PUTATIVE PATHOGENIC MUTATIONS CAUSING INHERITED HUMAN RETINAL DISEASE Zachry Tore Soens Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D.-Department of Molecular & Human Genetics Purpose Currently the identification of human disease-causing mutations has been largely limited to protein coding regions due to our limited knowledge of the rest of the genome. The purpose of this study is to assess the contribution to disease of identified novel sequence variations in non-protein coding regions in a cohort of more than 1000 human retinal disease patients. Methods Our approach combines computational and experimental methods. Based on the exon sequence of all known retinal disease genes in over 1000 patients, a statistical method was developed to estimate the contribution of noncoding mutations in each gene to each disease. Noncoding variants of the top ranked disease genes were identified using a combination of next generation sequencing and array comparative genomic hybridization. Identified variants were analyzed using an integrative approach for their potential to disrupt the function of genes required for proper retinal function. Candidate pathogenic variants were Sanger verified and functionally validated to alter gene function or expression. Results 16 genes that were likely to contain a significant number of noncoding pathogenic mutations, such as USH2A and ABCA4, were identified. Characterization of the genomic region of these genes in our patient cohort identified several novel candidate pathogenic noncoding variants in multiple disease cohorts including Leber congenital amaurosis, Usher syndrome, and Stargardt disease. Identified variants are predicted to disrupt gene splicing causing a proportion of transcripts to lose a typical exon or gain a cryptic exon. Conclusions We have developed a new statistical method to estimate the mutation load in the noncoding region for each known retinal disease gene. Follow up studies of top ranked genes identified multiple types of non-protein coding mutations including SNVs and indels. As a significant proportion of patients’ disease can be attributed to mutation in noncoding regions, further improving our ability to identify and interpret these types of mutations is crucial. Contributors: Soens, Zachry; Zaneveld, Jacques; Gelowani, Violet; Jiang, Lichun; Sui, Ruifang; Koenekoop, Robert; Chen, Rui 295 BAYLOR COLLEGE OF MEDICINE OLFACTORY REGULATION OF C. ELEGANS REPRODUCTIVE AGING Jessica Nichole Sowa Department of Molecular & Human Genetics Advisor: Meng Wang, Ph.D.-Department of Molecular & Human Genetics As women age, they experience both a decline in fertility and an increased risk of miscarriage and birth defects. In addition to being a significant public health concern, this ageassociated reproductive decline is one of the first aging phenotypes to manifest in humans. Although predicted to be a regulated process, little is known about the molecular mechanisms regulating the onset and progression of reproductive aging. In addition to purely genetic contributions, reproductive aging is thought to be regulated by complex interactions of environment-sensitive genetic signaling pathways and environmental conditions. However, the molecular mechanisms integrating environmental and genetic signals to regulate reproductive aging remain unknown. As soil-dwelling nematodes, Caenorhabditis elegans experience their diet of bacteria as a source of both metabolic and sensory input. We took advantage of this relationship as a convenient method of introducing environmental variation by raising C. elegans on different bacterial diets. We found that C. elegans exposed to different bacterial environments show significant differences in the duration of their reproductive span. To further dissect the molecular mechanisms underlying these reproductive span differences we chose to focus on two strains of Escherichia coli that gave drastically different effects on C. elegans reproduction. OP50 E. coli is the standard lab diet for C. elegans, and we found that worms raised on this strain reproduce longer and maintain their fertility later than worms raised on the alternate E. coli strain HB101. This effect is mediated by a pair of olfactory neurons which perceive a volatile odorant signal from the HB101 E.coli. The presence or absence of this environmental cue affects germline proliferation and maintenance in C. elegans, ultimately contributing to the timing of reproductive senescence. We have identified a pair of olfactory neurons, the AWB neurons, that are specifically required for this olfaction-mediated reproductive adaptation as well as for chemotaxis of C. elegans to the smell of the HB101 bacterial diet. Furthermore, we found that AWB neuronal photoactivation is sufficient to promote this reproductive adaptation. Finally, we identified neuropeptide release as the mechanism of AWB neuronal signaling. Together, our results reveal a novel pathway for the regulation of reproductive aging in C. elegans, and suggest the relevance of environment-sensitive signaling mechanisms in regulating the onset and progression of reproductive aging. Contributors: Sowa, Jessica; Ozseker, Ayse Sena; Wang, Meng 296 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF C/EBPß IN MAMMARY STEM/PROGENITOR CELLS AND TUMOR INITIATING CELLS Aaron Jonathon Spike Department of Molecular & Cellular Biology Advisor: Jeffrey Rosen, Ph.D.-Department of Molecular & Cellular Biology The goal of these studies is to elucidate the mechanisms by which the transcription factor C/EBPβ regulates mammary stem cells (MaSC) and tumor initiating cells (TICs). There are three C/EBPß isoforms, LAP1, LAP2, and LIP. C/EBPβ is known as a master regulator of mammary gland (MG) development however, its precise function in MaSCs is unclear. C/EBPß-/- mammary epithelial cell (MEC) transplantation results in severe defects and a significant reduction in MaSCs. Expression of LIP in human patients correlates with poor prognosis in ER/PR-/- breast cancer. Gene expression analysis of C/EBPß-/- MECs revealed a set of genes whose expression was modulated by C/EBPß which includes: Eya1, Notch3, ∆Np63, and Stat3. Eya1 is a negative regulator of Notch, and is also necessary for MaSCs asymmetric cell division. Expression of Notch3 in the MG is a marker for luminal progenitor cells. ∆Np63 is known to be a master regulator of stemness and contains C/EBPß binding sites in its promoter. Deletion of Stat3 results in a reduction of MaSCs and leads to developmental defects in regenerated MGs. C/EBPβ binds the Stat3 promoter to drive its expression. Preliminary data shows that Stat3 activation is evident in a small pool of MECs suggesting that Stat3 may be activated in MaSCs. These observations lead to my hypothesis that C/EBPβ regulates the expression of a set of genes necessary for stemness in an isoform specific manner in MaSCs. I will test this with the following aims: Aim 1: I will determine that C/EBPβ regulates MG development in an isoform specific manner. Aim 2: I will dissect the relationship between C/EBPβ isoforms and the expression of the above mentioned gene set in MaSCs. Aim 3: I will determine if these interactions are also evident in TICs, and if so, what the consequence is. These studies will broaden our knowledge of MG development, MaSC function, and breast cancer and may also provide new insights into discovering novel interactions important for stem cell biology. Contributors: Spike, Aaron 297 BAYLOR COLLEGE OF MEDICINE DETERMINING THE ROLE OF WNT AND P53 PATHWAYS IN MAMMARY STEM CELL FATE DETERMINATION Amulya Sreekumar Department of Molecular & Cellular Biology Advisor: Jeffrey Rosen, Ph.D.-Department of Molecular & Cellular Biology Asymmetric cell division (ASCD) typifies stem cells and results in a stem cell and a differentiated cell via mitosis, thus maintaining a steady stem cell pool. ASCD is critical for organ growth and homeostasis. Tumours often display increased symmetric cell divisions (SCD), that exponentially expands the stem cell pool. Studies in embryos and in vitro have revealed the p53 and Wnt pathways as possible mediators of cell fate decision. We asked if this holds in adult organisms in vivo. We propose to study stem cells in the mammary gland and the aggressive, cancer stem cell (CSC) driven basallike breast cancer (BLBC) subtype. We hypothesize that p53 and Wnt are critical modulators of ASCD, perturbation of which skews towards SCD, and in tumours results in an expanded CSC pool. To test this, we propose the following aims. (1) To elucidate the role of Wnt and p53 in ASCD of mammary stem cells Pubertal mammary glands show a restricted Wnt signalling niche, which is the potential site of ASCD. We will modulate levels of Wnt +/- p53 to study the role of these pathways in ASCD. (2) To demonstrate that perturbation of Wnt and p53 are both required to increase SCD The MMTV-Wnt1 overexpression model, p53-/- transplanted mammary gland model and the MMTV-Wnt1;p53-/- model will be used to delineate the role of Wnt and p53 in precancerous lesions (3) To establish the dependence of tumour growth on the pool of symmetrically dividing CSCs A panel of p53-/- BLBC mouse tumours with varying proportions of CSCs will be used to study the effect of novel pharmacological inhibition of the Wnt pathway. This will yield a titrable system to study the effects of targetting SCD in the CSC population and will be tested with chemotherapy to effect complete BLBC regression. This study is significant as it could shed light on the mechanism underlying a core developmental process and generates a rationale for pharmacological targeting of modulatory components of stem cell biology in BLBC patients. Contributors: Sreekumar, Amulya; Rosen, Jeffrey 298 2014 GRADUATE STUDENT SYMPOSIUM SELECTIVE ENCODING OF INERTIAL AND GRAVITATIONAL SELF-MOTION IN THE CEREBELLUM OF ALERT MICE Trace L Stay Department of Neuroscience Advisor: Dora Angelaki, Ph.D.-Department of Neuroscience The vestibular system provides information about one’s movement through the world. This is a critical component in sensorimotor integration, which underlies accurate movements. One complication in sensory processing is the inherent ambiguity in the signals sent by the otolith organs, which encode inertial and gravitational accelerations identically. Theory has predicted that the brain can differentiate inertial and gravitational components of self-motion centrally by combining otolith and semicircular canal information. Cells in the macaque monkey cerebellum have responses to vestibular stimulation which match these predictions. However, the exact functions of individual synapses in the circuit underlying this central computation are not known. We have pursued this question directly with single-unit electrophysiological recordings of cerebellar lobules 9c and 10 in alert mice. We chose mice because of the opportunity to use powerful genetic methods to precisely manipulate the vestibular circuit. Using multiple stimulation methods, we find that mice do have significant populations of cells which selectively encode inertial or gravitational accelerations. This suggests that the fundamental mechanisms of vestibular processing are common between mice and macaques, and establishes mice as a relevant model for investigating the mammalian vestibular circuit. We can now use a novel genetic strategy to silence individual cell types in the cerebellum and determine their specific computational roles. Our findings thus establish an important framework for future studies on the role of the cerebellum in sensory processing. Contributors: Stay, Trace; Shinder, Michael; Sillitoe, Roy*; Angelaki, Dora* 299 BAYLOR COLLEGE OF MEDICINE LABELING OF MACROPHAGES WITH NOVEL GADOLINIUM OXIDE NANOPARTICLES FOR IN VIVO IMAGING OF NEURAL DEVELOPMENT IN CHICK EMBRYO Gary R. Stinnett Department of Molecular Physiology & Biophysics Advisor: Robia Pautler, Ph.D.-Department of Molecular Physiology & Biophysics Cell labeling with MR contrast agents has been a major focus of in vivo imaging. Labeling of cells with T2 agents has allowed the visualization of cell populations in vivo, but precise concentration and location measurements remains challenging. T1 labeling of cells is preferable to T2 labeling because T1 contrast exudes positive contrast. Quality labeling of cells with T1 agents has been difficult due to difficult synthesis of agents, toxicity, poor stability at physiological pH, and no assay to easily determine novel targeted T1 agent efficacy in vivo. The Colvin group has synthesized gadolinium oxide nanoparticles that are coated with PAMPS-LA. These nanoparticles are stable down to a pH of 3 and are non-toxic to cells at a labeling concentration of up to 250 uM. These nanoparticles also provide about 9000 times more contrast per molecule than Gd-DTPA in water. We have electroporated PAMPS nanoparticles into developing chick neural tube cells to track neural development in vivo. Imaging cells electroporated with PAMPS nanoparticles highlights the growth and development of the neural structure in chick embryos. Contributors: Stinnett, G.R. ; Cho, M. ; Zhu H. ; Pedersen, S.E. ; Deneen, B. ; Colvin, V.L. ; Pautler, R.G. 300 2014 GRADUATE STUDENT SYMPOSIUM DYSFUNCTIONAL NONSENSE-MEDIATED DECAY CAUSES SYNAPTIC AND BEHAVIORAL ALTERATIONS ASSOCIATED WITH AUTISM AND MENTAL RETARDATION Loredana Georgiana Stoica Department of Molecular & Cellular Biology Advisor: Mauro Costa-Mattioli, Ph.D.-Department of Neuroscience Eukaryotic cells have complex post-transcriptional quality control mechanisms that ensure correct gene expression in normal, physiological settings as well as under stress conditions. One of these mechanisms is nonsense-mediated decay (NMD), a surveillance pathway that targets for degradation mRNA transcripts carrying premature translation termination codons (PTC). Thus, NMD blocks translation of potentially harmful truncated proteins that might have dominant-negative functions. Given that 1) a growing list of neuronal genes are reported to be regulated by nonsense mediated decay during brain development and synaptic plasticity and 2) mutations in several core components of the NMD pathway have been described in patients suffering from mental retardation and autism, we decided to explore the consequences of dysfunctional NMD in the adult brain. Therefore, we generated a new mouse model in which UPF2, one of the key components of NMD machinery, is deleted in the forebrain in a αCaMKII Cre-dependent manner. Similar to patients carrying UPF2 mutations, UPF2-deficient mice display behavioral alterations including impaired social interaction, behavioral inflexibility, and learning & memory deficits. Currently, we are investigating the molecular mechanisms underlying these neurocognitive defects. Interestingly, we discovered that, in these mutant mice, the behavioral abnormalities are accompanied by reactive astrogliosis, recruitment of a variety of immune cells to the brain, and increased production of chemokines and proinflammatory cytokines. In summary, we found that dysregulation of UPF2-mediated NMD could contribute to cognitive dysfunction and propose that UPF2-deficient mice can serve as a useful tool to study the function of nonsense mediated decay in brain processes. Ultimately, understanding how impaired NMD mediates cognitive dysfunction and autism-like behaviors will lead to drug discovery and therapeutic intervention for patients with NMD-related intellectual disability. Contributors: Zhu, Ping Jun; Johnson, Jennifer; Buffington, Shelly; Bhattacharya, Abhisek ;Stinett, Gary; Broussard, John; Eissa, Tony; Pautler, Robia; Pedersen, Steen; Nakada, Daisuke; Porse, Bo; Costa-Mattioli, Mauro; 301 BAYLOR COLLEGE OF MEDICINE SUBSTITUTIONS IN THE OMEGA-LOOP OF TEM BETA-LACTAMASE CHANGE THE SUBSTRATE PROFILE OF THE ENZYME Vlatko Stojanoski Department of Biochemistry & Molecular Biology Advisor: Timothy Palzkill, Ph.D.-Department of Pharmacology Serine β-lactamases are bacterial enzymes that hydrolyze β-lactam antibiotics. Mechanistically, serine β-lactamases are very similar to serine proteases such as chymotrypsin. They both utilize acylation and deacylation of an active site serine in their mechanism of catalysis. TEM, a common plasmid-encoded serine β lactamase, catalyzes the hydrolysis of early penicillins and cephalosporins. Here we examine a previously identified triple mutant of TEM 165-TyrTyrGly-167 (wild type165-TrpGluPro167) with switched substrate specificity from ampicillin to ceftazidime. When compared to chymotrypsin, the Glu166Tyr substitution in the TEM triple mutant is analogous to a substitution of the general base His57 in chymotrypsin that results in an enzyme that maintains function. Our findings agree with previous observations of altered substrate specificity of the triple mutant, which displays increased hydrolysis of ceftazidime. Additionally, enzyme kinetic analysis shows that the hydrolysis of ceftazidime follows a branched pathway characteristic of substrate-induced reversible inactivation. Structural analysis of the triple mutant at 1.39 Å resolution reveals enlargement of the active site making it more accessible for larger substrates. Furthermore, in the crystal structure of the triple mutant Tyr166 is within hydrogen bond distance to Ser70 suggesting it acts as a general base to activate the serine in the catalytic mechanism of the enzyme. These findings change the current view that Glu166 is indispensible in the mechanism of serine β-lactamases. More generally, this provides insights into alternate mechanisms for hydrolysis reactions catalyzed by enzymes. Contributors: Vlatko Stojanoski, Liya Hu, BV Prasad, Dar-Chone Chow, Banumathi Sankaran, and Timothy G. Palzkill 302 2014 GRADUATE STUDENT SYMPOSIUM ASSESSMENT OF COMBINATORIAL GENE THERAPY FOR THE TREATMENT OF OSTEOARTHRITIS Adrianne Elayne Stone Program in Translational Biology & Molecular Medicine Advisor: Brendan Lee, M.D./Ph.D.-Department of Molecular & Human Genetics Carlos Bacino, M.D.-Department of Molecular & Human Genetics Osteoarthritis (OA) is a progressively debilitating condition involving joint degeneration that causes substantial joint stiffness and pain, decreased mobility, and increased healthcare costs. Despite approximately 27 million Americans being afflicted with this disease, there are no medical treatments that significantly alter its progression. Currently, an individual suffering from OA will only be proscribed pain management medication, physical therapy, and lifestyle modification, and in end-stage disease a joint replacement surgery will be advised. Injections of cytokines and proteoglycans have shown some promise in clinical and pre-clinical trials, yet due to the rapid degeneration and turnover of these compounds, repeated injections are necessary to obtain any longlasting results, potentially leading to an increase in adverse events. Major obstacles to the development of new treatments for OA include both the difficulty of identifying quantifiable endpoints for progression in animal models, and the complex cell autonomous and non-cell autonomous components of the pathophysiology of the joint. To overcome these obstacles, our lab developed a novel imaging technique combining staining, phase-contrast optics, and microCT analysis of the joint, which allows for the robust quantification of murine cartilage volume, surface area, and osteophyte formation. To address the issue of the complex etiology of OA development, our lab has identified secreted molecules as potential therapeutic targets – Proteoglycan 4 (PRG4), Interleukin 1 Receptor antagonist (IL1Ra), and the Insulin-like Growth Factor 1 (IGF1). PRG4 is a major component of the cartilage extracellular matrix and mediates mechanical stress by providing lubrication. Inflammation has also been attributed to disease progression, and IL1Ra has been proven to function as an anti-inflammatory through the inhibition of Interleukin 1 (IL1). IGF1 is an anabolic agent that can help maintain cartilage by promoting chondrocyte proliferation and inhibition of apoptosis. Both published work and preliminary data from our lab and others shows that individually each provides some protection from OA progression. We hypothesize that combinatorial therapy with PRG4, IL1Ra, and IGF1 will provide increased protection from OA development and progression through addressing cartilage matrix loss and inflammation. This will be tested by comparing the effects of combinatorial therapy to duo- and mono-therapy in a post-traumatic model which involves generation of joint injury through the Cruciate Ligament Transection surgery – a procedure which mimics the human condition of knee injury by ACL tear. We will also assess the effects of the combinatorial therapy in an age-related model of OA development, as the contribution of factors leading to disease progression is different in each model. Contributors: Stone, Adriane; Ruan, Merry Z; Dawson, Brian; Lee, Brendan 303 BAYLOR COLLEGE OF MEDICINE INTRINSIC DISORDER CONTRIBUTIONS TO BINDING AND ALLOSTERY Danielle Dee Stuhlsatz Program in Structural and Computational Biology and Molecular Biophysics Advisor: B. Pettitt, Ph.D.-Biochemistry Misfolded and unstructured domains of proteins represent important examples of disease states where the understanding of the recognition, or folding process has important potential therapeutic implications. The mechanisms governing molecular recognition and the transition of proteins from their unfolded state to their native state remain related but unanswered fundamental biophysical questions. Refolding of protein domains often occurs during DNA binding. Specifically, LacI contains a hinge region that is disordered without DNA or when nonspecifically bound, but becomes ordered when bound to the correct DNA sequence. Experimentally, the hinge region has a measurable disorder to order transition when binding its specific sequence. This system has a nontrivial sequence and allows us to test the importance of the protein folding transition versus that of the contributions of DNA bending and DNA protein interactions separately. The hinge-helix sequence of the LacI system will be computationally tested in various multicomponent aqueous solutions. Our recent theoretical work suggests the solvent effect on the initial collapse toward folding and the early processes of molecular recognition are related. Contributors: Pettitt, B.M. 304 2014 GRADUATE STUDENT SYMPOSIUM JAG1 PROMOTES METASTATIC POTENTIAL OF PROSTATE CANCER CELLS Qingtai Su Integrative Program in Molecular and Biomedical Sciences Advisor: Li Xin, Ph.D.-Department of Molecular & Cellular Biology Prostate cancer is the second leading cause of cancer-related deaths in western countries’ males. Within that metastasis contributes to most of mortality and morbidity. Tissue microarray assay had revealed that Jag1 protein was elevated in tumor samples derived from patients who developed metastatic recurrent diseases; however, the roles of Jag1 in prostate cancer development are inadequately defined. We sought to use transgenic mouse models and human prostate cancer cell lines to investigate the role of Jag1 in prostate cancer progression. Firstly, we evaluated the effects of Jag1 deregulation on the proliferation and migration of the prostate cancer cell lines. Up-regulation of Jag1 in Du145 did not affect their proliferation. However, Jag1 positively regulates the migratory capacities of Du145 in an in vitro wound-healing assay. To validate the role of Jag1 in metastasis of prostate cancer in vivo, we injected the control Du145 cells and the Du145 cells over-expressing Jag1 into NOD/SCID mice via tail veins. Our result showed that Jag1 up-regulation promoted distal colonization of tumor cells in lung, reduced latency of metastasis and shortened life span. We also generated a transgenic mouse model that allows conditional over-expression of Jag1 in prostatic epithelial cells. We found that over-expression of Jag1 alone did not transformed epithelial cells by the age of 26week. Growth rate and resistance to death of prostate epithelial cells were not altered by the presence of higher Jag1 expression. Nevertheless, we discovered that Cyclins levels were up-regulated in the epithelium, which might cause some effects in older mice. On the other hand, we are currently inducing Jag1 expression in the well-defined Pten-null prostate cancer model which otherwise is not aggressively metastatic in quest of functions of Jag1 in the prostate cancer metastasis. Contributors: Su, Qingtai; Kwon, Oh-Joon; Zhang, Li; Valdez, Joseph; Zhang, Boyu; Wei, Xing; Xin, Li 305 BAYLOR COLLEGE OF MEDICINE KU’S ROLE AT HUMAN TELOMERES Ann Tabitha Sukumar Department of Molecular & Human Genetics Advisor: Alison Bertuch, M.D./Ph.D.-Department of Pediatrics Telomeres, the nucleoprotein structures found at chromosome ends, consist of tandem repeats of TTAGGG sequence bound by proteins collectively known as the shelterin complex. The higher order DNA-protein structure of telomeres shields the chromosome ends from appearing as DNA double strand breaks. Without such protection, end-to-end fusion of chromosomes takes place via non-homologous end joining (NHEJ), which leads to cell growth arrest. Ku, a crucial upstream factor in the NHEJ pathway, binds to DNA ends at double strand breaks to initiate NHEJ. However, Ku also associates with telomeres and contributes to telomere function. How Ku is regulated to inhibit its NHEJ function at the telomeres and to perform its telomere protection function is not well understood. Ku interacts with shelterin proteins TRF1 and TRF2 at the telomeres. Previous data from our lab suggests that Ku-TRF2 interaction might prevent Ku from participating in NHEJ at telomeres. To investigate this idea, we aim to understand how Ku associates with telomeres. We hypothesize that Ku associates with telomeres via protein-protein interaction as opposed to end binding, which could result in deleterious fusions. We will perform in vitro studies using purified Ku and TRF2 proteins and DNA substrates with and without telomeric repeats or access to the ends to assess if Ku associates with telomeres via TRF2. In addition, we will use Ku mutants that are impaired for TRF2 binding or end binding to explore Ku’s mode of association with telomeres via chromatin immunoprecipitation assay. By studying how Ku associates with telomeres, we hope to gain a better understanding of Ku’s conflicting roles at telomeres. Contributors: Sukumar, Ann; Bertuch, Alison 306 2014 GRADUATE STUDENT SYMPOSIUM PROTECTING CAR T CELLS FROM IMMUNOSUPPRESSIVE IL4 CYTOKINE PRESENT IN THE TUMOR MICROENVIRONMENT Sujita Sukumaran Program in Translational Biology & Molecular Medicine Advisor: Ann Leen, Ph.D.-Department of Pediatrics William Fisher, M.D.-Department of Surgery The promising clinical results achieved using adoptively transferred chimeric antigen receptors (CAR)-modified T cells in patients with B cell malignancies has prompted investigators to explore this strategy for the treatment of solid tumors. However, while the infusion of CAR T cells may provide therapeutic benefit, solid tumors use a variety of immuneevasion mechanisms, including the production of immunosuppressive cytokines (IL4, IL10, TGF ) that can inhibit effector T cells. To protect our CAR T cells from the suppressive effects of IL4, we generated a chimeric cytokine receptor (4/7R) that comprises the ectodomain of the IL4 receptor fused to the endodomain of the immunostimulatory IL7 receptor, with mOrange coexpressed to allow transgene detection. Thus, transgenic expression of the 4/7R should enable T cells to utilize tumor-produced IL4 to promote CAR T cell expansion and cytotoxic activity. To implement this approach for pancreatic cancer, a highly aggressive disease with a five-year survival of <1%, we generated a CAR targeting the tumor-expressed prostate stem cell antigen (PSCA). When expressed on T cells (78±20%, n=5) transgenic cells were able to specifically kill PSCA-expressing cell lines (CAPAN1 and K562-PSCA) but not 293T (PSCAtarget) (74±4%, 73±6% and 9±3% specific lysis, respectively, 10:1 E:T, n=3). To next evaluate the protective effects of the 4/7R we double transduced T cells to express both CAR-PSCA and the 4/7R (30-72% double positive cells; n=4) and assessed the expansion of both control (CARPSCA) and 4/7R/CAR-PSCA T cells in the presence of antigen and IL4. Although CAR-PSCA T cells were unable to expand in the presence of IL4 (from 2x106 cells on day 0 to 6.1±3.8 x 107 on day 28), 4/7R/CAR-PSCA T cells expanded exponentially (from 2x106 cells to 5.1±3.6 x 109) (n=3). Moreover, culture in IL4 for 4 weeks enriched the 4/7R (mOrange+) transgenic cells (mean 45.9±15% - day 0 to 86±11% - day 28, n=3). We next assessed the cytokine profile of 4/7R/CAR-PSCA T cells exposed to IL4- a prototypic Th2 cytokine, while IL7 induces a Th1polarized cytokine profile. Exposure of CAR-PSCA T cells to IL2 induced prototypic Th1 cytokines including IFN- and TNF23±14 ng/mL and 2.7±2 ng/mL respectively), which was diminished in the presence of IL4 (1.8±1.7 ng/mL and 0.4±0.1 ng/mL). However, the 4/7R restored the Th1 cytokine profile of transgenic cells exposed to IL4 (IFN- - 30.2±20 ng/mL and TNF3.2±2 ng/mL). Importantly, the selected 4/7R CAR-PSCA T cells continue expanding only in presence of both antigen and cytokine confirming their safety for in vivo translation. Thus, we have demonstrated the feasibility of the 4/7R in protecting CAR T cells from inhibitory IL4 by inverting a tumor-derived suppressive signal to enhance anti-tumor activity. Contributors: Sukumaran, S; Mohammed, S; Watanabe, N; Bajgain, P; Anurathapan, U; Leen, A.M; Vera, J.F. 307 BAYLOR COLLEGE OF MEDICINE GENETIC CHARACTERIZATION OF NORADRENERGIC FUNCTION IN RESPIRATORY PHYSIOLOGY Jenny J Sun Department of Neuroscience Advisor: Russell Ray, Ph.D.-Department of Neuroscience The perpetual rhythm of breathing is essential to survival and emerges through the interactions of a complex array of neuronal networks. Noradrenergic (NA) neurons have been implicated as a critical participant in the circuitry of this homeostatic behavior, and past studies point to a high molecular and functional complexity within the small system of only 4000 neurons. However, the relationship between the system’s genetic heterogeneity and its underlying functional organization remains unknown. Because early gene expression is critical to the development of neural circuits and cell identity, we hypothesize that subpopulations of NA neurons originating from different rhombomeres, differential gene expression domains in the developing brainstem, regulate distinct and specific aspects of respiratory physiology. To test this hypothesis, we will take an intersectional genetic approach that subdivides the NA system into narrowly defined, genetically distinct cohorts that can be functionally characterized. Preliminary studies show that NA neurons are required for the respiratory response to hypercapnic (high CO2) and hypoxic (low O2) conditions, as pharmaco-genetic perturbation of NA-expressing neurons results in reduced ventilatory responses. We also see a decrease in room air ventilation and the hypoxic response when rhombomeres 3&5-derived neurons are perturbed. These data present a working model to query the function of a genetically defined neuron population and the observed phenotypes offer a potential mechanistic understanding for homeostatic disorders associated with noradrenergic abnormalities. Linking these developmentally distinct subtypes of neurons in respiratory homeostasis will give us greater insight into the functional organization of neuromodulatory networks and improve potential therapeutics for life-threatening respiratory disorders. Contributors: Sun, Jenny; Ray, Russell 308 2014 GRADUATE STUDENT SYMPOSIUM DNA METHYLATION ABERRATIONS IN OSTEOSARCOMA Jiayi Monika Sun Program in Structural and Computational Biology and Molecular Biophysics Advisor: Ching Lau, M.D./Ph.D.-Department of Pediatrics Osteosarcoma is the most common type of malignant bone cancer in children and young adults. Little improvement to the long term survival of osteosarcoma patients has been seen in the past 20 years, especially for high-risk patients who develop metastasis or respond poorly to standard therapy. Genetic aberrations associated with osteosarcoma pathogenesis include aberrations of well-known tumor suppressors such as TP53 and RB1 however, very little is known about the epigenetic landscape. DNA methylation is a type of epigenetic regulation by which a methyl group is added to the cytosine of a cytosine-guanine (CpG) dinucleotide. Aberrant DNA methylation has been known to play a major role in the progression of cancer. As part of the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) initiative, we have comprehensively profiled approximately 100 osteosarcoma samples on several genomic platforms. DNA methylation was processed using Illumina’s Infinium HumanMethylation450 BeadChip. This array interrogates more than 480,000 CpG sites, covering 99% of all RefSeq genes and 96% of known CpG islands. In addition to including a majority of the probes from the previous version of the array (HM27) this array also includes an additional type of assay chemistry which is designed to interrogate regions of low CpG density. We evaluated the array’s control probes as well as probes located on sex chromosome to check for quality control and sample annotation integrity. For pre-processing, we performed background subtraction and dyebias correction through an R package called Lumi. A final methylation level called a ‘beta-value’ is calculated from a ratio of the unmethylated(U) and methylated(M) intensities (M/(M+U)) for each site which ranges between 0 and 1, where 0 represents complete demethylation and 1 represents complete methylation. The beta-values are corrected for probe design bias due to the different probe chemistries using the Beta Mixture Quantile Dilation method (BMIQ). Finally, we filtered out any probes with detection p-values greater than 0.05, probes overlapping repeat regions, probes containing SNPs located within 10 bp or at the CpG site and any probes located on the sex chromosomes to correct for gender and potential technical artifacts. Through nonnegative matrix factorization and recursively partitioned mixture modeling we identified two groups within the data with significantly different overall survival. We observed that patients with lower survival were characterized by hypermethylated probes. A very high correlation was observed between DNA methylation and gene expression for FAS, a gene whose expression has previously been shown to correlate with metastatic potential of osteosarcoma patients. We are currently in the process of pursuing this study in functional models. Contributors: Sun, Jiayi; Yu, Alex; Voicu, Horatiu; Lin, Howard; Taylor, Aaron; Shen, Jianhe; Guerra, Rudy; Man, Chris; Lau, Ching 309 BAYLOR COLLEGE OF MEDICINE VACUOLE PROTEASE REGULATES SIR2 TURNOVER DURING AGING IN YEAST Yu Sun Integrative Program in Molecular and Biomedical Sciences Advisor: Weiwei Dang, Ph.D.-Department of Molecular & Human Genetics Sir2 is a class III histone deacetylase that is a key regulator of life span. Sir2 (Silencing Information Regulator) belongs to Sirtuin protein family. Overexpression of Sir2 extends life span and this is conserved among higher eukaryotes. Sir2 protein is lost during aging, leading to high levels of H4K16 acetylation at telomere region that affects yeast replicative life span 1. Previous study has shown that the transcriptional level of Sir2 is not significantly reduced in old cells. We raise the question that what mediates Sir2 loss during aging. We found that neither Ubiquitin-Proteasome System nor macroautophagy is responsible for Sir2 degradation in old cells. From a proteomic study comparing protein profile in old and young cells, Pep4p stands out as one of the most up-regulated proteins in old cells. Pep4p is the vacuolar protease A, required for precursor maturation of most vacuolar proteases and also the homolog of Cathepsin D in mammalian cells. When Pep4 is deleted, Sir2 protein is preserved and even increased in old cells, thus Pep4 is a Sir2 regulator during aging. We tracked Sir2-GFP fusion protein in wild type and pep4∆ old cells and found that Sir2 is largely preserved in the vacuoles in pep4∆ strains, indicating the translocation of Sir2 from nucleus to vacuoles during aging. In pep4∆ strains, the conformation of Sir2 is changed, as shown by limited proteolysis test, while the enzymatic activity of Sir2 is not affected. Immunoprecipitation-Mass Spectrometry study showed that Ssa2 and Ssb2, which belongs to HSP70 protein chaperone family, are potential Sir2 binding factors that are differently regulated in wild type and pep4∆ strains. Co-IP experiment confirmed that Ssa2 and Ssb2 interact with Sir2. Future study will be focused on elucidating the interacting patterns of Ssa2, Ssb2, Sir2 and Pep4, and how this interplay will cause the degradation of Sir2 during aging. Reference: 1. Dang, W. et al. Histone H4 lysine 16 acetylation regulates cellular lifespan. Nature 459, 802–7 (2009). Contributors: Sun, Yu; Berger, Shelley L.; Dang, Weiwei. 310 2014 GRADUATE STUDENT SYMPOSIUM FUNCTIONAL ROLES OF CO-VARYING RESIDUES IN DOPAMINE D2 RECEPTOR Yun-Min Sung Department of Biochemistry & Molecular Biology Advisor: Theodore Wensel, Ph.D.-Department of Biochemistry & Molecular Biology Previous studies of amino acid co-variation in G protein-coupled receptors (GPCRs) mainly focused on predicting structural proximity of residue pairs in order to construct protein 3D structures. However, whether evolutionary amino acid co-variation can be used to detect functional coupling between residue pairs has not been tested. We investigated the functional relationships between evolutionarily correlated residue pairs in GPCR, focusing on dopamine D2 receptors (D2Rs). The D2R ligand binding pocket is predicted to be structurally similar to that of 5-HT2A serotonin receptors (5HT2ARs); however, D2R responds and binds to dopamine better than serotonin, indicating the ability of D2R to discriminate between the neurotransmitters dopamine and serotonin. We hypothesized that residues may work in pairs or larger groups to maintain functional specificity of D2R. By applying the Evolutionary Trace (ET) algorithm to pairs of residues, we first predicted residue pairs in D2R that were likely to co-vary in evolution and replaced the co-varying ET-residues with the corresponding residues in 5HT2AR. By comparing the double swaps with individual swaps, we observed nonadditive effect on the efficacy of G i activation induced by dopamine-stimulated D2R, indicating functional coupling between the ET-residue pairs. We also found dramatic rescue effect on serotonin responses in some of the co-varying ET-residue pairs, suggesting their functional role in discriminating between dopamine and serotonin. Furthermore, we observed that one combined ET-residue swap showed higher binding affinity for serotonin compared to the wild-type, while the individual swaps had no effect on serotonin affinity. The non-additive free energy change upon binding revealed the functional coupling in ligand recognition. Taken together, these findings indicate that key co-varying ET-residue pairs have worked together during evolution to help fine-tune responses to dopamine, whereas other pairs have worked together to help confer selectivity, with the latter property likely more important for conferring selective evolutionary advantage. Contributors: Sung, Yun-Min; Wilkins, Angela D.; Rodriguez, Gustavo J.; Lichtarge, Olivier; Wensel, Theodore G. 311 BAYLOR COLLEGE OF MEDICINE STRICT REGULATION OF STEROID RECEPTOR COACTIVATOR-2 EXPRESSION IS REQUIRED FOR NORMAL ENDOMETRIAL FUNCTION Maria Magdalena Szwarc Department of Molecular & Cellular Biology Advisor: Bert O'Malley, M.D.-Department of Molecular & Cellular Biology Steroid receptor coactivator-2 (SRC-2) is a member of the p160/SRC family of coregulators, which also includes SRC-1 and SRC-3. Members of this coregulator class exert a wide-spectrum of physiological processes from mammary morphogenesis, metabolic homeostasis, to endometrial function. Our group has previously shown that knockout of SRC-2 in the murine endometrium leads to infertility caused by both a defect in embryo attachment and an inability of the endometrium to undergo decidualization, an essential cellular process that enables embryo implantation to occur. Importantly, deregulation of SRC expression levels is a causal factor for many tissue pathologies in both human and mouse. In the case of the endometrium, clinical studies reveal that SRC-2 and SRC-3 levels are elevated in endometrial biopsies from patients diagnosed with polycystic ovary syndrome (PCOS). Importantly, the endometrium of PCOS patients displays severe defects in functionality, including increased endometrial cancer susceptibility and miscarriage rate. Elevated expression of SRC-2 and SRC3 has also been found in hyperplastic and neoplastic endometrium. Collectively, these descriptive findings suggest a causal link between elevated expression of one or both of SRCs and the emergence of these endometrial disorders. To address this proposal further, we engineered a SRC-2 overexpressor (SRC-2: OE) mouse in which high levels of human SRC-2 expression are targeted to cells that express the progesterone receptor. Although ovulation and serum hormone levels are normal, breeding studies show that elevated levels of endometrial SRC-2 result in a severe subfertility defect. Importantly, an artificial decidual response assay revealed that the SRC-2:OE endometrium exhibits an impaired ability to undergo decidualization. The incapacity of the SRC-2:OE endometrium to decidualize is also reflected at the molecular level by significant decrease in the induction of the decidual biomarkers. Furthermore, estradiol treatment reveals that perturbation of SRC-2 levels markedly potentiates estradiol-induced uterine epithelial hyperplasia, providing strong support for SRC-2 in the promotion of unopposed estrogen-action, the main risk factor endometrial cancer development. These studies are supported by our findings in vitro where we have shown that maintenance of high SRC-2 levels is required for endometrial cancer cell growth as shown in colony formation and proliferation assays. Importantly, microarray analysis demonstrated that attenuation of SRC-2 levels in endometrial cancer cells leads to alteration of cellular signaling pathways critical for cell proliferation and differentiation. Also, knockdown of SRC-2 leads to a downregulation of the glycolytic flux and mitochondrial oxidation which is accompanied by a decrease of metabolic intermediates of the pentose phosphate pathway, which is critical for cancer cell growth. We conclude that strict control of SRC-2 expression levels is mandatory not only for normal endometrial functionality but also to prevent unscheduled endometrial hyperplasia which can lead to cancer. Contributors: Szwarc, Maria M; Kommagani, Ramakrishna; Jeong, Jae-wook; Wu, San-Pin; Lanz, Rainer B; Putluri, Nagireddy; Tsai, Sophia Y; Tsai, Ming-Jer; DeMayo, Francesco J; Lydon, John P; O’Malley, Bert W 312 2014 GRADUATE STUDENT SYMPOSIUM EARLY ACTIVATION OF P2Y2 PURINERGIC SIGNALING IS ESSENTIAL FOR EFFICIENT HEPATOCYTE PROLIFERATION IN RESPONSE TO PARTIAL HEPATECTOMY Bryan Christopher Tackett Program in Translational Biology & Molecular Medicine Advisor: Sundararajah Thevananther, Ph.D.-Department of Pediatrics Saul Karpen, M.D./Ph.D.-Department of Pediatrics Background & Aims. Partial hepatectomy (PH) induces hepatocyte proliferation via step-wise induction of immediate early genes, reorganization of extracellular matrix, and cytokine and growth factor-mediated signaling early on during liver regeneration. However, the identity of initial trigger(s) of liver regeneration has remained elusive. ATP is released into the extracellular milieu within minutes of 70% PH. We hypothesized that extracellular ATP, via activation of its cognate cell-surface P2Y2 purinergic receptors, might play a key role in the induction of early events critical for hepatocyte proliferation in regenerating livers. Methods. Wild type (WT) and P2Y2 purinergic receptor knockout (P2Y2-/-) mice were subjected to 70% PH and liver tissues were analyzed for efficiency of hepatocyte priming and proliferation. Influence of extracellular ATP and P2Y2 puringeric receptor signaling on hepatocyte proliferation was evaluated in vitro. Results. Our findings suggest that hepatocyte proliferation in response to 70% PH was impaired in P2Y2-/- mice. Early activation of p42/44 MAPK (ERK, 5 min), early growth response-1 (Egr-1) and activator protein-1 (AP-1) DNA-binding activity (30 min) were attenuated in the remnant livers of P2Y2-/-. Correspondingly, Egr-1 and AP-1 target gene and a key mediator of extracellular matrix remodeling, matrix metalloprotese-9 (MMP-9) protein induction and HGFα/c-Met signaling were attenuated in P2Y2-/-. Extracellular ATP alone, via the activation of P2Y2 purinergic receptors, was sufficient to activate ERK/Egr-1 and proliferation of primary mouse hepatocytes in vitro. Conclusions. Extracellular ATP-mediated rapid activation of P2Y2 purinergic receptors plays a key role in the initiation of hepatocyte proliferation in response to PH in mice. Contributors: Tackett, Bryan; Sun, Hongdan; Cheruvu, Sayuri; Mei, Yu; Mani, Arunmani; Hernandez-Garcia, Andres; Vigneswaran, Nadarajah; Karpen, Saul; Thevananther, Sundararajah 313 BAYLOR COLLEGE OF MEDICINE LOSS OF FUNCTIONAL KCA1.1 CHANNELS IMPAIRS MYOGENESIS IN MYOTONIC DYSTROPHY TYPE 1 Rajeev Babu Tajhya Department of Molecular Physiology & Biophysics Advisor: Christine Beeton, Ph.D.-Department of Molecular Physiology & Biophysics Myotonic dystrophy type 1 (DM1) is an autosomal dominant neuromuscular disorder that primarily causes skeletal muscle wasting. This muscle wasting, due to reduced myogenesis, is the main cause of disability in patients and there is no cure for DM1. Errors in alternative splicing due to accumulation of the splice regulators MBNL and CUGBP at (CUG)n hairpin structures cause prolonged or persistent myoblast proliferation and delayed myotube fusion. However, the downstream pathways contributing to reduced myogenesis remain unclear. K+ channels regulate proliferation of myoblasts and differentiation into myotubes by triggering changes in membrane potential, cell volume and regulating Ca2+ signaling. Therefore, we investigated the roles of K+ channels in primary myoblasts obtained from patients with DM1 and healthy donors. We have identified loss of calcium-activated and voltage-dependent functional K+ channel KCa1.1 at the plasma membrane of DM1 myoblasts by immunofluorescence and flow cytometry, and have shown loss of paxilline-sensitive K+ currents by patch-clamp. We hypothesized that the loss of KCa1.1 in DM1 myoblasts plays role in the delayed myogenesis, and that over-expressing KCa1.1 channels will rescue the pathological features in vitro. We found that inhibition of KCa1.1 in myoblasts from healthy donors enhanced proliferation, lowered matrix metalloproteinase-2 (MMP2) production, impaired wound healing, and decreased myotubes formation – all features of DM1 myoblasts, demonstrating that functional KCa1.1 channels are required for normal proliferation, migration and myotube fusion. On the contrary, over-expressing KCa1.1 in myoblasts from patients with DM1 normalized their proliferation to the levels observed in healthy myoblasts. We also showed that blocking KCa1.1 in healthy myoblasts increases activated-NFκB which is often associated with muscle wasting. We conclude that the loss of KCa1.1 in DM1 impairs early stage of myogenesis possibly by affecting Ca2+-mediated pathways. This suggests that KCa1.1 modulation could be utilized as a novel strategy to rescue delayed myogenesis in DM1. Contributors: Tajhya, Rajeev; Hu, Xueyou; Tanner, Mark; Timchenko, Lubov; Beeton, Christine 314 2014 GRADUATE STUDENT SYMPOSIUM THE E3 UBIQUITIN LIGASE, ARIADNE-1 (ARI-1), REGULATES SYNAPTIC ACTIVE ZONE DEVELOPMENT IN DROSOPHILA AND IS REQUIRED FOR PROPER SYNAPTIC TRANSMISSION Kai Li Tan Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics Synaptic transmission involves various cellular activities, including protein synthesis and degradation, vesicular and protein trafficking, and fusion and fission of membranes. Orchestrating these activities is important to ensure optimal synaptic function. In a forward genetic screen on the Drosophila X-chromosome to identify essential genes required for proper synaptic transmission, we identified mutations in ari1, the Drosophila homolog of mammalian HHARI. ari-1 is a highly conserved gene, which encodes a ring-between-ring E3 ubiquitin ligase. By recording electroretinograms, we found that mutant photoreceptors display a loss of ON-transients when activated by light, indicating defects in neurotransmission. To further characterize this synaptic transmission defect, we turned to the 3rd-instar larval neuromuscular junction (NMJ). We performed electrophysiological recordings and found that postsynaptic muscle cells show an increased excitatory junction potential (EJP) upon stimulation when compared to the control. To identify the cause of the increased evoked response, we performed transmission electron microscopy and immunostaining for different components in the synapse, and found that the size of the synaptic vesicles and the amount of postsynaptic receptors remained unchanged. However, the number of active zones (indicated by Bruchpilot staining), the sites in the synapse where vesicles cluster to allow neurotransmitter release, is increased in ari-1 mutants. These additional active zones are potentially functional, since increasing the level of a voltage-gated calcium channel, Cacophony (Cac) in ari-1 mutant background leads to early animal lethality, which is likely caused by calcium mediated excito-toxicity. A core component of the active zone, Bruchpilot (Brp), is homologous to mammalian ELKS, where it functions to recruit Cac to the active zone. It is likely that Ari-1 directly regulates the level of Bruchpilot, possibly through its ubiquitination activity. However, over-expressing Brp in the neuron does not alter the physiological properties and active zone distribution of the synapse. This indicates that either Ari-1 directly regulates Brp level locally at the synapse, or Ari-1 indirectly affects Brp level or distribution in the synapse by regulating other proteins. In order to understand the molecular mechanism of Ari-1 in promoting optimal synaptic transmission, we will identify the ubiquitinated targets of Ari-1 in the neuron through biochemical assays. Contributors: Tan, Kai Li*; Haelterman, Nele*; Nagarkar, Sonal; Lee, Pei-Tseng; Bellen, Hugo 315 BAYLOR COLLEGE OF MEDICINE BLOCKING KCA1.1 CHANNELS INHIBITS THE INVASIVE PROPERTIES OF FIBROBLAST-LIKE SYNOVIOCYTES AND REDUCES DISEASE SEVERITY IN ANIMAL MODELS OF RHEUMATOID ARTHRITIS Mark R. Tanner Program in Translational Biology & Molecular Medicine Advisor: Christine Beeton, Ph.D.-Department of Molecular Physiology & Biophysics David Corry, M.D.-Department of Medicine Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease of largely unknown etiology that mainly affects diarthrodial joints, leading to joint destruction, pain, and decreased mobility. The fibroblast-like synoviocyte (FLS) is a resident synovial cell that has a central role in RA pathogenesis, including the formation of synovial hyperplasia and increased invasiveness, along with increased cytokine, growth factor, and protease release. These factors contribute to cartilage and bone degradation within the joint. Currently, no RA therapy has been developed to specifically target FLS. We have found that FLS from patients with RA and from rats with the pristaneinduced and collagen-induced arthritis models of RA have increased expression of KCa1.1 at their plasma membrane when compared with FLS from patients with osteoarthritis or from healthy rats, respectively. Selectively blocking the function of this channel reduces many of the pathogenic aspects of RA-FLS, including decreasing proliferation, invasiveness, and release of cytokines, chemokines, angiogenic factors, and proteases. Furthermore, decreasing the amount of KCa1.1 expressed in FLS reduces their invasiveness and opening or increasing membrane expression of KCa1.1 increases invasiveness. This altered invasiveness is likely a result of KCa1.1’s regulation of the FLS cytoskeleton, as blocking KCa1.1 increases the channel’s association with actin and causes filamentous actin to have a modified cellular distribution. We have also found that inhibiting KCa1.1 with a selective small-molecule blocker after onset of clinical signs of arthritis significantly reduced disease severity in both the collagen-induced and pristane-induced rat models of RA, along with reducing the ex vivo invasiveness and proliferation of FLS from blocker-treated animals. These studies indicate the importance of KCa1.1 as a novel target for RA and emphasize the potential efficacy of directly inhibiting FLS in reducing the severity of this debilitating disease. Contributors: Tanner, Mark R.; Hu, Xueyou; Huq, Redwan; Tajhya, Rajeev; Khan, Fatima S.; Gulko, Percio; Beeton, Christine 316 2014 GRADUATE STUDENT SYMPOSIUM MOLECULAR PROFILING OF PEDIATRIC EPENDYMOMAS REVEALS PROGNOSTIC SUBTYPES Aaron Michael Taylor Program in Structural and Computational Biology and Molecular Biophysics Advisor: Ching Lau, M.D./Ph.D.-Department of Pediatrics Background. Ependymomas (EPN) account for ~10% of intracranial tumors in children, typically presenting in the first five years of life. Several studies to date have described subtypes of EPN by mRNA expression and DNA copy number that have prognostic significance. Still, there has been little improvement in survival and the search for effective targeted therapy continues. We seek to increase our understanding of EPN by adding phenotypes defined by mRNA regulation in an exclusively pediatric cohort of patients, and in doing so unlock new routes for therapeutic intervention. Materials & Methods. We performed expression and miRNA profiling of 65 cases of pediatric infratentorial EPN collected from a single clinical protocol (TCH n = 16 and Children’s Oncology Group n = 49), and 16 normal brain controls using the Affymetrix U133 Plus 2.0 and ABI TaqMan platforms (367 miRNAs), as well as copy number profiling using the Affymetrix SNP array on a subset of 54 samples. Subjects ranged between 1 day & 18 years of age, with 51% under the age of four. This patient cohort was consistent with the disease epidemiology, with 38% anaplastic cases (WHO grade III). All but 5 received postoperative radiotherapy, and 75% of initial surgeries were complete resections. Results. Examination of the two current infratentorial subtypes PFA and PFB, as defined by a literature-based gene expression signature, revealed no prognostic differences (PFS log rank p = 0.890). However, supervised mRNA-based profiling based on age at diagnosis demonstrated further within-phenotype heterogeneity dominated by differential homeobox (HOX) gene expression. Utilizing this signature we were able to separate all PFB cases with genomic instability and better outcome from PFB cases with young age, poor prognosis, and absent expression of the anterior HOX genes, which are involved in embryogenesis and have been associated with several pediatric brain tumors. Furthermore, miRNA within HOX genomic loci, which regulate the HOX gene group, also show low expression in this latter subtype, whereas the homeobox homologue gene EN2 and co-regulated gene CNPY1, markers of early midbrain-hindbrain development, are strongly overexpressed. The elucidation of this novel phenotype, designated PFBHOX-, restores prognostic value to the existing subgroup designations (PFS log rank p = 0.0133). Conclusions. Examination of existing infratentorial phenotypes in our all-pediatric samples did not appear prognostic, but reproducible mRNA-based subtypes within the known phenotypes were discovered. These subtypes, defined by an absence of anterior HOX genes expression and an overexpression of EN2 and CNPY1, revealed strong differences in clinical outcome. We intend to validate these markers in an independent set of EPNs while further exploring their biological implications. Contributors:Taylor, Aaron; Burstein, Matthew D.; Shen, Jianhe; Chow, Thomas; Su, Jack; Adesina, Adekunle; Dauser, Robert; Whitehead, William; Jea, Andrew; Curry, Daniel; Chintagumpala, Murali; Lau, Ching C. 317 BAYLOR COLLEGE OF MEDICINE EFFECT OF CELLULAR ELIMINATION ON THE CIRCUIT INTEGRATION OF ADULT BORN NEURONS IN OLFACTORY BULB Burak Tepe Program in Developmental Biology Advisor: Benjamin Arenkiel, Ph.D.-Department of Molecular & Human Genetics The mammalian brain has a remarkable capacity for continued neurogenesis throughout life. Every day, thousands of adult-born neurons are generated in the subventricular zone and migrate anteriorly along the rostral migratory stream where they eventually populate the granule cell layer of the olfactory bulb (OB). During this process, however, only fifty percent of adult-born neurons will survive long-term to populate the OB circuitry. This dramatic reduction suggests that certain selection events act on these neurons to determine which ones will be integrated. To elucidate how survival dynamics modulate adult-born neuron maturation, plasticity, and integration in the brain, we hypothesize that the balance between the death rate of resident granule cells (GC), and survival rate of adult-born neurons, determines the integration efficiency of adult born neurons. To determine if integration of adult-born neurons depends on the loss of resident neurons, we propose to increase neuronal cell death in the OB and determine the effects on long-term survivability and circuit integration of incoming adult-born neurons. To do so, we genetically targeted resident OB interneurons for toxin-mediated cellspecific ablation. Upon death of resident OB neurons, we injected animals with the proliferation marker EdU to follow a population of adult-born neurons and investigate their integration probability and spatial distribution. Preliminary data suggest that the loss of resident GCs increases the integration probability of newly integrating neurons. To examine the increased survival of resident and adult born GCs, we generated a novel mouse strain which we have targeted the ROSA26 locus for insertion of a Cre recombinase-dependent expression construct that contains viral apoptosis inhibitor p35. By injecting these mice with Cre-expressing virus at different time points of adult-born neuron development, we propose to increase the survival of these neurons and observe their integration efficiency. Similarly, we will target resident OB neurons for inhibition of apoptosis by targeting them with a Cre-expressing virus while assessing effects on adult-born neuron integration. Contributors: Tepe, Burak; Arenkiel, Benjamin R. 318 2014 GRADUATE STUDENT SYMPOSIUM EVOLUTIONARY TRACE-GUIDED MUTATIONS TO GENERATE D2 DOPAMINE RECEPTOR EFFECTOR BIAS IN RESPECT TO THE BETA-ARRESTIN2 PATHWAY María Elisa Terrón-Díaz Integrative Program in Molecular and Biomedical Sciences Advisor: Olivier Lichtarge, M.D./Ph.D.-Department of Molecular & Human Genetics One-third of all available medications target G-Protein Coupled Receptors (GPCR). Therefore, specificity towards ligands and downstream effectors is important for understanding protein function and the development of selective therapeutic drugs with fewer side effects. The serotonin (5-HT2AR) and D2 dopamine (D2R) receptors are both part of the bioamine subfamily of GPCRs and have similar sequence identity, however, they discriminate stringently in their cellular responses between endogenous agonists. Using Difference Evolutionary Trace (ET) analysis, functional determinants were previously identified and rationally targeted by swapping residues from 5-HT2AR into D2R. This swap altered D2R ligand potency, affinity, and specificity. Beyond the ligand bias, we aim to utilize these sixteen D2R mutants to determine the effector bias in respect to the β-arrestin2 pathway, involved in receptor desensitization and resensitization, using a live cell-based assay known as Bioluminescent Resonance Energy Transfer2 (BRET2). The BRET2 assay is being used to detect the proximity between D2R-RlucII and beta-arrestin2-GFP10 at less than a distance of 100Å. Five plasmids for the BRET assay have been generated: GFP10 fused to RLucII (positive control), GFP10 (negative control), RLucII (negative control), D2R-RLucII (donor), and beta-arretin2-GFP10 (acceptor). Plasmid titrations are currently being tested and sitedirected mutagenesis is being performed on D2R-RLucII to generate the 16 pointmutations. We hypothesize that one or more of the 16 D2R mutants that where previously shown to have ligand bias will also have beta-arrestin2 bias. The betaarrestin2 bias will be detected in the BRET assay as an increased GFP10/RLucII signal in the mutant D2R after agonist treatment as compared to wild type. This project will give light as to how allosteric regulation of D2R can influence effector bias aiding in the understanding of pathway selectivity. Contributors: 319 BAYLOR COLLEGE OF MEDICINE SECRETED B. ANTHRACIS PROTEASES TARGET HEMOGLOBIN AS AN AMINO ACID SOURCE Austen Lee Terwilliger Integrative Program in Molecular and Biomedical Sciences Advisor: Anthony Maresso, Ph.D.-Department of Molecular Virology & Microbiology Bacterial infections are sustained by acquiring essential nutrients (e.g. carbon, nitrogen, phosphorous, sulfur, and metals) from the host to support cellular metabolism and replication. Necessary for bacterial survival, numerous studies focus on iron and its acquisition systems within blood borne pathogens, many of which target the oxygen carrier protein hemoglobin as an iron reservoir. However amino acid acquisition systems, supplying carbon, nitrogen and sulfur, remain uncharacterized and understudied as therapeutic targets. Blood, the likely source of nutrients during bacteremia, contains free amino acids as well as a multitude of plasma proteins that could fulfill a bacterium’s nutritional needs. Employing a novel defined medium to mimic human blood serum and Bacillus anthracis as a model pathogen, we determined that hemoglobin and other serum proteins can serve as an amino acid source during nutrient depravation. This effect is independent of the iron-bound heme in hemoglobin, and genetic analysis reveals the immune inhibitor metalloprotease, InhA1, as the main proteolytic contributor. These studies highlight the emergence of amino acid acquisition as a novel point for therapeutic intervention and challenge a previous model, which views hemoglobin solely as an iron source for bacterial pathogens during infection. Contributors: Austen Terwilliger, Anthony Maresso 320 2014 GRADUATE STUDENT SYMPOSIUM THE DEVELOPMENT OF HLANULLCD1D-EXPRESSING K562 CELLS AS ARTIFICIAL ANTIGEN-PRESENTING CELLS FOR SELECTIVE AND EFFICIENT CLINICAL SCALE NKT CELL EXPANSION Gengwen Tian Department of Pathology & Immunology Advisor: Leonid Metelitsa, M.D./Ph.D.-Department of Pediatrics CD1d-restricted type-I Natural Killer T cells (NKTs) have been shown to mediate antitumor responses in mouse models and are associated with improved outcome in several types of cancer. However, the therapeutic application of NKTs has been limited by low numbers and functional defects of these cells in patients with cancer. To provide a means for safe and effective ex-vivo NKT-cell expansion for cell and gene therapy applications we explored native and engineered properties of K562 cells to function as artificial antigen-presenting cells (aAPC). Although clinical grade K562-based aAPC products have been shown to be effective for T- and NK-cell expansion for therapeutic use in patients, K562 cells express HLA-Cw3, an NK-cell activating ligand which stimulates expansion of NK and potentially alloreactive T cells that causes competition with NKTs in culture and poses a clinical hazard. Because HLA-C is the only HLA allele in K562, we rendered K562 cells HLAnull by eliminating HLA-C gene from K562 cell genome using a HLA-C-specific zinc finger nuclease. We then transduced parental and HLAnull K562 cells with CD1d cDNA followed by single cell sorting and clonal expansion. The clones were pulsed with αGalactosylceramide and tested as aAPC for NKTs using CFSE proliferation assay. We found that in contrast to K562/CD1d, HLAnullK562/CD1d clones selectively expanded NKTs when added to primary PBMC, and clones with an intermediate level of CD1d expression induced the highest rate of NKT-cell proliferation. Next, a selected HLAnullCD1dmed clone was further modified to express CD86 alone or in combination with 4-1BBL and/or OX40L followed by single cell sorting and clonal expansion. The comparison of the APC function allowed us to select a clone with the phenotype HLAnullCD1dmedCD86highOX40Lmed4-1BBmed (B8-2) which consistently induced the highest rate of NKT-cell expansion. B-8-2 cells were able to efficiently expand both primary NKTs as well as NKTs modified to express a chimeric antigen receptor (CAR) specific to CD19 antigen, a clinically validated therapeutic target for B-cell malignancies. CAR expression in NKTs rendered them highly cytotoxic against CD19+ Raji leukemia cells while preserving their CD1drestricted specificity. Importantly, CAR-NKTs expanded with B-8-2 cells exhibited a Th1skewed cytokine profile and had potent therapeutic activity in Raji leukemia model. Therefor, the engineered K562 B-8-2 can be used as highly efficient aAPC for ex vivo propagation of primary human as well as gene modified NKTs amenale for adoptive cell therapy application for human patients. Contributors: Gengwen Tian1, Bipulendu Jena2, Daofeng Liu1, Andras Heczey1, Hiroki Torikai2, Dean Lee2, Laurence Cooper2, and Leonid Metelitsa11Texas Children's Cancer Center, Center for Cell and Gene Therapy, Departments of Pediatrics, Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030 2Cell Therapy, Children's Cancer Hospital, Department of Immunology, MD Anderson Cancer Center, Houston, TX, 77030 321 BAYLOR COLLEGE OF MEDICINE T LYMPHOCYTES FUNCTION TO NORMALIZE TUMOR-ASSOCIATED VASCULATURE BY INCREASING PERICYTE COVERAGE Lin Tian Department of Biochemistry & Molecular Biology Advisor: Xiang Zhang, Ph.D.-Department of Molecular & Cellular Biology Tumor-associated vasculatures are usually considered to be abnormal and lack the support of pericytes. High percentage of pericyte coverage is considered to be the good prognosis marker for some cancers such as colorectal cancer and breast cancer. However, the field lacks the information on whether the adaptive immune system is involved in tumor-associated vascular normalization. We utilized a p53-null murine mammary tumor model to determine the role of adaptive immune cells in vascular normalization. The p53-null tumor pieces were inoculated into the mammary gland of immunocompetent (Balb/c) mice and immunodeficient (nude) mice. We found that pericyte coverage in tumors of Balb/c mice was significantly higher than that of nude mice. To directly test the function of T lymphocytes in vascular normalization, we reconstituted T lymphocytes in tumor bearing nude mice, and found that pericyte coverage of tumor-associated vasculatures increased significantly in nude mice with T cell reconstitution. This study indicates that adaptive immune cells function to normalize tumor-associated vasculature by increasing pericyte coverage, which might prevent cancer cells entering into circulation and further decrease metastasis frequency. Contributors: None 322 2014 GRADUATE STUDENT SYMPOSIUM KCNQ2 PORE MUTATIONS CAUSING SEVERE EPILEPSY DISRUPTS CHANNEL TARGETING TO THE AXON INITIAL SEGMENT Baouyen Tran Department of Neuroscience Advisor: Edward Cooper, M.D./Ph.D.-Department of Neurology KCNQ2/3 voltage-gated potassium channels underlie the M-current (IM) regulating neuronal excitability. About 150 mutations in KCNQ2 have been published, about half leading to mild neonatal-onset epilepsy (Benign Familial Neonatal Seizures) and half leading to severe epileptic encephalopathy. High concentrations of functional KCNQ2/3 channels are found at the axon initial segment (AIS) where they exert control over action potential generation. Experimental determination of the pathophysiological mechanisms in severe KCNQ2 encephalopathy is important for guiding therapy in this disorder. Candidate mechanisms include alterations in voltage-dependent gating, trafficking, or stability. We used immunofluorescence and confocal imaging mice to determine localization of KCNQ2/3 in both the cortex and hippocampus of transgenic hKCNQ2-G279S, and in transfected cultured rat hippocampal neurons. We used surface biotinylation assays in CHO cells transfected with mutant KCNQ2 in order to determine whether or not localization of the KCNQ2/3 channels are altered or abnormally degraded. Immunofluorescence microscopy performed on tissue sections from transgenic mice over-expressing the dominant negative mutant G279S revealed a strikingly aberrant pattern: KCNQ2 was completely absent from the AIS and was retained at numerous intracellular puncta in the soma and dendrites. KCNQ3 weakly labeled the AIS, and was partially redistributed to these puncta. Voltage-gated sodium channel concentration at the AIS was normal. In addition, cultured hippocampal neurons electroporated with mutant KCNQ2 at the pore show absence or abnormal pattern of KCNQ2 labeling at the AIS at DIV7. However, surface biotinylation assays in CHO cells show no changes in mutant KCNQ2’s ability to be detected at the surface. Some KCNQ2 mutations may act by preventing trafficking to the AIS, leading to an imbalance in sodium and potassium channel activity, excessive action potential initiation, and cellular hyperexcitability. KCNQ2 pore mutations appear to fall into this category. Since such effects that may not be easily revealed through heterologous expression, further development of neuronal expression and in vivo models is warranted. Contributors: Tran, Baouyen; Li, Li; Xu, Mingxuan; Cooper, Edward 323 BAYLOR COLLEGE OF MEDICINE ROLE OF P53 IN MYODIFFERENTIATION DURING PROSTATIC DISEASE PROGRESSION Linda Lien Tran Integrative Program in Molecular and Biomedical Sciences Advisor: David Rowley, Ph.D.-Department of Molecular & Cellular Biology In prostate cancer, reactive stroma is observed to co-evolve with disease and be indicative of rate of progression, rate to biochemical recurrence after prostatectomy, and mortality. Previous xenograft studies in our lab involving the recombination of carcinoma and stromal cells have shown that reactive stroma is tumor promoting. Myofibroblasts are a key component of this microenvironment, yet little is known about the mechanisms guiding their expansion differentiation. p53 is a transcription factor that has been widely studied for its role in the regulation of DNA damage response. Modifications to this protein can determine whether a cell is fated to apoptosis or cell cycle arrest. The Cooney lab and collaborators have implicated p53 in the differentiation potential of mouse embryo fibroblasts (MEFs). p53-deficient MEFs spontaneously differentiated into several different cell types. RUNX1 is a transcription factor critical in normal hematopoiesis. Recent work performed in the Rowley lab has identified RUNX1 as a key transcription factor in TGFeta1-mediated activation of human prostate mesenchymal stem cells (hpMSCs) to myofibroblasts. These data indicate a role for RUNX1 in stem cell biology and multipotency. While p53 status appears to be important for differentiation commitment and resistance to plasticity, RUNX1 seems to be essential for transit amplification of MSCs and maintenance of their plasticity. RUNX1 is known to complex with p53 and affect transcriptional activity of downstream target genes. Therefore RUNX1 and p53 may be coordinating MSC expansion and myofibroblast differentiation. We hypothesize that as RUNX1 levels decrease with myodifferentiation, p53 levels/activity will increase, signaling the commitment of these cells to a specific lineage and removing them from the multipotent pool. To determine the correlation of p53 levels with myodifferentiation, hpMSCs were treated with TGF-beta1 and RNA isolated at 6, 12, 24, and 48 hours. To investigate whether RUNX1 regulates p53 expression, RUNX1 was knocked down in hpMSCs using siRNA and treated with TGF-beta1. RNA was isolated at 6, 12, 24, and 48 hours and analyzed by qRT-PCR. Preliminary studies show that RUNX1 knockdown appears to raise p53 threshold expression levels and illicit a more pronounced response to TGFbeta1 treatment. Interestingly, there was a decrease in p53 expression at 12 and 24 hours; timepoints at which reactive stroma markers TNC and ACTA2 were at their peak expression. This data is to be corroborated with protein expression via Western blots and IHC. Contributors: Tran, Linda; Cooney, Austin; Rowley, David 324 2014 GRADUATE STUDENT SYMPOSIUM HOW CHEMOATTRACTANT AND MECHANOSENSORY CUES ARE INTEGRATED DURING EPIDERMAL WOUND CLOSURE IN DROSOPHILA Chang-Ru Tsai Program in Developmental Biology Advisor: Michael Galko, Ph.D.-Biochemistry and Molecular Biology Efficient wound closure is critical for survival and is conserved across species. Epidermal cell migration into the wound gap requires both soluble chemoattractants and mechanical cues. However, it remains mysterious how these different types of directional information are integrated by the migrating cell. Previously our lab identified the Drosophila Vascular Endothelial Growth Factor (VEGF) ligand and receptor (VEGFR) as wound closure genes. Surprisingly, VEGFR appears to function as both a chemoreceptor and a mechanoreceptor during wound healing, suggesting integration of chemical and mechanical cues may occur at the level of the same receptor. Both the VEGF ligand and mechanical stimuli/wounding act through the downstream transcription factor Yorkie (Yki). Yki is also required for wound closure but is typically thought to function in the context of Hippo signaling, a growth control signaling pathway. Upon wounding, Yki translocates from the cytoplasm into the nucleus nearly instantaneously and even at large distances from the wound. A major goal of my project is to figure out exactly how this spectacular response occurs. With the power of fly genetics, I will manipulate the expression pattern and function of both VEGF and VEGFR in the epidermis and monitor Yki translocation upon mechanical stimulation and/or wounding. This approach will test the roles of VEGF and VEGFR during both signal initiation and propagation. Second, to obtain more detailed insights into the unique properties of the VEGF/VEGFR/Yki signaling axis, an in vivo structure/function study of these factors will be performed. Different genomic deletion constructs of VEGFR and VEGF will be engineered into VEGFR and VEGF mutant backgrounds, respectively, and their abilities to rescue chemoattraction and mechanotransduction will be assessed. This project is likely to lead to fundamental insights into how cells respond to damage cues and perceive their mechanical environment. Given that both the VEGFR and Hippo/Yki signaling pathways are important for cancer signaling, these insights will likely be relevant not just to wound healing but also to cancer metastasis and tissue growth regulation. Contributors: Tsai, Chang-Ru; Michael Galko 325 BAYLOR COLLEGE OF MEDICINE THE ROLE OF G3BP1 ARGININE METHYLATION IN STRESS GRANULE ASSEMBLY AND ANTIVIRAL Wei-Chih Tsai Department of Molecular Virology & Microbiology Advisor: Richard Lloyd, Ph.D.-Department of Molecular Virology & Microbiology Stress granule (SG) is formed when eukaryotic cells cope with different external stresses, including virus infection. These stressors are sensed by different eIF2a kinases, and eventually shutdown global translation initiation. In the meanwhile, RasGAP-SH3-binding protein 1 (G3BP1) will be dephosphorylated, targeting to stalled mRNP/mRNA complexes to nucleate SG assembly. In contrast, viruses have evolved to manipulate SG formation via digesting or sequestering G3BP1 and other essential SG components. Our previous data has demonstrated the localization and activity of PKR in G3BP1 induced SGs. In addition, expression of G3BP1 dampens enteroviruses titer and stimulates NFκB reporter activity. Given the important role for SGs in innate immune response; however, it is still unclear how G3PB1 be regulated in this process. Type I protein arginine methyltransferases (PRMT) are the family of enzymes in eukaryotic cells that are responsible for arginine methylation in the RGG domains of many RNA-binding proteins, including G3BP1. Herein, we demonstrated a dramatic reduction of SG assembly in stressed PRMT3 (40%) and PRMT4 (80%) knockout mouse embryonic fibroblast (MEFs). These data implies the importance of arginine methylation in SG formation. Sequence analysis of G3BP1 reveals a C-terminus RGG domain where contains four potential arginine methylation sites. Deletion of the RGG domain abolished SG assembly in stressed G3PB1 KO MEFs and repressed NFκB reporter activity. In addition, there was an average of 40% reduction of SGs with the 435, 443, or 447 methyl mimetic (R to F) mutants. Taken together, these data illustrate that arginine methylations are new SG regulation signals in the RGG domain of G3PB1. That can also modulate innate immunity by controlling NFκB activation. Stress granules (SGs) are dynamic cytoplasmic structures, which contain aggregates of translationally stalled messenger ribonucleoprotein (mRNP) complexes. SGs are formed in response to environmental stressors such as heat shock, oxidative stress, amino acid deprivation, and viral infection. Protein arginine methyltransferases (PRMTs) are the enzymes responsible for arginine methylation, which provides a means to modulate the charge and localization of target proteins. A central component of SGs is Ras-GTPase activating protein SH3 domain-binding protein 1 (G3BP1), which contains an arginine-rich RGG domain in the C-terminus. It has been shown that PRMT1 can also bind in this region; however, the role of methylation in SGs dynamics is yet to be determined. The first aim of this study is to investigate whether G3BP1 methylation is required for SG formation. We found three potential methylation sites (R435, R443, R447) in the RGG region of G3BP1. In order to characterize the functional significance of these sites, I will generate methylation mimics (R to F) and deficient mutants (R to K). These mutants will be transfected into U2OS cells and microscopy will be used to study SG behavior under arsenate and thapsigargin stresses. It is reasonable to infer that G3BP1 will switch its binding partners after changing its charge. The second aspect of this aim is to verify the components of G3BP1 complex to depict the possible biological function of G3BP1 via pull down assay and Ingenuity pathway analysis software. The second aim of this study will be to investigate the role of methylation in SG dynamics. For this aim, methylation deficient mouse embryonic fibroblasts will be used to ablate specific methylation pathways. Stress granules will be induced in these cells and stress granule assembly and disassembly kinetics will be monitored by immunofluorescence microscopy. Additionally, in vitro and in vivo methyltransferase assays using PRMTs found to be important in the above SG dynamics studies will be employed with G3BP1 as a substrate. These studies will provide insight into whether R435, R443 and R447 are modified by PRMTs. The last part of this study will focus on the relationship between innate immunity and methylation modification on G3BP1. First, I will overexpress those mutants in either G3BP knockout or PRMT knockout cells. Then infect cells with enterovirus to investigate viral replication rates. If methylation effects viral replication, I will investigate cytokine expression to determine whether SG modulation of various cytokines is partly responsible for the observed changes in virus replication. Contributors: Wei-Chih Tsai, Lucas C. Reineke, Jon Dougherty, Richard E. Lloyd 326 2014 GRADUATE STUDENT SYMPOSIUM THE CELL-ADHESION GPCR BRAIN-SPECIFIC ANGIOGENESIS INHIBITOR 1 REGULATES DENDRITICSPINE MORPHOLOGY AND SYNAPTOGENESIS Yen-Kuei Tu Integrative Program in Molecular and Biomedical Sciences Advisor: Kimberley Tolias, Ph.D.-Department of Neuroscience Synapses are specialized sites that mediate communication between neurons. Most excitatory synapses in the brain reside on actin-rich structures called dendritic spines. The formation, regulation and maintenance of excitatory synapses are crucial for normal cognitive function. Recently, we have identified the adhesion G protein-coupled receptor (GPCR) brain-specific angiogenesis inhibitor 1 (BAI1) as a key regulator of synapse development. We show that BAI1 is highly localized to spines. Furthermore, knockdown of BAI1 results in decreased spine and synapse density in both cultured hippocampal neurons and cortical neurons from intact mouse brains. Synaptic loss caused by BAI1 knockdown can be rescued by full-length BAI1, but not by a BAI1 truncation mutant, which fails to interact with the Par3/Tiam1 polarity complex. Tiam1 is a Rac1-guanine nucleotide exchange factor (GEF) that promotes spine and synapse development by inducing Rac1-dependent actin remodeling. Tiam1 is restricted to spines by the polarity protein Par3, enabling for spatial control of Rac1 activation. We show that BAI1 regulates spine and synapse development by recruiting the Par3/Tiam1 complex to spines, resulting in localized Rac activation and actin polymerization. Although these findings elucidate how BAI1 signals inward to promote post-synaptic development, it is unclear whether BAI1 also signals across the synapse to induce pre-synaptic differentiation. Utilizing a COS7 cell-neuron co-culture system, we show that BAI1 increases pre-synaptic termini formation on the axons of cultured hippocampal neurons that contact BAI1-expressing COS7 cells. These results indicate that BAI1 can induce pre-synaptic as well as post-synaptic development. Our future investigations are twofold: (1) We have also shown that BAI1 interacts with important ligands that regulate synaptic development such as Neuroligin-1 and the complement factor C1ql3. Investigating the importance of N-terminal domains that interact with these ligands will further elucidate mechanisms for BAI1's regulation of synaptogenesis. (2) Given that the two other BAI family members are also highly expressed in the brain, affect neuron morphology, and have been linked to neurological disease, we are investigating their role as regulators of spine and synapse development. Results from our study should help to elucidate the mechanisms that regulate excitatory synapse development, and provide potential therapeutic targets for the treatment of neurological disease. Contributors: Tu, Yen-Kuei; Duman, Joseph; Tolias, Kimberley 327 BAYLOR COLLEGE OF MEDICINE BIASED AGONISM OF TOLL LIKE RECEPTOR 4 IN MEDIATING IMMUNE SYSTEM Hui-Ying Tung Department of Pathology & Immunology Advisor: David Corry, M.D.-Department of Medicine The immune basis of allergic versus non-allergic diseases differ vastly, however, both utilize the immune pattern like recognition receptor- Toll like receptor (TLR) 4. While the canonical TLR4 ligand, lipopolysaccharide or LPS, induces innate and adaptive responses characterized by T helper (Th) 1 type of immune response; our laboratory has recently shown that during allergic airway infections, LPS-free fibrinogen cleavage product (FCP) triggers a Th2 type immune response in a TLR4-dependent manner. We hypothesize that TLR4 exhibit biased agonism in response to different ligands, such as LPS and FCP, and aim to determine the fibrinogen-derived TLR4 ligand that drives allergic responses and the distinct signaling mechanisms activated by FCP through TLR4. Column fractionation of fibrinogen hydrolysates, in addition with coomassie blue staining analysis and NFkB seap reporter assay, yielded two large size fractions that are potentially the TLR4 ligand; one of which had the molecular size similar to that of Ddimer, a known FCP that has been implicated in many diseases. Reverse phase protein array (RPPA) of LPS vs FCP treated RAW 264.7 cells demonstrated a distinctive signaling pathway induced by FCP in comparison to that triggered by LPS. Western blot analyses further showed distinctive kinetic differences between LPS and FCP in activating various NFkB subunits and phosphorylation of Mitogen-activated protein kinases (MAPKs) such as ERK 1/2 and p38. Moreover, as FCP can induce both mucin gene expression and airway eosinophilia, both of which are induced by IL-13 through STAT6, our western blot results based on RAW 264.7 cells and bone marrow derived macrophages (BMDMs) showed that STAT6 was clearly induced by FCP, but not LPS. These results indicate that LPS and FCP, both ligands for TLR4, activate NF-kB and MAPKs through distinct signaling pathways and that activation of STAT6 by FCP but not by LPS further support our hypothesis. Collectively, TLR4 appears to exhibit biased agonism for controlling immune responses elicit by different ligands. Contributors: Tung, Hui-Ying; Landers, Cameron; Yuping, Qian; Luz, Roberts; Corry, David. 328 2014 GRADUATE STUDENT SYMPOSIUM SCREENING FOR PHENOTYPIC PATTERNS ASSOCIATED WITH MITOCHONDRIAL MUTANTS TO UNRAVEL PATHWAYS BY WHICH MITOCHONDRIA CAUSE NEURONAL DEMISE Berrak Ugur Program in Developmental Biology Advisor: Hugo Bellen, Ph.D./D.V.M.-Department of Molecular & Human Genetics Mitochondria are highly dynamic organelles that are responsible for energy production, steroid biosynthesis, Ca+2 homeostasis, cell differentiation and cell cycle progression. Defects encompassing any of the mitochondrial metabolic pathways cause mitochondrial diseases and a breath of different symptoms in humans including myopathies and gastrointestinal dysfunction. The mitochondrial proteome contains approximately 1200 nuclear encoded genes and the vast majority of these genes remain uncharacterized. Hence, studying mitochondrial function in model organisms will provide important insights into the complex etiology and phenotypic heterogeneity of different disorders. We performed an unbiased, forward genetic screen for essential genes on the Drosophila X-chromosome that cause late developmental defects and neurodegeneration (X-screen). We isolated 165 genes, 100 of which were previously uncharacterized. Interestingly, 32/165 genes function in mitochondria and are highly conserved in humans. Mutations in 45% of these mitochondrial genes have already been reported to be causal in human disease. The remaining 55% are likely to be novel disease or lethality causing genes. We decided to perform a detailed phenotypic analysis of these 32 mutants to assess mitochondrial morphology, reactive oxygen species (ROS) levels, mitochondrial potential, lipid droplets (LD) in larval muscles, and morphology and function of homozygous mutant clones in the retina. All the Xmitochondrial mutants, except those that are cell lethal, cause neuronal defects in photoreceptors. We therefore rationalized that a detailed phenotypic analysis of the mutants will allow us to define specific pathways by which mitochondria cause neuronal demise. We isolated multiple phenotypic groups including two novel genes that encode proteins that may localize to mitochondria but have not yet been associated with them. For example, we observed that the loss of the fly homolog of human RFT1 (CG3149) causes excessive LD accumulation and increased ROS levels in the larval muscles. In parallel, 3 other X-mitochondrial mutants display increased ROS and muscle LD accumulation and hence define a phenotypic group. We are finishing the phenotypic analysis and grouping of X-mitochondrial mutants and we aim to investigate the molecular mechanisms that underlie these phenotypes. Contributors: Jaiswal, Manish; Sandoval, Hector; Haelterman, Nele A.; Yamamoto, Shinya; Bellen, Hugo J. 329 BAYLOR COLLEGE OF MEDICINE THE ROLE OF LOCAL ASTROCYTES IN ADULT-BORN NEURON CIRCUIT INTEGRATION Kevin Ung Program in Developmental Biology Advisor: Benjamin Arenkiel, Ph.D.-Department of Molecular & Human Genetics Adult neurogenesis in the mammalian brain represents an extraordinary example of continued cellular and structural neuronal plasticity. Although much has been learned about the general phenomenology associated with adult-born neuron development, the molecular and genetic mechanisms that guide adult-born neuron synapse formation, synapse maintenance, and circuit integration are not well understood. Previous studies show that multiple cellular interactions and diverse forms of presynaptic input affect the proliferation, survival, and synapse formation of adult-born neurons. However, the nature of how these presynaptic inputs and/or cellular interactions translate to the adultborn neuron circuit development remains unknown. We have recently combined Rabies Virus (RV)-mediated transsynaptic circuit tracing and mouse genetics to elucidate cell types that provide presynaptic inputs to adult-born neurons. Interestingly, in addition to diverse neuron-neuron interactions, we revealed numerous direct contacts between adult-born neurons and local resident astrocytes. We have also found that targeted activity manipulations of astrocytes directly influence proper adult-born neuron development and function. Astrocytes have been implicated in many aspects of neuronal development, ranging from trophic support to neuromodulation and plasticity. Moreover, defective astrocytes have been associated with several conditions, such as Alzheimer’s disease, schizophrenia, epilepsy, and tumorigenesis. Taking into account the known functional roles of astrocytes along with our preliminary data, we are currently testing the hypothesis that: specific local astrocyte populations play essential roles in adult-born neuron synapse formation and circuit integration in the olfactory bulb. Elucidating the molecular and cellular interactions between neurons and astrocytes that influence postnatal synaptogenesis and circuit integration will enhance our understanding of circuit formation in the developing, aging, and diseased brain. Uncovering novel functional roles for astrocytes toward adult-born neurons may not only better inform us of normal neural development and brain function, but also help explain how defects in astrocytes may contribute to neurodegenerative diseases. Contributors: 330 2014 GRADUATE STUDENT SYMPOSIUM EXPRESSION OF NR5A1 DISRUPTS FERTILITY AND PROMOTES ECTOPIC ENDOMETRIAL GROWTH Yasmin1 Margarita Vasquez Department of Molecular & Cellular Biology Advisor: Francesco Demayo, Ph.D.-Department of Molecular & Cellular Biology The nuclear receptor subfamily 5, group A, member 1 (Nr5a1), is a transcription factor expressed at high levels in the gonads where it promotes the expression of key steroidogenic enzymes. In the endometrium, the promoter of Nr5a1, is heavily methylated and thus the gene is silenced. However, in women with endometriosis, a chronic gynecological disease characterized by the growth of endometrial tissue outside the uterine cavity, this epigenetic regulation is lost and the gene is aberrantly expressed. The presence of endometriosis causes inflammation and severe pain, and it is associated with infertility lacking any other identified causes. We hypothesize that high expression of Nr5a1 in endometrial tissues results in the local production of estrogen and the disruption of hormone signaling. To test our hypothesis in vivo, we developed a mouse model in which Nr5a1 was conditionally expressed in the uterus. Nr5a1 expression resulted in the rapid development of cystic endometrial glands, observable by 6 weeks of age. Following a 6-month breeding trial, mice presented depleted stromal and myometrial compartments, a worsening of the cystic gland morphology, and were infertile. Endometriosis was induced by auto-transplantation of a 2 mm punch biopsy of uterine tissue sutured to the mesenteric membrane of cycling mice at 6 weeks of age. Ectopic lesions were excised four weeks later, revealing that those from Nr5a1 expressing mice were on average 4-fold larger than control lesions. Thus, Nr5a1 expression conferred distinct advantages to ectopic endometrial lesions. To determine which genes are misregulated in the endometrium in the presence of Nr5a1, microarray analysis was conducted, demonstrating 1992 genes to be differentially regulated. Pathway analysis identified these genes to be involved in development (Dcc, Klf9, Notch1 and Msx1), cellular movement and morphology (Shh, Cdkn1c, Gli2 and Wnt4) and vascular development and function (Hoxa10, Vegfa and Vcam1). Most notably, we identified a significant increase in the expression of genes involved in the activation of the immune response including the migration of lymphocytes and leukocytes, indicating that aberrant activation of Nr5a1 expression not only disrupt the hormone signaling pathways required for proper uterine morphology, function and fertility, but it also promotes endometriotic pathogenicity by inducing pathways responsible for a robust immune response. The present results indicate the Nr5a1 pathology in endometriosis may not be merely due to the production of estrogen as previously postulated, but also have a direct role in endometrial gland function and activation of inflammatory pathways. This research was supported by R01 HD042311 and U54 HD007495 to FJD. Contributors: Wu, San-Pin; Anderson, Matthew L.; Hawkins, Shannon M.; Creighton, Chad J.; Tsai, Sophia Y.; Tsai, Ming-Jer; Lydon, John P. and DeMayo, Francesco J. 331 BAYLOR COLLEGE OF MEDICINE G i2 IS REQUIRED FOR PARACRINE STIMULATION OF MAMMARY EPITHELIAL CELL PROLIFERATION BY ACTIVATED SMOOTHENED. Hugo Villanueva Department of Molecular & Cellular Biology Advisor: Michael Lewis, Ph.D.-Department of Molecular & Cellular Biology Background: Smoothened (SMO), the primary Hedgehog signaling effector, is overexpressed in breast cancers, and constitutive activation in mouse mammary glands leads to paracrine stimulation of proliferation. SMO can function canonically via GLI transcription factor activation, or non-canonically as a heterotrimeric G-protein coupled receptor (GPCR). Whether SMO functions non-canonically in mammals is not known. Our goal is to determine whether SMO signals as a GPCR in the mammary gland. Experimental Design and Methods: Differential gene expression analysis was performed using RNAseq, with validation by qRT-PCR in FACS-enriched SMOoverexpressing or non-expressing cells vs. wild type cells. G i function was assessed pharmacologically by in vivo treatment with pertussis toxin (PTX), and genetically using G i null alleles, in the presence or absence of activated SMO. GLI transcription factor function was assessed using a small molecule antagonist. Results: Gene expression analysis indicated that G i1 was the only G subunit upregulated in SMO-overexpressing mammary epithelial cells. Consistent with this observation, in vivo PTX treatment of SMO-induced mammary gland proliferation suggests G i function. Genetic deletion of G i2, and not G i1 or G i3, in the mammary gland in the presence of activated SMO suggests G i2 mediates SMOinduced proliferation. Gli expression was upregulated in SMO overexpressing cells, and treatment with a GLI antagonist is ongoing. Conclusion: Our data support a model of SMO function as a GPCR coupling via the PTX sensitive G i2 G-protein. Contributors: Villanueva, Hugo; Yu, Peng; Wu, Jessie; Plummer, Nicholas; Birnbaumer, Lutz; Hilsenbeck, Sue; Shaw, Chad; Lewis, Michael 332 2014 GRADUATE STUDENT SYMPOSIUM PUTATIVE STRUCTURES IN THE 3’ UNTRANSLATED REGION OF DENGUE VIRUS IMPROVE REPLICATION AND DISEASE Megan Brittany Vogt Integrative Program in Molecular and Biomedical Sciences Advisor: Rebecca Rico, M.P.H.-Department of Molecular Virology & Microbiology Dengue virus (DENV) is the most prevalent cause of viral hemorrhagic fever worldwide, with 500,000 cases of dengue hemorrhagic fever (DHF) and 50,000 DENV-related deaths occurring each year. Mechanisms of DENV pathogenesis are not well understood, primarily because this virus only causes disease in humans; however, DHF is often correlated with high blood DENV titers and infection with specific DENV strains. All DENV strains possess a single stranded positive sense RNA genome with a single 3’ untranslated region (3’UTR). In order to replicate the genome during viral replication, the host cell translation machinery must be utilized to translate the viral polymerase off of the viral genome. The genomes of DENV strains commonly associated with severe disease have distinctive putative RNA folding patterns, notably pseudoknots, in the 3’UTR that are thought to recruit the translation machinery and the viral polymerase to the viral RNA. Thus, we hypothesize that RNA folding patterns in the 3’UTR improve replication of DENV, ultimately increasing disease severity. To determine the impact of 3’UTR folding patterns on DENV genome replication, chimeric viruses will be created in which the 3’UTR of a more severe strain replaces the 3’UTR of a less severe strain and vice versa. Genome replication and production of infectious virus in vitro will be assessed upon infection of these chimeras into human dendritic cells, the only cell culture system that mimics natural infection. The impact of 3’UTR folding patterns on DENV genome translation will be assessed in vitro via luciferase assay with reporter constructs engineered with the 3’UTRs of specific DENV strains. If the 3’UTR structure does impact DENV genome replication and translation, then chimeras and reporter constructs containing the 3’UTR of more severe DENV strains should produce more infectious virus and have higher luciferase activity than those containing the 3’UTR of less severe DENV strains. The effects of these 3’UTRs on DENV pathogenesis will be further assessed in vivo using a humanized mouse model that develops dengue disease and mimics natural DENV infection in humans. Selected chimeras will then be evaluated for production of dengue disease in these humanized mice (NOD/SCID/IL-2r gamma null mice engrafted with human stem cells) by evaluating signs of disease, such as fever, viremia (virus titers in blood), thrombocytopenia (platelet loss), and rash. This study could improve our knowledge of DENV replication and help measure viral determinants of pathogenesis. Contributors: Vogt, Megan; Rico-Hesse, Rebecca 333 BAYLOR COLLEGE OF MEDICINE GLANDULAR MORPHOGENESIS IN THE MOUSE UTERUS Zer Vue Program in Developmental Biology Advisor: Richard Behringer, -Molecular Genetics - M.D. Anderson Endometrial glands secrete substances (or histotrophs) that are essential for endometrial receptivity to the embryo, implantation, conceptus survival, development and growth. During pregnancy, in response to increasing demands of the developing conceptus for histotrophs, endometrial glands undergo extensive hyperplasia and hypertrophy. In models where uterine glands have been phenotypically knocked out either through the use of genetics (mouse) or hormones (sheep), the females are infertile due to defects in implantation and early pregnancy loss, which suggests that uterine glands are essential for fertility. Uterine adenogenesis is the formation of glands within the stroma of the uterus of mammalian females. Like other tissues, uterine glands develop from fundamental processes such as tubulogenesis and branching morphogenesis. Uterine gland development is a unique postnatal process that occurs between Postnatal Day [P] 5 and P12 in the mouse. During P5, the luminal epithelium (LE) of the uterus will invaginate into and form epithelial buds. By P12, we have found that these initial buds extend into the adjacent stroma as single, essentially unbranched tubes. We are currently using various methods to examine later stages of adenogenesis. Uterine glands are considered to be highly coiled and branched, except for rodents. However, many of these conclusions were based on two-dimensional histological analysis of the uterus and limited data exists on the three-dimensional shape of a uterine gland. Contributors: Vue, Zer; Stewart, C. Allison; Gonzalez, Gabriel; Behringer, Richard 334 2014 GRADUATE STUDENT SYMPOSIUM RETHINKING THE APERTURE PROBLEM Edgar Yasuhiro Walker Department of Neuroscience/M.D.-Ph.D. Program Advisor: Andreas Tolias, Ph.D.-Department of Neuroscience How the visual system resolves ambiguity is a fundamental problem in vision science. A classic example is the aperture problem, in which a moving grating is viewed through an aperture. Although the stimulus is consistent with many motion directions, it typically produces a reliable motion direction percept. However, since very few studies have explored the effect of the shape of the aperture and of its orientation relative to the grating on perceived motion direction, the brain’s strategy for resolving ambiguity in this problem is not fully understood. We conducted an experiment in which subjects reported the perceived motion direction of a grating moving behind an elliptical or rectangular aperture with a variable aspect ratio and variable relative orientation. We found strong effects of relative orientation, aspect ratio, and shape on the perceived motion direction. These effects could not be captured by previous models – one based on a prior favoring low speeds, and one based on line terminators. Instead, we reframed the observer’s decision process as Bayesian inference on the motion direction of an infinitely long patterned strip with fixed but unknown width viewed through the aperture. In the model, the observer a) computes for each candidate motion direction the speed and minimum strip width consistent with the scene, b) assigns posterior probability using both the low-speed prior and a prior we propose here, which favors narrower strips, and c) reports the posterior mean. The resulting model not only outperformed the other two models, but also captured the observed dependencies with high accuracy. One potential interpretation of the narrow-strip prior is as a “little-unseen stuff” prior, favoring scenes that require the fewest assumptions about unobserved regions of the scene. Perhaps our brain resolves ambiguity by performing a process analogous to model selection, where simpler models are favored over complex ones. Contributors: Walker, Edgar; Ma, Wei Ji 335 BAYLOR COLLEGE OF MEDICINE DIFFERENT AFFINITIES OF ANKYRIN/SPECTRIN COMPLEXES IN THE CLUSTERING OF SODIUM AND POTASSIUM CHANNELS AT NODES OF RANVIER Chih-Chuan Wang Integrative Program in Molecular and Biomedical Sciences Advisor: Matthew Rasband, Ph.D.-Department of Neuroscience Myelination and the formation of nodes of Ranvier have been proposed to be an evolutionarily critical advance allowing saltatory and rapid propagation of action potentials in axons. The saltatory conduction of action potential depends on the nodal clustering of Na+ and K+ channels. It has been proposed that AnkyrinG is of critical importance in the clustering of Na+ and K+ channels. AnkyrinG interacts with NF-186, IV-spectrin, Na+ channels and K+ channels. NF-186 is the axonal receptor of glialderived extracellular matrix (ECM) and mediates ECM-based clustering of ion channels. IV-spectrin is the scaffold protein bridging nodal protein complex, including Na+ and K+ channels, to the actin cytoskeleton. However, we found that both AnkyrinG and IVspectrin are dispensable for nodal clustering of Na+ channels. In the AnkyrinG conditional knockout mice and IV-spectrin mutant mice, the nodal clustering of Na+ channels is still intact. We also found that the nodal clustering of Na+ channels is compensated by AnkyrinR and I-spectrin. Although it has been shown that Na+ and K+ channels share conserved domains that interact with Ankyrin, our results indicated that AnkyrinG and IV-spectrin are required for nodal clustering K+ channels. In contrast to Na+ channels, the nodal clustering of K+ channels cannot be compensated by AnkyrinR and I-spectrin in the AnkyrinG conditional knockout mice and IVspectrin mutant mice. In summary, we found that AnkyrinG and IV-spectrin are dispensable for nodal clustering of Na+ channels. Their function in nodal clustering of Na+ channels can be compensated by AnkyrinR and I-spectrin. However, the nodal clustering of K+ channels cannot be compensated by AnkyrinR and I-spectrin in the AnkyrinG conditional knockout mice and IV-spectrin mutant mice. There are different affinities of the AnkyrinR/ I-spectrin complex for Na+ and K+ channels. Contributors: Wang, Chih-Chuan; Ho, Tammy Szu-Yu; Rasband, Matthew N. 336 2014 GRADUATE STUDENT SYMPOSIUM MUTATIONS IN A METABOLIC KINASE GENE LEAD TO AUTOSOMAL DOMINANT RETINITIS PIGMENTOSA Feng Wang Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D.-Department of Molecular & Human Genetics Retinitis pigmentosa (RP) is a genetically heterogeneous disease with over 60 causative genes known to date. Nevertheless, approximately 40% of RP cases remain genetically unsolved, suggesting many novel disease-causing genes are yet to be identified. Here, we identified a glucose metabolism-related kinase gene as a novel causative gene for autosomal dominant RP (adRP). This is the first report that associates the glucose metabolic pathway with human retinal degenerative disease, suggesting a potential new disease mechanism. A large adRP family with negative result from known-retinal-disease-gene screening was recruited. Linkage analysis identified a minimal disease region of 8 Mb with a peak parametric LOD score of 3.500. Further whole-exome sequencing identified a heterozygous missense mutation that segregated with the disease phenotype in the family. Screening over 200 unsolved RP patients identified two unrelated simplex RP cases carrying an identical mutation which is only 11 amino acids away from the initial mutation identified in the adRP family, suggesting that both alleles may impact the same functional domain. Biochemical assays showed that the mutation does not affect kinase enzymatic activity or the protein stability, suggesting that the mutation may impact other uncharacterized function or result in a gain-of-function. Contributors: Feng Wang1,2*, Yandong Wang3*, Bin Zhang1,2*, Huajin Li4, Li Zhao1,5, Keqing Wang1,2, Mingchu Xu1,2, Yumei Li1,2, Frances Wu6, Cindy Wen6, Paul S. Bernstein7, Hui Wang1,2, Ruifang Sui4#, Kang Zhang6,8#, Rui Chen1,2,5,9,10# 337 BAYLOR COLLEGE OF MEDICINE COLLAPSED HAPLOTYPE PATTERN METHOD FOR LINKAGE ANALYSIS OF NEXT-GENERATION SEQUENCE DATA Gao T. Wang Program in Structural and Computational Biology and Molecular Biophysics Advisor: Suzanne Leal, Ph.D.-Department of Molecular & Human Genetics Traditionally, linkage analysis was used to map Mendelian diseases and genes within the linked regions were sequenced to identify the causal variants. Recent advances in next generation sequencing (NGS) make it possible to directly sequence genomes and exomes of individuals with Mendelian diseases and identify causal mutations by filtering variants in an affected individual(s) family member(s), removing those variants with higher allele frequency, e.g. >0.1% in variant databases. Linkage analysis of SNP data are sometimes used in conjunction with NGS to increase the success of identifying the causal variant. With the reduction in cost of NGS, DNA samples from entire families can be sequenced and linkage analysis can be performed directly using NGS data. Inspired by “burden” tests which are used for complex trait rare variant association studies, we developed the collapsed haplotype pattern (CHP) method to generate markers from sequence data for linkage analysis. To demonstrate the power of the CHP method compared to analyzing individual variants, we analyzed and performed empirical power calculations using the allelic architecture for several known non-syndromic hearing loss genes, i.e. GJB2, SLC26A4, MYO7A & MYH6. Power analysis demonstrated that the CHP method is substantially more powerful than analyzing individual SNVs in the presence of inter-allelic familial heterogeneity, i.e. families have different pathological variants within a gene or intra-familial heterogeneity e.g. compound heterozygotes. Specifically for an autosomal recessive model with allelic heterogeneity and locus heterogeneity of 50%, it requires 12 families for the CHP method to achieve a power of 90% for the SLC26A4 gene, while analyzing individual SNVs requires >50 families to achieve the same power at a genome-wide significance level of α=0.05. Unlike the commonly practiced filtering approaches used for NGS data, the CHP method provides statistical evidence of the involvement of a gene in Mendelian disease etiology. Additionally because it incorporates inheritance information and penetrance models it is less likely than filtering to exclude causal variants in the presents of phenocopies and/or reduced penetrance. We recommend the use of the CHP method in parallel to filtering methods to take full advantage of the power of NGS in families. The CHP method is incorporated in the SEQLinkage software which is freely available http://www.bioinformatics.org/seqlink/. Contributors: Gao Wang, Di Zhang, Biao Li, Hang Dai, Suzanne M. Leal 338 2014 GRADUATE STUDENT SYMPOSIUM IDENTIFICATION OF KINASES THAT CONTROL STABILITY AND LEVEL OF SHANK3 Li Wang Department of Molecular & Human Genetics Advisor: Huda Zoghbi, M.D.-Department of Pediatrics SHANK3 haploinsufficiency causes Phelan-McDermid Syndrome whereas SHANK3 duplications lead to manic-like behavior and epilepsy, suggesting a proper dosage of SHANK3 is critical for normal brain function. Currently, however, treatment approaches for normalizing SHANK3 abundance are lacking. Post-translational modifications like phosphorylation are common mechanisms for controlling protein level. We explored Shank3 phosphorylation pattern and identified 16 phosphorylated residues, suggesting a phospho-dependent regulation. We hypothesize that kinase pathways regulate the stability and abundance of Shank3, and that targeting these pathways could restore SHANK3 level and ameliorate corresponding Shankopathies. To identify such kinase pathways, I made a cell line to stably express a DsRedIRES-EGFP-Shank3 construct. The ratio of EGFP to DsRed serves as a read-out for Shank3 levels controlling for transgene transcription efficiency. Using this cell line, I performed a kinome-wide RNA interference (RNAi) screen with each of the 638 kinases targeted by 3 siRNAs independently. The RNAi hits with the greatest change in EGFP/DsRed ratio are considered positive. The screen revealed that multiple components of ERK1/2 pathway are involved in SHANK3 regulation. To validate this pathway, I treated mouse primary cortical neurons with two highly selective MEK1/2 inhibitors and detect a significant increase in Shank3 level. By q-RT-PCR, I found that this increase is not dependent on higher transcription of Shank3, suggesting translational or post-translational modifications. Considering that kinases in ERK1/2 pathway may directly phosphorylate SHANK3 and determine its stability, I performed a bimolecular fluorescence complementation (BiFC) assay to detect transient interactions between Shank3 and kinases in ERK1/2 pathway as well as other hits coming out of my screen. MEK1, Erk1 and Erk2 are all found to interact with Shank3. In parallel, immunoprecipitation followed by mass-spectrometry analysis identified ERK2 as one of the top interactors of Shank3. Currently I am performing an in vitro kinase assay to explore kinase-substrate interaction between SHANK3 and ERK. At the same time, I am generating shRNA lenti-viruses to knock down Erk1/2 pathway for further validation. In the future, I will validate other kinase hits for potential combination therapy to reduce side effects of strong inhibition of a single kinase. Contributors: Wang, Li; Han Kihoon; Holder Jimmy; Zoghbi, Huda 339 BAYLOR COLLEGE OF MEDICINE THE STOCHASTIC KINETICS OF E. coli TRANSCRIPTION AT A SINGLE GENE LOCUS Mengyu Wang Program in Structural and Computational Biology and Molecular Biophysics Advisor: Ido Golding, Ph.D.-Department of Biochemistry & Molecular Biology Gene regulation consists of a series of stochastic, single-molecule events, resulting in substantial randomness in mRNA production between individual cells, and even between the individual copies of the same gene within a single cell. To characterize the stochastic kinetics of transcription in E. coli, we combine fluorescently labelled DNA-binding proteins and single-molecule fluorescence in situ hybridization (smFISH), to simultaneously detect a gene of interest and measure its transcriptional activity, in individual bacteria. Our preliminary results, using the lactose promoter (Plac), indicate that we can reliably detect the site of active transcription. They also indicate that active transcription involves a change in the gene’s spatial position in the cell. Under conditions of strong repression, we find that transcription is more likely to occur close to the time of gene replication. Next, we will use mathematical modeling to map the observed nascent and total mRNA copy-number statistics to the underlying stochastic kinetics of transcription at a single gene locus. We will examine how this stochastic kinetics is modulated by transcription factors that regulate gene expression, and whether the activity of multiple copies of the same gene is correlated within the single cell. Contributors: Wang, Mengyu; Zhang, Jing; Sepulveda, Leonardo; Golding, Ido. 340 2014 GRADUATE STUDENT SYMPOSIUM CRYO-ET OF PLATELET Rui Wang Program in Structural and Computational Biology and Molecular Biophysics Advisor: Wah Chiu, Ph.D.-Department of Biochemistry & Molecular Biology In the United States, heart attack and stroke are two major killers, which are the products of arterial thrombosis and thromboembolism. Platelets, blood cellular fragments that are key components in hemostasis, play a critical role in the thrombosis pathophysiology. Platelets adhere to the exposed subendothelium of ruptured atherosclerotic plaques, aggregate and thereby forming a life-threatening occlusive thrombus that blocks blood flow and kills the surrounding cardiac or neural tissue. Platelet activation, which is obligatory to both hemostasis and thrombosis, catalyzes their granular contents, thereby propagating aggregation. Within this context, it is clear that a better understanding of the structural mechanisms underlying platelet activation will aid in the development of the appropriate interventions for thrombosis and bleeding disorders. However currently, the structural changes associated with platelet activation are poorly understood. Although conventional electron microscopy reveals structural details of platelet, it fails to uncover platelet structure in its native solution state. Cryo-electron tomography (CET) is a cellular imaging technique that offers some great advantages over other methods, particularly in the study of platelet structure. CET allows direct visualization of cellular structures at molecular resolution. Importantly, CET could deconvolute platelet structure in its native solution state without chemical embedding and fixation. CET and time-dependent rapid vitrification methods also permits kinetic analysis of activation according to changes in morphology. Unlike conventional methods that require chemical pretreatment that distort structure, CET reveals platelet morphology in a native physiological state. Contributors: Wang, Rui; Chiu, Wah; Michael Schmid; Dong, Jing-Fei; Wensheng Sun; Khant, Htet 341 BAYLOR COLLEGE OF MEDICINE SUPPRESSOR MUTATION DEFINES DIFFERENT FUNCTIONS OF TGRC1 IN DICTYOSTELIUM Yue Wang Program in Structural and Computational Biology and Molecular Biophysics Advisor: Gad Shaulsky, Ph.D.-Department of Molecular & Human Genetics Dictyostelium, commonly known as social amoeba, live as single cells with nutrition. After nutrition is consumed, thousand of cells aggregate to form multicellular structures and develop into fruiting bodies. In the final fruiting body, there are two types of cells: 80% of them are spores in the sorus, and the remaining 20% become vacuolized and make up the stalk. Only spores can germinate into cells again. TgrC1 is an adhesion molecule that has been demonstrated essential in both cell type differentiation and kin recognition. Knockout of tgrC1 results in a mutant defective in both differentiation and kin recognition. Protein Kinase A (PKA) regulates every stage of the development and its over-expression has been shown to overcome many developmental gene defects. However, the over-expression of PKA fails to rescue tgrC1-, which indicates that tgrC1 may represent a parallel pathway that is equally important as the PKA pathway for Dictyostelium development. How tgrC1 regulates differentiation and kin recognition therefore has been of great interests for scientists studying Dictyostelium and for those studying differentiation and kin recognition. We started to investigate the TgrC1 pathway by identifying the downstream players of TgrC1. Through an unsaturated suppressor screen we have discovered three genes that are likely to act downstream of TgrC1. They are stcA, stcB and stcC, stc short for suppressor of tgrC. All mutations partially rescue the tgrC1- developmental defects. We then investigated TgrC1’s role in differentiation and kin recognition using stcAins as a probe. Our study revealed TgrC1 has distinctive downstream pathways for cell type differentiation and for kin recognition. Furthermore, TgrC1 regulates stalk differentiation in a cell-autonomous way, whereas it regulates spore differentiation in a non-cell autonomous way. We propose that TgrC1 may function as a central regulator that integrates and responds to different signal inputs from cell adhesion, differentiation and kin recognition through the development of Dictyostelium. Contributors: Wang, Yue; Shaulsky, Gad 342 2014 GRADUATE STUDENT SYMPOSIUM HIGH-RESOLUTION EVOLUTIONARY-GUIDED DE NOVO STRUCTURE PREDICTION OF TRANSMEMBRANE HELICAL PROTEIN HOMOOLIGOMERS Yumeng Wang Program in Structural and Computational Biology and Molecular Biophysics Advisor: Patrick Barth, Ph.D.-Department of Pharmacology How specific protein associations regulate the function of membrane receptors remains poorly understood. Conformational flexibility currently hinders the structure determination of several classes of membrane receptors and associated oligomers. In absence of monomeric protein structures and experimental information on their binding sites, the prediction of protein-protein interactions and associated structures remains also very challenging. To address this problem, we have developed a general method to predict transmembrane (TM) helical protein homooligomeric structures from sequence guided by co-evolutionary constraints. We show that a combination of sequence-based prediction of TMH binding surfaces and stringent convergence criteria in folding simulations allow to considerably enrich sequence covariation signals in inter-monomer residue pairwise contacts. When applied to all 13 structurally characterized right and left handed homodimers and 3 higher-order oligomers, the method predicts near-native binding interface structures (Cα-RMSD ≤ 2.5 Å) and reach near-atomic accuracy (CαRMSD ≤ 1.5 Å) for 15 and 12 of these targets, respectively, using less than 3 contacts on average. The method also successfully predicts the structure of alternative oligomeric receptor conformational states and of combined TM and juxtamembrane (JM) receptor domains. Blind predictions of structurally-uncharacterized FGFR and KIT receptors provide testable hypothesis on the molecular mechanisms of diseaseassociated point TM and JM mutations and potential binding surfaces for the rational design of novel selective inhibitors. The method sets the stage for uncovering novel sequence/structure determinants of molecular recognition and mechanisms of signaling in single-spanning membrane receptor homo-oligomers. Contributors: Wang, Yumeng; Barth, Patrick 343 BAYLOR COLLEGE OF MEDICINE EXPRESSION AND FUNCTION OF CD133 IN PROSTATE EPITHELIA Xing Wei Integrative Program in Molecular and Biomedical Sciences Advisor: Li Xin, Ph.D.-Department of Molecular & Cellular Biology Prostate cancer (PCa) is one of the most common malignancies in the United States. Although most of the local tumors can be cured through surgery, chemo- and radio-therapy, a high percent of cases will eventually relapse as metastatic androgenrefractory PCa which is still lethal so far. The cell-of-origin of PCa has been indicated to be related with disease progression and potential resistance to androgen-deprivation therapy. As the tumorigenic mutation hits in cells with different differentiated statuses may cause diverse outcomes, the study on lineage hierarchy of prostate epithelia can shed light upon distinct properties of these cell subpopulations and specific therapeutic targets on the transformed cells. The long term goal of my project is to investigate the function of CD133 as a putative molecular marker for normal adult stem cells and/or cancer stem cells. Although CD133 together with some other cell surface molecules has been used to identify a subpopulation of prostate basal epithelia containing the capacity of self-renewal and differentiation, the function of this protein is still elusive in the prostate. In addition, the indeterminacy of CD133 antibody generates a lot of controversies in the previous stem cell marker studies. In my project, I will utilize better approaches to investigate the role of CD133 as a stem cell marker and to elucidate the function of CD133 in the prostate. My study shows that most of the CD133-expressing cells in murine prostate are luminal cells, but they can also be detected in the basal and stromal compartments. By performing lineage tracing with a CD133-CreER/R26-LSLYFP mouse model, I find that the percentage of CD133-expressing cells doesn't change after the induction of prostate epithelia turnover. And after castration-induced prostate involution, there is not an enrichment of CD133-expressing cells. These results indicate that prostate stem cells are not enriched in CD133-expressing cells. By using BrdU staining and transplantation-regeneration assay, I will determine the proliferation ability and in vivo stem cell capacity of CD133-expressing cells. As for the function study, CD133 knockout mice have been examined, and they have normal prostate pathology and in vitro sphere-forming ability. By overexpressing CD133 in human prostate cancer cell line PC-3, I find that the colony-forming and migration abilities are not affected, indicating that the function of CD133 is not essential. In the end, my thesis work will generate a definitive conclusion about the role of CD133 in prostate stem cell biology and prostate cancer cell biology, and will provide a solid basis for determining whether CD133 can be employed as a potential therapeutic target for PCa. Contributors: Wei, Xing; Xin, Li 344 2014 GRADUATE STUDENT SYMPOSIUM THE SPATIOTEMPORAL EXPRESSION PATTERN OF THE PROGESTERONE RECEPTOR DEFINES THE PERMISSIVE PERIOD OF EMBRYO IMPLANTATION WITHIN THE MURINE UTERUS Margeaux Wetendorf Integrative Program in Molecular and Biomedical Sciences Advisor: Francesco Demayo, Ph.D.-Department of Molecular & Cellular Biology In the nation today, millions of women suffer from the inability to reproduce. The female uterus is a hormone-responsive organ dependent on the presence of the ovarian steroid hormones, estrogen and progesterone, functioning via their receptors, the estrogen receptor and progesterone receptor (Pgr). Progesterone signaling is necessary for the initiation and continuance of a healthy pregnancy. Although comprised of two isoforms, the Pgr-A isoform is the most abundant within the murine uterus. During early murine pregnancy, the Pgr is expressed in the uterine epithelium on days 2-3 and decreases in expression by day 4, the time of embryo implantation. This decrease in epithelial Pgr expression is thought to initiate the beginning of what is known as the “window of receptivity” or the limited time frame in which the uterus is prepared to receive the embryo. We hypothesize that this unique Pgr expression pattern is critical for embryo implantation and if the Pgr is expressed in the epithelium through the window, implantation will fail to occur. To test this hypothesis, a conditional mouse model continually expressing the Pgr-A isoform was generated utilizing a Wnt7acre driven lox-STOP-lox system. Through continuous expression of Pgr-A within the uterine epithelium, the mice proved to be sterile due to defects in implantation and decidualization. The implantation defect was attributed to the decreased expression of leukemia inhibitory factor (LIF), a cytokine absolutely necessary for embryo implantation. Although not a direct target of the Pgr, Lif is thought to be regulated by a variety of transcription factors as the proximal promoter consists of Nfkb, Ets, and AP-1 binding sites. Furthermore, in vitro experiments using human endometrial adenocarcinoma cells expressing high levels of the Pgr-A isoform demonstrate that Lif is downregulated upon treatment with progestin. Our data suggest an indirect role of Pgr in the inhibition of Lif at the time of implantation. This mouse model and utilization of human endometrial cells will further our understanding of Pgr function in pregnancy and disease. This work was supported by NIH Grants: R01HD042311, 5U54HD007495 (to FJD) and R01CA77530 (to JPL) and NURSA grant: U19DK62434 (to MJT, SYT & FJD).] Contributors: Wu, San-Pin; Lydon, John; Tsai, Sophia; Tsai, Ming-Jer; DeMayo, Francesco 345 BAYLOR COLLEGE OF MEDICINE DISRUPTING SYNAPTIC TRANSMISSION IN THE CEREBELLUM CAUSES SEVERE DYSTONIA IN MICE Joshua James White Department of Neuroscience Advisor: Roy Sillitoe, Ph.D.-Department of Pathology & Immunology Dystonia is a severe neurological disorder that can cause painful muscle contractions. Although recent work has identified several brain regions that are altered in dystonia, a major obstacle to understanding why and how the brain sends erroneous signals to the muscles has been the lack of an accessible animal model. While genetically engineered mouse models have been useful for defining the molecular mechanisms of dystonia, none of these mice show dystonic motor behaviors. And although spontaneous mutant models do show dystonia, they also have unrelated pathological defects. Additionally, despite the severe dystonia that can be induced by certain chemicals, the methods of delivery often damage brain circuits and the dystonia that’s induced is usually transient and variable. To overcome these limitations, we developed a novel dystonia model by using a conditional genetic approach to remove vesicular glutamate transporter 2 (Vglut2) from olivo-cerebellar synapses, a connection that may be central to the defects in humans and animal models with dystonia. Here, we show using high-resolution anatomy and in vivo, awake electrophysiology that constitutive loss of olivo-cerebellar synaptic transmission alters the patterning of Purkinje cell dendrites, induces abnormal burst firing within the cerebellar circuit, and causes severe muscle co-contractions that produce dystonic movements. We also used a surgical approach that transiently inhibits the abnormal cerebellar output to overcome the dystonic phenotype. Our new model offers a unique opportunity to determine the circuit defects that trigger dystonia and an ideal approach for testing drug efficacy in a model that displays obvious dystonic movements and severe brain dysfunction. Contributors: White, Joshua J; Sillitoe, Roy V 346 2014 GRADUATE STUDENT SYMPOSIUM TARGETING THE TUMOR VASCULATURE WITH TEM8-SPECIFIC T CELLS LaTerrica Chemise Williams Program in Translational Biology & Molecular Medicine Advisor: Stephen Gottschalk, M.D.-Department of Pediatrics Xiao-Tong Song, Ph.D.-Department of Pathology & Immunology Background: T-cell immunotherapy with genetically modified T cells expressing chimeric antigen receptors (CARs) has shown promise in preclinical models as well as early clinical studies. However, patients with solid tumors often do not respond as well as patients with hematological malignancies. This lack of efficacy for solid tumors is most likely due to several factors including a) emergence of immune escape mutants, and b) inability of tumor-specific T cells to recognize and destroy the vascular bed of solid tumors, which is critical for their malignant growth. The aim of this project is to generate CARs specific for tumor endothelial marker (TEM)8, and evaluate their antivasculature and anti-tumor activity in preclinical tumor models. Methods/Results: We generated a retroviral encoding a TEM8-specific CAR consisting of the TEM8-specific single chain variable fragment AF344, a hinge/transmembrane domain, and a CD28.41BB.z endodomain. CD3/CD28-actiavted T cells were transduced with RD114-pseudotyped retroviral particles to generate TEM8specific T cells and CAR expression was confirmed by FACS analysis. To evaluate the functionality of TEM8-specific T cells we used TEM8-negative cell lines (U373, A549, LM7, 293T) and 293T cells that were genetically modified to either express human TEM8 (293T.hTEM8) or murine TEM8 (293T.mTEM8). TEM8-specific T cells recognized target cells in an antigen-dependent fashion as judged by their ability to secrete pro-inflammatory cytokines (IFN-g and IL-2) in coculture assays, and kill TEM8positive target cells. Importantly, TEM8-specific T cells readily recognized mTEM8positive target cells, which will allow us to evaluate the safety and efficacy of TEM8specific T cells in xenograft and immune competent murine tumor models. Conclusion: We have constructed a TEM8-specific CAR and have shown that T cells expressing this CAR recognize and kill hTEM8- or mTEM8-positive target cells. Animal studies are in progress to determine their safety and efficacy. Targeting the tumor vasculature with TEM8-specific T cells may improve current T-cell immunotherapies for solid tumors. Contributors: Williams, LaTerrica; Krebs, Simone; Karla, Mamta; Kakarla, Sunitha; Phung, Thuy; Rowley, David; Gottschalk, Stephen 347 BAYLOR COLLEGE OF MEDICINE SOCIALLY INDUCED RETRIEVAL OF FEAR MEMORY Chun-Ting Wu Department of Neuroscience Advisor: Daoyun Ji, Ph.D.-Department of Molecular & Cellular Biology Previously people have shown that rats that have fear memory shows increased freezing while observed conspecifics being shocked. My hypothesis is that during socially induced freezing, previous fear memory is recalled, and the hippocampal representations associated with the place of fear experience is reactivated. To test this, we set up a task in which the rats can observe a conspecific being shocked at the place where they had been shocked before. In the set-up, there are two boxes separated by a transparent glass. One rat (Observer) in non-shock box (NS) can observe the other rat (Demonstrator) in the shock box (S) being shock. These two boxes have distinct cues so that rats can differentiate two contexts. There were two groups of animals - Shocked Observer and Naïve Observer. Shocked Observer was shocked in S on Day1 but Naïve Observer was not. Both groups were in NS to observe Demonstrator being shocked in S on Day2, and the freezing levels of two groups were measured. To make sure that the freezing in Shocked Observer is not induced by the context of NS box, in another set of experiments (Control), rats are shocked in S on Day1 and then freezing level was tested on Day2 while rats are in NS but without Demonstrator in S. The preliminary result showed that Naïve Observer didn’t have increased freezing during observation. The freezing level of Shock Observer is low before the Demonstrator was shocked, but increased significantly after Demonstrator was shocked, indicating that freezing in Shock Observer is induced by the shock event of Demonstrator. For the Control rats, the freezing level is also low. In summary, I have demonstrated that rats had increased freezing while conspecifics were shocked in the environment where they had been shocked before, and this response is dependent on previous fear memory. To see the hippocampal representations during socially induced retrieval of fear memory, one rat with 9 place cells have been recorded simultaneously. In these cells, 5 cells have place field in S and 4 cells have place field in NS. While the rat was in NS, before Demonstrator was shocked, place cells encoding NS were activated. However, after shock to Demonstrator, place cells encoding S occasionally fire together in a short time period (~200 ms). This activity co-occurred with freezing behavior. The finding supports our hypothesis. I will confirm this by recording larger population of hippocampal neurons and finishing the quantitative analysis. Contributors: Wu, Chun-Ting; Ji, Daoyun 348 2014 GRADUATE STUDENT SYMPOSIUM THE DNA DAMAGE-CONTROL NETWORK: A NEW CLASS OF CANCER GENES DISCOVERED IN E.COLI Jun Xia Integrative Program in Molecular and Biomedical Sciences Advisor: Susan Rosenberg, Ph.D.-Department of Molecular & Human Genetics Two broad functional classes of cancer genes are the “gatekeepers”, and the “genomic caretakers”. The caretakers are DNA-repair and -damage-response genes, mutations in which increase mutation rate, which promotes cancer. The DNA damage that caretaker pathways ameliorate results mostly from endogenous cellular sources. We reasoned that the genes that affect endogenous DNA-damage levels are potentially cancer genes if either mutations in, or overexpression of these genes increase endogenous DNA damage to levels above repair capacity. This would create cells that act like caretaker (repair-defective) mutants, without having a caretaker mutation. We developed a high-throughput screen using E. coli that fluoresce upon DNA damage and screened an ordered E. coli overexpression library to model cancer-causing gene amplifications. We report a DNA-damage-control network of 214 damage-up and 24 damage-down genes that cause more or less DNA damage, respectively, when overexpressed. We validated all positives from our screen by sensitive flow-cytometric assay. We find that the vast majority are not traditional caretakers, indicating a new class of genes important to DNA damage. We identified 282 human homologues of the E.coli damage-control genes and found that these genes are significantly overrepresented in cancer-genome databases, mostly as amplifications, supporting the expectation that their overproduction promotes cancer. Only 5% of these human homologs are traditional caretaker genes. These data indicate that the human homologs are new class of cancer genes. We are characterizing the reasons for increased DNA damage in the E. coli model: (1) many of the DNA damage-control genes, when overexpressed, show sensitivity to DNA damaging agents, implying that excessive DNA damage overwhelms DNA repair pathways. (2) Fifteen of 15 selected high damage-up overexpressers show increased mutation rate, validating the proposal that excessive DNA damage leads to increased mutagenesis in E.coli. (3) We developed high-throughput microscopy to scan for overexpressers that have increased DNA double-strand breaks (DSBs) visualized as foci of an engineered fluorescent DSBbinding protein developed by our lab (eLife 2013). (4) We also showed increased stalled replication forks visualized as fluorescent foci of a new protein we have developed. (5) We also developed flow-cytometric assays to screen for overexpressers with chromosome-segregation defects and (6) increased oxygen radicals. Our data show the existence and functions of a new class of genes controlling genome stability: the DNA damage control genes, and show that in human, these are a new class of cancer gene. Contributors: Xia, Jun; Nehring, Ralf; Bravo, Maria; Guzman, Meztili; Mei, Qian; Frisch, Ryan; Gibson, Janet; Herman, Christophe; Miller, Kyle; Rosenberg, Susan; 349 BAYLOR COLLEGE OF MEDICINE MOLECULAR AND GENETIC STUDIES OF ATOH1 REGULATION Wei Xie Integrative Program in Molecular and Biomedical Sciences Advisor: Huda Zoghbi, M.D.-Department of Pediatrics Proper development of the cerebellum is highly dependent on the basic helix loop helix (bHLH) transcription factor Atoh1. Previous studies on Atoh1 have demonstrated its requirement for both proliferation and differentiation of cerebellar granule cell precursors (CGPs), which give rise to all granule cells of the mature cerebellum. Our lab’s previous studies have also implicated Atoh1 in the formation of Sonic hedgehogdriven medulloblastoma, the most common solid pediatric brain tumor. How Atoh1 promotes both cell proliferation and differentiation is not known. A better understanding of this mechanism not only expands our basic understanding of how a transcription factor can regulate seemingly opposing functions, but will also shed light on the mechanism of medulloblastoma formation. We hypothesized that phosphorylation and/or different interactors of Atoh1 can regulate its function, ultimately affecting CGP proliferation and differentiation status. Based on evolutionary conservation, serine 193 (S193) emerged as a potential phosphorylation site for regulation of Atoh1 function. In vitro studies in our lab demonstrated that altering S193 phosphorylation affects Atoh1 transcriptional activity. Further studies showed that the DNA binding properties of the S193A phosphomutants were altered. In sum, our results pointed to a pivotal role of S193 phosphorylation on Atoh1 function. Therefore, we generated a phosphomutant knock in mouse in order to investigate the role of S193 phosphorylation in vivo. Concurrently, we performed an in vivo immunoprecipitation and mass spectrometry screen to identify novel interactors of Atoh1. Several proteins from the screen have been further validated with two assays: bimolecular fluorescence and in vitro immunoprecipitation. We are currently pursuing knockdown studies to identify interactors that affect Atoh1 function. Future studies will focus on characterizing the Atoh1 S193 phosphomutant knock in mice and investigating the functional interaction between Atoh1 and its interactors. Ultimately, our studies will shed light on the underlying mechanisms in which a single transcription factor can drive seemingly opposing functions in a specific cell population. Contributors: Xie, Wei; Klisch, Tiemo; Zoghbi, Huda 350 2014 GRADUATE STUDENT SYMPOSIUM IN SEARCH OF NOVEL DISEASE-CAUSING GENES OF FAMILIAL EXUDATIVE VITREORETINOPATHY (FEVR), A GENETICALLY HETEROGENEOUS EYE DISEASE Mingchu Xu Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D.-Department of Molecular & Human Genetics Familial exudative vitreoretinopathy (FEVR) is a rare genetic disease that cause visual impairment and retinal detachment due to abnormal retinal vascularization To date, five disease-causing genes, including NDP, FZD4, LRP5, TSPAN12 and ZNF408, have been associated with FEVR and together account for ~50% of FEVR cases. We collected a cohort of about 100 FEVR patients and a two-step strategy was used to unravel the molecular etiology. First, targeted panel NGS sequencing was performed to screen mutations in all known FEVR-causing and other retinal disease genes. Patients who are negative during the first step are subject to whole exome sequencing (WES) as they are likely caused by mutations in novel disease genes. By capture panel sequencing, we have solved ~40% cases. We also identified many novel variants in known FEVR-causing genes, especially in LRP5 (15 missense variants). For the unsolved cases, we have enriched some candidate genes both in large families and in sporadic cases. These genes have functional implications in tissue vascularization based on mouse phenotypes. Their mutations also recurred in multiple cases, which make them good candidates. They are now undergoing genetic validation and probably some of them will be tested functionally. Contributors: Xu, Mingchu; Salvo, Jason; Wang, Hui; Wang, Keqing; Li, Yumei; Nguyen, Duy; Luo, Hongrong; Zhang, Kang; Chen, Rui 351 BAYLOR COLLEGE OF MEDICINE CLONAL EVOLUTION OF THE HER2 L755S MUTATION LEADS TO ACQUIRED HER-TARGETED THERAPY RESISTANCE THAT CAN BE REVERSED BY THE IRREVERSIBLE HER1/2 INHIBITOR AFATINIB Xiaowei Xu Department of Biochemistry & Molecular Biology Advisor: Rachel Schiff, Ph.D.-Department of Medicine Background: Targeting HER2 with lapatinib (L), trastuzumab (T), or the LT combination, is effective in HER2+ breast cancer (BC), but acquired resistance commonly occurs. In our 12week neoadjuvant trial (TBCRC006) of LT without chemotherapy in HER2+ BC, the overall pathologic complete response (pCR) rate was 27%. To investigate resistance mechanisms our lab developed 10 HER2+ BC cell lines resistant (R) to these drugs (LR/TR/LTR). To discover potential predictive markers/therapeutic targets to circumvent resistance, we completed genomic profiling of the cell line panel and a subset of pre-treatment specimens from TBCRC006. Methods: Parental (P) lines and LR/TR/LTR derivatives of 10 cell line models were profiled with whole exome/RNA sequencing. Mutations detected in R lines but not in P lines of the same model were identified. Single cells of the BT474AZ-LR line were cloned and sequenced. Mutation-specific Q-PCR was designed to sensitively quantify mutations. Whole exome sequencing (>100X) and Ion AmpliSeq of 17 TBCRC006 baseline tumor/normal pairs were performed. Results: We found and validated the HER2 L755S mutation in the BT474ATCC-LTR/BT474AZ-LR lines (~30% of DNA/RNA/cDNA in BT474AZ-LR), in which the HER pathway was reactivated for resistance. Overexpression of this mutation was previously shown to induce LR in HER2-negative BC cell lines, suggesting a role as an acquired L/LT resistance driver in HER2+ BC. Sequencing of BT474AZ-LR single cell clones found the HER2 L755S mutation in ~30% of HER2 copies in every clone. Using mutation-specific Q-PCR, we found statistically higher HER2 L755S levels in BT474ATCC-P/ BT474AZ-P compared to other HER2+ BC parentals. These data suggest that this mutation exists subclonally within BT474 parentals and was selected as the more dominant population in the two resistant lines. The HER1/2 irreversible tyrosine kinase inhibitor (TKI) afatinib (Afa) robustly inhibited growth of BT474ATCC-LTR/AZ-LR cells (IC50: Afa 0.02µM vs. L 3 µM). Western blots confirmed HER and downstream Akt and MAPK signaling inhibition in the LR cells by Afa. Whole exome sequencing/Ion Ampliseq of TBCRC006 baseline found the HER2 L755S mutation in 1/17 subjects. This patient did not achieve pCR. The variant was present in 2% of the reads, indicating a subclonal event. Conclusion: Acquired resistance in BT474 LR/LTR lines is due to selection of HER2 L755S subclones present in parental cells. The higher HER2 L755S levels in BT474 parentals compared with other parentals, and detection of its subclonal presence in a pre-treatment HER2+ BC patient, suggest that sensitive mutation detection methods will be needed to identify patients with potentially actionable HER family mutations in primary tumor. Treating this patient group with an irreversible TKI like Afa may prevent resistance and improve clinical outcome of this subset of HER2+ BC. Contributors: Xu, Xiaowei; Nardone, Agostina; Qin, Lanfang; Hu, Huizhong; Nanda, Sarmistha; Heiser, Laura; Wang, Nicholas; Covington, Kyle; Chen, Edward; Renwick, Alexander; Wang, Tao; De Angelis, Carmine, Contreras, Alejandro; Gutierrez, Carolina; Fuqua, Suzanne; Chamness, Gary; Shaw, Chad; Wheeler, David; Gray, Joe; Hilsenbeck, Susan; Rimawi, Mothaffar; Osborne, C. Kent; Schiff, Rachel. 352 2014 GRADUATE STUDENT SYMPOSIUM HUMAN EFFECTOR-MEMORY T CELLS ARE RESISTANT TO HYPOXIA Yang Xu Department of Pathology & Immunology Advisor: Gianpietro Dotti, M.D.-Department of Medicine The clinical efficacy of chimeric antigen receptor (CAR)-redirected T cells remains marginal in solid tumors compared to leukemia. Several immunosuppressive mechanisms may be responsible for this lack of efficacy including the low oxygen tension or hypoxia that is a common feature of the solid tumor microenvironment. In particular, hypoxia is known to severely impair proliferation and viability of human peripheral blood T cells (PBT cells). However, it remains unknown if hypoxia uniformly and equally suppresses all T-cell subsets, and specifically whether it affects CAR-T cells generated for clinical use. We thus generated T-cell lines by expanding ex vivo T lymphocytes from PBT after activation with CD3/CD28 cross-linking antibodies in the presence of IL-2 (TEXP cells), and tested them in 1% O2 tension. In sharp contrast with freshly isolated PBT cells, TEXP cells featured enhanced cell divisions in hypoxia vs normoxia (proliferation index: 1.8±0.2 vs 1.3±0.1, respectively; p<0.01). In addition, viability of activated TEXP cells was significantly higher in hypoxia (71±1%) than normoxia (48±6%)(p<0.01). Cytotoxic function of TEXP cells redirected with a GD2-specific 2nd generation CAR was also enhanced in hypoxia as residual neuroblastoma cells were reduced in hypoxia (9±5%) than in normoxia (227%) (p<0.05) in co-culture assays. Using in vivo hypoxia labeling in NSG mice, we found higher percentages of CAR-redirected TEXP cells actively cycling within hypoxic tumor areas (KI67+ T cells: 68±21%), than in normoxic tumor areas (KI67+ T cells: 35±11%), suggesting that CAR- redirected TEXP cells are functionally superior in hypoxic environment. As the great majority of TEXP cells consist of effector-memory (EM) cells, we dissected whether their enhanced function in hypoxia reflected an endogenous property of EM cells or was rather acquired through ex vivo cultures. We found that EM cells directly isolated from PBT showed comparable properties to TEXP cells in hypoxia (proliferation index: 0.36±0.13 in normoxia vs 0.94±0.05 in hypoxia; p<0.05; viability 42±6% in normoxia vs 61±7% in hypoxia; p<0.05), while freshly isolated naive and central-memory cells were highly suppressed. Resistance to hypoxia of TEXP cells was associated with an increased expression of hypoxia ¬adaptation genes and glycolytic activities. HIF¬1α and downstream glycolytic genes are highly expressed in EM and TEXP cells. Consequently, glucose uptake and lactate production were also significantly elevated in these cells compared to PBT ±. Our findings thus suggest a “pre¬-adaptation status” of EM and TEXP cells to hypoxia that can be further exploited to enhance the functionality of ex-vivo generated tumor-specific T cells. Contributors: Xu, Yang; Savoldo, Barbara; Metelitsa, Leonid; Dotti, Gianpietro 353 BAYLOR COLLEGE OF MEDICINE BACK-TO-BACK MECHANISMS DRIVE ACTOMYOSIN RING CONTRACTION DURING DROSOPHILA CELLULARIZATION Zenghui Xue Department of Biochemistry & Molecular Biology Advisor: Anna Sokac, Ph.D.-Department of Biochemistry & Molecular Biology While Myosin-2 motor activity is required for actomyosin ring contraction during cytokinesis in some cell types, recent findings suggest that motor activity is dispensable in other cell types, including mammalian cultured cells and budding yeast. One simple possibility is that different cell types use different mechanisms of actomyosin contraction during cytokinesis. Another possibility is that Myosin-2 dependent and independent mechanisms act in the same cell type, but make different contributions to contraction. In this work, we address the contribution of Myosin-2 motor activity during the cytokinetic event of cellularization in early Drosophila embryos. During cellularization, actomyosin rings contract to build the bottom of newly forming epithelial cells. By quantitative live cell imaging, we find that ring contraction proceeds in two morphologically and kinetically distinct phases. In Phase 1, hexagonal rings become circular, and the contraction rate is slow (0.17 ± 0.04 μm/min, mean ± S.E.M.). In Phase 2, rings get smaller, and ring contraction is fast (0.66 ± 0.03 μm/min, mean ± S.E.M.). F-actin and Myosin-2 levels increase or stay constant, respectively, during Phase 1, but both decrease during Phase 2, suggesting that the Phases are mechanistically distinct. Indeed, using genetic mutants and drug strategies to manipulate Myosin-2 motor activity and F-actin polymerization dynamics, we find that contraction during Phase 1 depends on Myosin-2 motor activity, while Phase 2 does not. Instead, Phase 2 contraction depends on F-actin depolymerization. Taken together, our work shows that two back-to-back mechanisms drive distinct phases of actomyosin ring contraction during cellularization. Our data supports a model whereby Myosin-2 dependent and independent mechanisms conspire to drive ring contraction, within the same cell type, and even during the same cytokinetic event. Cellularization now provides a unique opportunity to compare distinct contraction mechanisms in the same cell type, and to understand what molecular components control the switch between Myosin-2 dependent and independent mechanisms. Contributors: Xue, Zenghui; Sokac, Anna 354 2014 GRADUATE STUDENT SYMPOSIUM IS PERTURBED PULSE WAVE VELOCITY IN SYSTEMIC ARTERIES PRESENT IN PATIENTS WITH PULMONARY ARTERIAL HYPERTENSION? Eric Yota Yang Clinical Scientist Training Program Advisor: Christie Ballantyne, M.D.-Department of Medicine BACKGROUND: Increased systemic arterial stiffness has been demonstrated in and is thought to contribute to heart failure with preserved ejection fraction (HFpEF). Pulmonary arterial hypertension [PAH] results from vascular dysfunction through multiple pathways, some of which may be shared with the systemic circulation. In concert with this a similar female preponderance has been observed in patients with HFpEF and in those with PAH. We hypothesized that a shared perturbation in both pulmonary and systemic arterial stiffness as evaluated by carotid-femoral pulse wave velocity [PWV] may be present in PAH patients. METHODS: PWV and derived central pressures were measured with applanation tonometry in sequential consenting hemodynamically confirmed PAH patients (N=25). Exclusion criteria included hypertension, diabetes, scleroderma, liver disease, renal disease, congenital heart disease. RESULTS: No patient was treatment naïve though 2 were on only calcium channel blocker therapy. 15 patients had idiopathic PAH; 4, heritable PAH; 7 PAH, associated with connective tissue disease; and 1, anorexiant associated PAH. WHO functional classes 2 and 3 were present in 17 and 10 patients respectively. The group was middle-aged (median 43 [interquartile range (IQR) 34, 54] years) and mostly female (89%) with brachial systolic and diastolic blood pressures (BP) of 105 (IQR 100, 115) and 65 (IQR 60, 70) mm Hg, respectively. These BP were correlated (p<0.01) with central systolic (95 [IQR 91, 102] mm Hg, tau-b = 0.60) and diastolic BP (67 [IQR 61, 72] mm Hg, tau-b = 0.93). The median PWV was 6.7 m/s (IQR 5.9, 7.7). No difference (p=0.75) in PWV was noted between PAH patients and controls reported in literature. CONCLUSIONS: Systemic arterial stiffness appears to be normal in patients with pulmonary arterial hypertension although the impact of PAH specific therapy cannot be excluded. Further evaluations of systemic arterial stiffness should be considered in treatment naïve PAH patients. Contributors: Yang, Eric; Nambi, Vijay; Frost, Adaani 355 BAYLOR COLLEGE OF MEDICINE DNMT3A LOSS DRIVES AML AND ALL IN FLT3-ITD LEUKEMOGENESIS Liubin Yang Department of Molecular & Human Genetics/M.D.-Ph.D. Program Advisor: Margaret Goodell, Ph.D.-Department of Pediatrics Epigenetic modifiers including the de novo DNA methyltransferase (DNMT) 3A are amongst the most frequently occurring recurrent mutations in hematologic malignancies, yet the mechanism by which DNMT3Amut contributes to leukemic transformation remains unknown. Interestingly, 41% of these mutations coexist with the FLT3-ITD oncogene. Here, we report that Dnmt3a haploinsufficiency cooperates with FLT3-ITD to generate acute myeloid leukemia and Dnmt3a loss generates FLT3-ITD early immature T-cell-like leukemia, arising from transformed HSCs, by conditional knockout murine models. We found that Dnmt3a loss results in upregulation of stem-cell and myeloid gene signatures likely causing hypomethylation of critical distal regulatory regions in hematopoietic stem and progenitor cells. Our results demonstrate that Dnmt3a loss of function releases otherwise suppressed stem-cell and myeloid genes to participate in leukemogenesis. More broadly, our findings support a role for Dnmt3a as a guardian of distal regulatory regions important in the epigenetic control of leukemic transformation. Contributors: Yang, Liubin; Rodriguez, Benjamin; Mayle, Allison; Luo, Min; Jeong, Mira, Curry, Choladda; Ruau, David; Zhang, Xiaotian; Challen, Grant; Rau, Rachel; Gottgens, Berthold; Li, Wei; Goodell, Margaret 356 2014 GRADUATE STUDENT SYMPOSIUM FUNCTIONAL CONNECTIVITY IN POPULATIONS OF CORTICAL NEURONS DURING SENSORY PROCESSING Dimitri Yatsenko Department of Neuroscience Advisor: Andreas Tolias, Ph.D.-Department of Neuroscience Ambitious projects aim to record the activity of ever larger and denser neuronal populations in vivo. Correlations in neural activity measured in such recordings can reveal important aspects of neural circuit organization. However, estimating and interpreting large correlation matrices is statistically challenging. Estimation can be improved by regularization, \ie by imposing a structure on the estimate. The amount of improvement depends on how closely the assumed structure represents dependencies in the data. Therefore, the selection of the most efficient correlation matrix estimator for a given neural circuit must be determined empirically. Importantly, the identity and structure of the most efficient estimator informs about the types of dominant dependencies governing the system. We sought statistically efficient estimators of neural correlation matrices in recordings from large, dense groups of cortical neurons. Using fast 3D random-access laser scanning microscopy of calcium signals, we recorded the activity of nearly every neuron in volumes 200 µm wide and 100 µm deep (150–350 cells) in mouse visual cortex. We hypothesized that in these densely sampled recordings, the correlation matrix should be best modeled as the combination of a sparse graph of pairwise partial correlations representing interactions between the observed neuronal pairs and a low-rank component representing common fluctuations and external inputs. Indeed, in cross-validation tests, the covariance matrix estimator with this structure consistently outperformed other regularized estimators. The sparse component of the estimate defined a graph of interactions. These interactions reflected the physical distances and orientation tuning properties of cells: The density of positive ‘excitatory’ interactions decreased rapidly with geometric distances and with differences in orientation preference whereas negative ‘inhibitory’ interactions were less selective. Because of its superior performance, this ‘sparse+latent’ estimator likely provides a more physiologically relevant representation of the functional connectivity in densely sampled recordings than the sample correlation matrix. Contributors: Yatsenko, Dimitri; Josic, Kresimir; Ecker, Alexander; Froudarakis, Emmanouil; Cotton, Ronald; Tolias, Andreas. 357 BAYLOR COLLEGE OF MEDICINE THE ROLE OF NALCN IN CARDIORESPIRATORY CONTROL Szu-Ying Yeh Program in Developmental Biology Advisor: Huda Zoghbi, M.D.-Department of Pediatrics Cellular and circuit excitability of the nervous system is essential for important rhythmic behaviors, including breathing and heartbeats. Respiration and cardiac cycle continue to maintain physiological homeostasis throughout life, and cardiorespiratory coupling ensures precise and efficient gas exchange in organisms through highly overlapping respiratory and cardiac networks within the brainstem. Prematurity and several brain disorders are associated with deterioration of breathing and cardiac rhythms such as apneas of prematurity (AoP), Rett syndrome, sudden infant death syndrome (SIDS), and sudden unexpected death in epilepsy (SUDEP). Dissecting the mechanism underlying breathing control is of central importance to understand the basis of cardiorespiratory neural circuits and life-threatening cardiorespiratory disorders. I focus on a recently identified ion channel NALCN, which is highly expressed in respiratory rhythm generators, the preBötzinger complex and the retrotrapezoid nucleus/parafacial respiratory group. NALCN is critical for maintaining respiratory activity and sustaining life. Also, NALCN current contributes to about 72% of the basal Na+ leak current, which is the characteristic of all pacemaker cells that generate autonomous neuronal activity, and has been purposed to underlie neuromodulation of neuronal bursting rhythm. Using genetic mouse models, we will delete Nalcn gene in cells within the hindbrain to determine the NALCN-dependent neuronal population that is essential for neonatal survival. On the other hand, we purpose that the NALCN current govern the respiratory rhythm generation and/or modulation. Combining genetic mouse models and electrophysiological approaches, we will assess the biophysical properties of rhythm generators within the respiratory center. We expect that loss of NALCN current in respiratory rhythm generators would lead to perturbations, or even cessations in spontaneous oscillation and responses to neuromodulators, and ultimately lethality that will illuminate the functional roles of NALCN. We will also exploit this model to study whether the rhythm generators themselves are sufficient to drive motor behavior and whether the circuit excitability dictates respiratory activity. Revealing the functions of NALCN in our study will advance our understanding of the mechanism in respiratory rhythm and pattern generation. Contributors: Yeh, Szu-Ying; Huang, Wei-Hsiang; Zoghbi, Huda 358 2014 GRADUATE STUDENT SYMPOSIUM CELLULAR AND MOLECULAR MECHANISM IN MÜLLERIAN DUCT FORMATION Shuo-Ting Yen Program in Developmental Biology Advisor: Richard Behringer, -Molecular Genetics - M.D. Anderson Tubulogenesis is a fundamental process in development. Several mechanisms are already well-studied in many tubular systems, such as neural tube, kidney, as the trachea system in fly. However, the tube formation of reproductive tract development still remains unclear. The female reproductive tract primordium, Müllerian duct, forms in an intriguing manner. Its formation is now defined as three phase: initiation, invagination, and elongation. Several models of Müllerian duct formation have been proposed but none of them agree with each other. New Methodologies should be applied to elucidate the cellular mechanism of Müllerian duct formation. By time-lapse imaging of Wnt7a-Cre; R26R-YFP mouse embryo, we have observed digit like cell process and changes in diameter of the tube. These findings may provide new clues to explain Müllerian duct formation. Furthermore, Müllerian duct formation is unique in its dependence on Wolffian duct. Most tube formation does not depend on another existing tube. It is known that some molecules, such as WNT9B produced by the Wolffian duct, play a tropic role in Müllerian duct formation (Carroll et al., 2005; Grünwald, 1937). However, the detailed mechanisms of this Wolffian-duct dependent guidance are still needed to be elucidated. Contributors: Yen, Shuo-Ting; Huang, Cheng-Chiu; Behringer, Richard 359 BAYLOR COLLEGE OF MEDICINE TRANSGENE EXPRESSION IN THE NEUROPSIN TET-OFF DRIVER LINE IS NOT RESTRICTED TO THE ENTORHINAL CORTEX Michael Joseph Yetman Department of Neuroscience Advisor: Joanna Jankowsky, Ph.D.-Department of Neuroscience The entorhinal cortex (EC) plays a central role in episodic learning and memory formation, and is among the earliest sites of neuronal loss and neurofibrillary tangle formation in Alzheimer’s disease. The EC has therefore been an attractive target for genetic manipulation to selectively modify gene expression or neuronal function in various models of neurological disease. Many such conditional models utilize the neuropsin (Nop) promoter to limit spatial distribution of the tetracycline transactivator (TTA). When crossed with a second tet-responive transgenic line, the resulting bigenic mice will express the transgene of interest in neurons where TTA is active. The NopTTA mouse line was reported to restrict tet-responsive transgenes to the superficial layers of medial EC and parts of the pre- and parasubiculum (Yasuda and Mayford, Neuron, 2006), and Nop-TTA mice have been used in several experimental studies examining functional properties of the entorhinal-hippocampal circuitry. The utility of this transgenic driver line is contingent on the specificity of the spatially restricted gene expression, yet detailed neuroanatomical mapping of its expression has not yet been done. We therefore crossed the Nop-TTA driver line with a reporter strain expressing βgalactosidase and green fluorescent protein, and established an online histological atlas of Nop-TTA regulated gene expression. This atlas resource (available through the Rodent Brain Workbench, www.rbwb.org) was used to perform a detailed brainwide analysis of β-galactosidase labeling in bigenic (Nop-TTA-LacZ) mice. Our findings highlight strong expression in regions beyond the EC and suggest caution in interpreting experiments that depend on precise localization of gene products controlled by the NopTTA driver. Contributors: Yetman, Michael; Lillehaug, Sveinung, Bjaalie, Jan, Leergaard, Trygve, Jankowsky, Joanna 360 2014 GRADUATE STUDENT SYMPOSIUM BEHAVIORAL PHENOTYPING OF MICE DEFICIENT IN CHRNA7 Jiani Yin Department of Molecular & Human Genetics Advisor: Huda Zoghbi, M.D.-Department of Pediatrics 15q13.3 microdeletion syndrome is a rare genetic disorder caused by a deletion of a segment of chromosome 15. The deletion is commonly 1.5Mb in length and encompasses 6 genes, but patients with small deletions, which only encompass the CHRNA7 gene, have also been reported. The clinical phenotypes associated with this syndrome are variable, but commonly include developmental delay/intellectual disability and impaired social interaction. Other clinically important features include epilepsy, impulsive behavior, aggression, and schizophrenia. A mouse model deficient of CHRNA7 was generated and reported to be grossly normal in growth, anxiety-like behaviors, learning and memory, as well as sensorimotor gating. However, deficits in social interaction and repetitive behavior have not been assessed in this mouse model. We tested heterozygous and homozygous CHRNA7 mutant mice and their wildtype littermates for repetitive behaviors in self-grooming, holeboard exploration, and marble burying test, and for social interaction behaviors in the three-chamber paradigm, partition test, and social interaction video scoring. A detailed assessment of the aforementioned behaviors will be presented and be discussed in the context of human 15q13.3 microdeletion phenotypes. Contributors: Yin, Jiani; Schaaf, Christian 361 BAYLOR COLLEGE OF MEDICINE NOVEL ROLES OF AN AXON-GUIDANCE MOLECULE SEMAPHORIN-3E IN PANCREATIC CANCER Lin-Kin Yong Program in Translational Biology & Molecular Medicine Advisor: Qizhi Yao, M.D./Ph.D.-Department of Surgery William Fisher, M.D.-Department of Surgery Pancreatic cancer (PC) is the 4th leading cause of cancer death in the US with a 5-year survival rate of less than 6%. One characteristic feature of PC is that it has dense stroma which makes PC cells especially difficult to respond to treatment and often develop resistance to chemotherapeutic drugs. Hence, there is an urgent need for better understanding of PC stroma. Two recent large-scale genomic analyses of human PC have uncovered increased copy numbers of an axon-guidance gene SEMA3E that codes for the glycoprotein Semaphorin-3E (Sema3E). Several recent reports have implicated Sema3E in metastasis of breast and colon cancers. Sema3E has also been reported to have functions in inflammation and immune cell migration. Hence, we hypothesize that Sema3E contributes to PC progression by promoting metastasis of PC, as well as modulating immunosuppression in PC stroma. Using qRT-PCR of matched patient tumor and adjacent normal tissue, we found that Sema3E was significantly upregulated (>2-fold) in 46% of samples, while the receptor of Sema3E, PlexinD1, was significantly upregulated in 33% of samples. We also performed immuno-histochemical (IHC) staining for Sema3E and PlexinD1 expression in patient and mouse PC (KrasG12D/-;P53R172H/-;PDX-1-Cre PC mouse model) tissue samples, and found that, for both human and mouse tissues, Sema3E and PlexinD1 were expressed at much higher levels in the epithelial tumor cells compared to normal acinar cells, highlighting the significance of the roles Sema3E and PlexinD1 play in PC. We are currently in the process of generating and characterizing Sema3E-overexpression and –knockdown PC cell lines to further understand the precise effects Sema3E has on PC. Given reports of Sema3E’s role in mediating inflammation in atherosclerosis, we hypothesize that Sema3E can modulate immunosuppression in PC stroma by skewing macrophage polarization towards the protumoral M2 subtype. Mouse bone marrow-derived macrophages (BMDM) were first polarized to an M1 or M2 subtype via addition of LPS or IL4+IL-13, respectively, followed by addition of recombinant Sema3E to the cells, after which the cells were harvested for analysis. qRT-PCR and WB analysis showed that the levels of iNOS, a M1 macrophage marker, were suppressed upon adding Sema3E to M1-polarized cells. Flow cytometry analysis revealed that addition of Sema3E to BMDM enhanced the production of a M2 macrophage-related cytokine, IL-10. These preliminary results suggest that Sema3E can skew polarization of macrophages towards the M2 subtype, which may point towards an immunosuppressive role of Sema3E in the tumor stroma. Altogether, we have shown that Sema3E is overexpressed in majority of pancreatic cancer, and plays a role in modulating immunosuppressive stroma by skewing macrophage polarization towards the protumoral subtype. These findings indicate that Sema3E could be an attractive novel therapeutic target by acting on both PC tumor cells and tumor stroma. Contributors: Yong, Lin-Kin; Li, Dali; Liang, Zhengdong; Fisher, William; Chen, Changyi Johnny; Yao, Qizhi Cathy 362 2014 GRADUATE STUDENT SYMPOSIUM FLOW CYTOMETRIC ISOLATION OF MOUSE EMBRYONIC MACROPHAGES FOR MOLECULAR AND FUNCTIONAL STUDIES Nejla Yosef Department of Molecular Physiology & Biophysics Advisor: Mary Dickinson, Ph.D.-Department of Molecular Physiology & Biophysics In mouse development, the first macrophages appear in the yolk sac at embryonic day 7.5 (E7.5), and between E8.0 and E9.5 they can be found in both extraembryonic and embryonic tissues [1]. Several studies suggested that embryonic macrophages (EMs) play various roles during the development and remodeling of the vascular system [2], but the mechanisms behind this diversity are not fully understood. Relatively little is known about the phenotypic and functional capabilities of embryonic macrophages (EMs) at the molecular level. Here, we report the establishment of a highly efficient protocol to isolate EMs using the Csf1r-EGFP+/tg mouse reporter line expressed within EMs. E9.5 Csf1r-EGFP+/tg transgenic embryos were enzymatically digested to generate a single cell suspension and EMs were sorted via fluorescence activated cell sorting (FACS) using GFP signal. Further marker analysis showed that it is possible to sort brightly >98% purified EMs. Such a pure EM population is suitable for a wide range of molecular and functional studies to define specialized properties of EMs that are different from other macrophage classes Contributors: 363 BAYLOR COLLEGE OF MEDICINE CARBON BLACK-INDUCED LUNG INFLAMMATION IN EMPHYSEMA AND LUNG CANCER Ran You Department of Pathology & Immunology Advisor: Farrah Kheradmand, M.D.-Department of Medicine Environmental pollutants and cigarette smoke are major causative factors for lung diseases, such as COPD (chronic obstructive pulmonary diseases) and lung cancer, which are respectively the 3rd leading cause of death and the 1st cancerrelated death, however the shared pathogenic substance by all risk factors for these diseases is still unknown. In our study, carbon black (CB), as the component of coal mine dust, cigarette smoke and airborne particles, is found to deposit in the lung antigen-presenting cells (APCs) of emphysema/COPD patients. Therefore, we hypothesized that CB-induced activation of APCs and lung inflammation promote progression of emphysema and lung cancer. By using mouse model with the intranasal challenge of CB, we found that CBchallenged mice developed similar immunopathological changes in emphysema patients and mice exposed to chronic smoke, including enlarged lung volume, infiltration of immune cells into the lungs, upregulated disease-related gene and strong Th17 responses. Moreover, CB directly activate APCs by upregulation of pro-Th17 cytokines, IL-6 and IL-1β. Furthermore, by comparing the soluble nanoparticle polyethylene glycol (PEG)-CB (hydrophilic) and elemental CB (hydrophobic), we found that hydrophobic CB induced more severe inflammation and emphysema, indicating that surface features of CB contribute to its pathogenesis. More interestingly, CB-induced inflammation also decreases lung tumor latency. In airway specific Pten/Smad4 deficient mice, CB promotes early lung cancer progression concomitant with early stomach metastasis. Further on, CB directly increases Pten-deficient bronchoepithelial cell invasion. To figure out the underlying mechanism of how hydrophobic CB induces emphysema and promotes cancer progression, we did RPPA analysis of CB-treated cells and found that CB directly induced DNA double strand break and Erk signaling, which can stimulate genomic instability, inflammation and cell invasion. APCs treated with the inhibitors to either protein kinases sensing DNA damage or MAPK kinase were less sensitive to CB treatment indicated by less IL-6 production. In all, CB, as the shared pathogenic substance in environmental pollutants and cigarette smoke, induces severe inflammation and promotes emphysema and early lung cancer progression by direct induction of DNA damage. Potential mechanisms of CBinduced APC activation revealed in our study provide new insights into the immunopathogenesis of lung diseases caused by environmental risk factors. Contributors: You, Ran; Shan, Ming; Lu, Wen; Cho, Sungnam; Seryshev, Alexander; Marcano, Daniela, Song, Lizhen; Yuan, Xiaoyi; Demayo, Francesco; Tour, James; Corry, David; Kheradmand, Farrah 364 2014 GRADUATE STUDENT SYMPOSIUM COMPUTATIONAL MOEDELING AND DESIGN OF ORTHOGONAL GPCRMEDIATED SIGNALING COMPLEXES Melvin Chen Young Department of Biochemistry & Molecular Biology Advisor: Patrick Barth, Ph.D.-Department of Pharmacology G protein-coupled receptors (GPCRs) commonly exhibit an inherently high level of promiscuity in ligand and effector binding and activation that hinders accurate understanding of their regulation and specific role in diseases. Consequently, many cell and gene therapy strategies are limited by the inability to reprogram receptor signaling properties without affecting endogenous cellular signaling pathways. We hypothesize that orthogonal, highly specific, GPCR-mediated signaling complexes will allow for the properties of specific receptor and downstream effector systems to be finely tuned without perturbing the function of related receptors and alternative pathways. A combination of Rosetta’s multi-state design (MSD) and docking protocols have been used to evolve and select in-silico novel and highly specific binding interfaces unrelated to native GPCR/G protein complexes. The MSD protocol does not allow for a flexible backbone, limiting the available sequence space. To compensate for this limitation, we use the docking protocol to produce a larger diversity of GPCR/G protein binding interfaces that allows access to a substantially larger sequence space than MSD alone. The most promising computationally designed orthogonal GPCR/G protein complex has been experimentally cross validated by monitoring agonist-induced specific activation of design and WT complexes using cell-based assays. Our activation assay shows that the change in membrane potential with the designed GPCR and designed G protein pair compares favorably with the WT/WT pair, while both design/WT pairs have drastically lower activation as intended by the design process. This suggests that our methodology should greatly aid in the creation of highly specific GPCR-mediated signaling complexes. Contributors: Young, Melvin Chen; Chen, Kuang-Yui Michael; Barth, Patrick 365 BAYLOR COLLEGE OF MEDICINE A PERTURBED TRANSCRIPTOME UNDERLIES CORNELIA DE LANGE SYNDROME AND RELATED PHENOTYPES Bo Yuan Integrative Program in Molecular and Biomedical Sciences Advisor: James Lupski, M.D./Ph.D.-Department of Molecular & Human Genetics Cornelia de Lange syndrome (CdLS) is a genetically heterogeneous disorder manifesting extensive phenotypic variability. To date, mutations in NIPBL, SMC1A, SMC3, RAD21 and HDAC8, which encode subunits or regulators of the cohesin complex, are found in about 65% of patients. Wiedemann-Steiner syndrome (WDSTS), caused by mutations in KMT2A, shares phenotypic features with CdLS. We utilized genomic approaches to determine additional molecular etiologies for CdLS-like phenotypes and investigate molecular underpinnings of shared clinical features. Whole exome sequencing (WES) of two male siblings clinically diagnosed with WDSTS revealed a hemizygous, predicted-deleterious, missense mutation in SMC1A. Extensive clinical evaluation and WES of a Turkish cohort of 30 patients clinically diagnosed with CdLS revealed a de novo heterozygous nonsense KMT2A mutation in one patient without characteristic WDSTS features. Moreover, a de novo heterozygous frameshift mutation in SMC3 was identified in a patient with combined CdLS and WDSTS features. Furthermore, in families from two separate world populations segregating an autosomal recessive disorder with CdLS-like features, we identified homozygous mutations in TAF6, which encodes a core component in a transcriptional regulation pathway. Our findings suggest CdLS and related phenotypes may result from a “transcriptomopathy” rather than a cohesinopathy: a conclusion supported by recent transcriptomic studies. Contributors: Yuan, Bo; Pehlivan, Davut; Karaca, Ender; Patel, Nisha; Gambin, Tomasz; Gonzaga-Jauregui, Claudia; Sutton, V. Reid; Yesil, Gozde; Bozdogan, Sevcan; Tos, Tulay; Beck, Christine R.; Gu, Shen; Aslan, Huseyin; Yuregir, Ozge Ozalp; Rubeaan, Khalid; Nakeeb, Dhekra; Alshammari, Muneera; Bayram, Yavuz; Atik, Mehmed M.; Aydin, Hatip; Geckinli, Bilge; Seven, Mehmet; Ulucan, Hakan; Fenercioglu, Elif; Jhangiani, Shalini; Muzny, Donna M.; Boerwinkle, Eric; Baylor-Hopkins Center for Mendelian Genomics; Tuysuz, Beyhan; Alkuraya, Fowzan S; Gibbs, Richard A.; Lupski, James R. 366 2014 GRADUATE STUDENT SYMPOSIUM COMPLEMENT C1Q-BASED THERAPY ATTANUATES SMOKE-INDUCED EMPHYSEMA Xiaoyi Yuan Department of Pathology & Immunology Advisor: Farrah Kheradmand, M.D.-Department of Medicine Smoking-induced chronic obstructive pulmonary disease (COPD), which encompasses chronic bronchitis and emphysema, is a progressive inflammatory lung disease with no known effective treatment. Sterile inflammation induced by cigarette smoke can activate lung antigen presenting cells (APCs) expressing CD11b/CD11c markers that can differentiate T helper type 1 (Th1) and Th17 cells. Maturing APCs are associated with expression of complement components because a large number of their proteins and receptors have been detected in mice and humans. In particular whereas most other complement factors are synthesized in the liver, the first component of the classical complement pathway, C1q, is primarily produced by the APCs and macrophages. Interestingly deficiency of C1q in humans and mice is associated with a lack of immunoregulatory response that results in inflammation and autoimmunity characteristic of systemic lupus erythematosus (SLE). Thus we hypothesized that C1q serves a regulatory role in the inflammation and autoimmunity observed in emphysema. We show here that C1q mRNA level is reduced in lung APCs from emphysema patients and mice exposed to cigarette smoke, and smokers with emphysema showed lower plasma C1q concentration, which correlates with decreased lung function. Moreover, C1q augments the differentiation of T regulatory cells in vitro and in vivo, with the increase of IL-10 production. Furthermore, APCs treated with C1q also exhibit inhibitory phenotype marked by elevated CD103 expression and the ability to induce more robust T regulatory cells differentiation in co-culture assays. Finally, relative to vehicle control, complement C1q intranasally treated mice showed attenuated smokeinduced emphysema with reduced lung pathology measured by Micro-CT, less inflammatory cells infiltration to the airway and antigen presenting cell (APCs) infiltration to the lung, as well as reduction in inflammatory cytokine production. These findings suggest a critical role for C1q in the inhibition of smoke-induced lung inflammation, and should be further explored to develop specific new therapeutic targets for the treatment of emphysema. Contributors: Yuan, Xiaoyi; Kheradmand, Farrah 367 BAYLOR COLLEGE OF MEDICINE THE PAN-CANCER ANALYSIS OF PSEUDOGENE EXPRESSION REVEALS BIOLOGICALLY AND CLINICALLY RELEVANT TUMOR SUBTYPES Yuan Yuan Program in Structural and Computational Biology and Molecular Biophysics Advisor: Han Liang, Ph.D.-Bio informatics and Computational Biology Although individual pseudogenes have been implicated in tumor biology, the biomedical significance and clinical relevance of pseudogene expression have not been assessed in a systematic way. Here we generate pseudogene expression profiles in 2,808 patient samples of seven cancer types from The Cancer Genome Atlas RNA-seq data using a newly developed computational pipeline. Supervised analysis reveals a significant number of pseudogenes differentially expressed among established tumor subtypes; and pseudogene expression alone can accurately classify the major histological subtypes of endometrial cancer. Across cancer types, the tumor subtypes revealed by pseudogene expression show extensive and strong concordance with the subtypes defined by other molecular data. Strikingly, in kidney cancer, the pseudogeneexpression subtypes not only significantly correlate with patient survival, but also help stratify patients in combination with clinical variables. Our study highlights the potential of pseudogene expression analysis as a new paradigm for investigating cancer mechanisms and discovering prognostic biomarkers. Contributors: Han, Leng; Yuan, Yuan; Zheng, Siyuan; Yang, Yang; Li, Jun; Edgerton, Mary; Diao, Lixia; Xu, Yanxun; Verhaak, Roeland; Liang, Han 368 2014 GRADUATE STUDENT SYMPOSIUM MODULATION OF FXTAS NEURODEGENERATION IN A MOUSE FMR1 PREMUTATION MODEL BY CUGBP1 Zachary A Zalewski Department of Molecular & Human Genetics Advisor: David Nelson, Ph.D.-Department of Molecular & Human Genetics FXTAS is hypothesized to arise by an RNA-mediated toxic gain-of-function mechanism, by which the rCGG sequesters RNA binding proteins (RBPs), inhibiting their normal function. Molecular hallmarks of FXTAS include ubiquitin-positive intranuclear inclusion bodies throughout the brain and marked dropout of cerebellar Purkinje neurons. We have previously developed transgenic mouse models to drive premutation-length rCGGs specifically in Purkinje neurons. Purkinje neuron-specific promoter L7/Pcp-2 drives the expression of 90 CGG repeats fused to the cDNA of Fmr1 or EGFP. These mice have been shown to successfully model human phenotypes including inclusion formation (in Purkinje neurons), Purkinje neuron loss, and impaired motor coordination. CUG-Binding Protein Elav-Like Family Member 1 (CELF1, aka CUGBP1) is an RBP that has previously been demonstrated in a Drosophila model to be a potent modifier of the rCGG premutation. We predict that, in the mouse, CELF1 protein expression is inversely proportional to phenotypic severity – the less CELF1 is being expressed, the more severe the phenotype and vice versa. We have developed a novel CELF1 knockout mouse model and have utilized existing models to overexpress CELF1 in Purkinje neurons specifically using a Tet-off system, and are assessing their effect on phenotype in the transgenic premutation models by immunohistochemistry, immunofluorescence, and behavioral studies. Thus far, we have demonstrated that haploinsufficiency of CELF1 in the context of the Fmr1 premutation may enhance rCGG toxicity. In littermates aged six to eight months CELF1 depletion in the context of CGG-Fmr1 significantly (p<0.05) increases the percentage of Purkinje neurons containing an inclusion body, relative to CGG-Fmr1 expressing normal levels of CELF1. We are also in the process of gathering iterative behavioral data – primarily rotarod but also additional motor coordination assays – for mice between the ages of 4-12 months. Our current findings support the significance of CUGBP1 and other RBPs in mammalian models of FXTAS, as well as the RNA toxic gain-of-function hypothesis. This serves to underscore the long-term implications of RBP inhibition/sequestration by premutation-length rCGG, whose presence in intranuclear inclusions is coincident with neuronal loss, and highlight the potential future role of RBPs as possible therapeutic targets. Contributors: Zalewski, Zachary A.; Jin, Peng; Timchenko, Lubov T.; Nelson, David L. 369 BAYLOR COLLEGE OF MEDICINE DETERMINING GENES CONTRIBUTING TO MISSING HERITABILITY IN HUMAN DISEASE THROUGH LARGE COHORT ANALYSIS Jacques Eric Timothy Zaneveld Department of Molecular & Human Genetics Advisor: Rui Chen, Ph.D.-Department of Molecular & Human Genetics Despite complete sequencing of exonic regions in disease genes, there remains significant missing heritability for human Mendelian disease. Here, we calculate the contribution of known disease genes to this missing heritability by examining the frequency of lone recessive mutations in undiagnosed disease vs control populations that have undergone exon sequencing. This novel statistical analysis was applied to our massive cohort of >600 retinal disease patients whose disease could not be explained by exonic mutations. Interestingly, certain known retinal disease genes, including ABCA4 and USH2A, contribute significantly to missing heritability in retinal disease. Other known retinal disease genes such as MYO7A show no contribution. This analysis can be applied to any sufficiently large cohort and any disease at least partially caused by recessive mutations. The results of this analysis allow resources to be efficiently directed towards the genes with the largest contribution to missing heritability. Much of the missing heritability associated with genes identified through this approach is likely due to mutations that are not observed in exon sequencing data (such as intronic mutations and copy number variations). To search for these types of mutations, we performed in depth analysis of the retinal disease genes we predicted to have the strongest contribution to missing heritability. Through aCGH, intron sequencing and bioinformatics we identified 13 novel mutations that were recurrent in our disease cohort but absent from controls. Further, we identified 20 intronic mutations predicted to cause splice site changes either leading to cryptic exon inclusion or exon loss. Ongoing preliminary molecular tests confirm altered splicing caused by some of these candidates. This data shows that directed investigation of genes contributing to missing heritability allows for the rapid discovery of novel types of disease causing mutations. More importantly, this technique improves the efficiency of molecular diagnosis and research into the cause of human disease. Contributors: Zaneveld , Jacques*; Gelowani, Violet*; Salvo, Jason; Soens, Zachary; Wang, Feng; Li, Huajin; Wang, Hui; Wang, Keqing; Li, Hui; Ren, Huanan; Lopez, Irma; Dorfman, Allison; Khan, Ayesha; Li, Yumei; Sui, Ruifang; Koenekoop, Robert and Chen, Rui. *These authors contributed equally to this manuscript. 370 2014 GRADUATE STUDENT SYMPOSIUM THE FUNCTION OF MICRORNA MIR-23A CLUSTER IN OSTEOGENESIS Huan-Chang Zeng Program in Developmental Biology Advisor: Brendan Lee, M.D./Ph.D.-Department of Molecular & Human Genetics MicroRNAs are negative regulators of target genes and are involved in various biological processes. The miR-23a cluster is a group of microRNA which is highly enriched in bone tissues but the function of this cluster during osteogenesis is not yet clear. To study the function of miR-23a cluster in bone development in vivo, we generated a transgenic mouse model overexpressing miR-23a cluster using the osteoblast (Ob)-specific Col1a1-2.3kb promoter. Micro-computed tomography (uCT) and histomorphometric analysis showed that transgenic mice exhibited low bone mass with decreased Ob number and decreased mineral apposition rate. Interestingly, we also found that osteocytes were significantly increased in transgenic mice supporting dysregulation of both early Ob fate commitment and late Ob terminal differentiation into osteocytes. To study the loss of function of miR-23a cluster in vivo, we also generated transgenic mice expressing decoys for miR-23a, miR-27a or miR-24-2 individually driven by the Col1a1-2.3kb promoter. Mice with the miR-23a decoy and miR-27a decoy showed dental fractures and low bone mass by μCT while miR-24 decoy lines had no apparent abnormalities. RNASeq analysis of mouse models showed that TGF-β signaling pathway may be directly regulated by the miR-23a cluster. Contributors: Zeng, Huan-Chang; Bae, Yangjin; Dawson, Brian; Chen, Yuqing; Bertin, Terry; Munivez, Elda; Chen, Rui; Lee, Brendan 371 BAYLOR COLLEGE OF MEDICINE INTEGRATIVE GENOMICS ANALYSIS OF HUMAN INFLUENZA VIRUS INFECTION Yijie Zhai Department of Molecular & Human Genetics Advisor: John Belmont, M.D./Ph.D.-Department of Molecular & Human Genetics Gene expression profiling of human blood cells might uncover the complex dynamics of host response to ARIs such as pandemic H1N1. However, only limited data are available on the system level response to ARIs in naturally acquired infections. To understand the molecular bases and network orchestration of host responses, we prospectively enrolled 1600 healthy adults in fall 2009-2010, followed the subjects with influenza-like illness (N=133) for 3 weeks, and examined changes in their peripheral blood gene expression. The results, which were replicated between seasons, showed a dramatic upregulation of interferon pathway and innate immunity genes. This persisted for 2-4 days. The data show a recovery phase at days 4 and 6 with differentially expressed transcripts implicated in cell proliferation and repair. By day 21 the gene expression pattern was indistinguishable from baseline (enrollment). We identified two biomarkers that differentiate influenza from non-influenza virus infection. Using lineage and activation state specific transcripts to produce cell composition scores, patterns of B and T lymphocytes depression accompanied by a major activation of dendritic cells and NK cells were found in the acute phase of illness. We then moved the focus from gene expression patterns to gene co-expression patterns. We detected gene modules that are related to core features of regulatory networks and found a substantial increase in the connectivity of the influenza responsive genes. Finally, we identified a molecular signature that strongly correlated with antibody response to pH1N1 virus. Taken together, our findings offer insights into the molecular mechanisms underlying host response to influenza virus infection, and provide a valuable foundation for investigation of the global coordinated responses to acute infection. Molecular correlates of the immune response suggest targets for intervention and improved vaccines. Contributors: Zhai, Yijie; Belmont, John; Atmar, Robert; Quarles, John; Arden, Nancy; Bucasas, Kristine; Wells, Janet; Niño, Diane; Wang, Xueqing; Zapata, Gladys; Shaw, Chad; Franco, Luis; Couch, Robert 372 2014 GRADUATE STUDENT SYMPOSIUM QUANTIFYING TRANSCRIPTION KINETICS IN INDIVIDUAL E. COLI CELLS Jing Zhang Department of Biochemistry & Molecular Biology Advisor: Ido Golding, Ph.D.-Department of Biochemistry & Molecular Biology Gene expression and its regulation are traditionally studied by averaging over large cell populations. These “bulk” measurements miss significant cell-to-cell differences, as well as all spatial information within the single cell. To overcome these limitations, we plan to quantify transcription kinetics at the level of individual gene copies in individual E. coli cells. For that purpose, the gene of interest will be labeled using the Fluorescent Repressor Operator System (FROS), which combines fluorescently-tagged DNA binding proteins with a synthetic array of cognate binding sites. The transcriptional activity of the gene will be tracked using an analogous method, where the MS2 RNAbinding protein labels an array of binding sites on nascent mRNA. We have constructed these two reporter systems separately, and are now optimizing single-cell imaging and data acquisition in a microfluidic device. Next, we will combine both reporters to simultaneously follow the individual copies of the gene, and their transcriptional activity, in individual cells. We will examine whether transcription is coupled to cellular events such as gene replication or cell division, and whether transcription is accompanied by changes in the spatial position of the gene. We will also ask whether the transcriptional activity of two gene copies in the same cell is correlated. In the future, we will extend the work to examine the dynamics and correlation of two different genes in the same cell. Contributors: Zhang, Jing; Wang, Mengyu; Golding, Ido. 373 BAYLOR COLLEGE OF MEDICINE MUTUAL POTENTIATION OF DNMT3A LOSS-OF-FUNCTION AND IDH2 NEOMORPHIC MUTATIONS IN MALIGNANT HEMATOPOIESIS Xiaotian Zhang Department of Molecular & Human Genetics Advisor: Margaret Goodell, Ph.D.-Department of Pediatrics Recent studies of impaired hematopoiesis in acute myeloid leukemia (AML), have implicated frequent somatic mutations in genes that participate in the epigenetic regulation of transcription1-4. In contrast to so-called Class I or Class II mutations in classical models of leukemogensis, alterations in epigenetic modifiers such as DNMT3A and IDH1/2 do not appear sufficient by themselves to induce myeloid malignancies5-7. Interestingly, mutations in two DNA methylation regulators, DNMT3A and IDH1/2, occur together nonrandomly in 5~8% of patients with AML and myelodysplastic syndrome (MDS)3,4,8-10, suggesting that the two genetic lesions may interact to promote the induction of myeloid malignancies. Here we demonstrate that overexpression of a neomorphic mutant Idh2 in a Dnmt3a loss-of-function background in hematopoietic stem and progenitor cells (HSPCs) can potentiate the development of several different myeloid malignancies. Our transcriptome analysis identified a distinct stem cell-like gene expression signature in Dnmt3a—/—; Idh2R140Q HSPCs in that was absent in hematopoietic progenitors with a single-mutant background. This signature is activated by the loss of DNA methylation in hematopoiesis-associated enhancer regions due to the loss of Dnmt3a and the gain of trimethylation on histone H3 Lysine 4, induced by high 2-hydroxylglutarate (2-HG) levels caused by over expression of Idh2R140Q. Metabolome analysis shows 2-HG in Dnmt3a—/—; Idh2R140Q cells is synthesized mainly from glucose by active oxidative phosphorylation. Importantly, treatment with a histone deactylase (HDAC) inhibitor rapidly reversed the abnormal epigenetic changes in Dnmt3a—/—- Idh2 mutant HSPCs disrupting leukemia self-renewal. Our research shows, for the first time, that mutations in two DNA methylation regulators, Dnmt3a and Idh2, can interact to potentiate the development of myeloid malignancy Contributors: Xiaotian Zhang1,2,3, Benjamin Rodriguez4*, Putluri Nagireddy5*, Ting Zhou6,7*, Mira Jeong2,3*, Min Luo2,3,Hongcang Gu8, Liubin Yang1,2,3, George Michailidis9, Alexander Meissner8, Wei Li4, Arun Sreekumar5, Vivienne I. Rebel6,7, and Margaret A. Goodell1,2,3 374 2014 GRADUATE STUDENT SYMPOSIUM NEXT-GENERATION SEQUENCING BASED MOLECULAR DIAGNOSIS OF 82 RETINITIS PIGMENTOSA PROBANDS FROM NORTHERN IRELAND Li Zhao Program in Structural and Computational Biology and Molecular Biophysics Advisor: Rui Chen, Ph.D.-Department of Molecular & Human Genetics Retinitis pigmentosa (RP) is a group of inherited retinal disorders characterized by progressive photoreceptor degeneration with a prevalence of approximately 1:3,500 to 1:4,000. RP is highly heterogeneous, as nearly 3100 mutations in more than 50 genes have been reported cause the disease. Next-Generation Sequencing (NGS)based molecular information can contribute to precise clinical diagnoses enabling better disease management. A retinal capture panel that enriches 186 known retinal disease genes, including 55 known RP genes, was developed. Targeted Next-Generation Sequencing was performed for a cohort of 82 unrelated RP cases from Northern Ireland, including 46 simplex cases and 36 familial cases. Disease-causing mutations were identified in 49 probands, including 28 simplex cases and 21 familial cases, achieving a solving rate of 60%. In total, 65 pathogenic mutations were found, and 29 of these were novel. Interestingly, the molecular information of 12 probands was not consistent with their initial inheritance pattern nor clinical diagnosis. Further clinical reassessment resulted in a refinement of the clinical diagnosis in 11 patients. The benefit of molecular diagnoses is particularly highlighted by two patients and their families. One patient with a clinical re-diagnosis to Refsum disease from RP can now be treated with the appropriate diet which has been shown to reduce the progression of Refsum disease. In another case, the reclassification of the inheritance pattern from xlRP to arRP saved the family from performing taxing offspring selection. This is the first study to apply Next-Generation Sequencing based, comprehensive molecular diagnoses to a large number of RP probands from Northern Ireland. Our study shows that molecular information can aid clinical diagnosis, potentially changing treatment options, current family counseling and management. Contributors: Zhao, Li; Wang, Feng; Wang, Hui; Li, Yumei; Alexander, Sharon; Wang, Keqing; Willoughby, Colin; Zaneveld, Jacques; Jiang, Lichun; Soens, Zachry; Earle, Philip; Simpson, David; Silvestri, Giuliana; Chen, Rui 375 BAYLOR COLLEGE OF MEDICINE DEVELOPMENT BIODEGRADABLE POLYPEPTIDES FOR IN VIVO NON-VIRAL GENE DELIVERY Mingkun Zhao Integrative Program in Molecular and Biomedical Sciences Advisor: Jin Wang, Ph.D.-Department of Pharmacology Lung cancer remains the leading cause of cancer related mortality in the US and is estimated to be responsible for 27% of all cancer related deaths in 2013. The majority of lung cancer incidences are non-small cell lung cancer (NSCLC). The majority of patients (77%) are diagnosed with spread beyond the primary site and 55% of patients have metastatic disease, both of which are associated with high mortality. Currently surgery, EGFR/ALK inhibitors, and chemotherapies are the first-line treatments for advanced NSCLC. Unfortunately, drug resistance and metastasis remain the major hurdles to the outcome of NSCLC treatment. Resistance to TKIs typically occurs less than a year from treatment initiation. The resistance mainly originates from EGFR mutations and crosstalk between signaling pathways. In addition to the severe side effects associated with chemotherapies, selection pressure induced by nonspecific chemotherapeutic drugs and development of drug resistance can also promote metastasis. Surgical removal of the primary tumor can also promote the proliferation of metastases in part due to excessive release of growth factors needed for wound healing. Therefore, there is an urgent need to identify new targets and develop new therapeutics to overcome drug resistance and inhibit metastasis as both a pre-surgical neoadjuvant therapy and in combination with standard-of-care drugs to improve advanced NSCLC management. Steroid receptor coactivator 3 (SRC-3) is a master regulator of cellular growth and development which sits at the nexus of many intracellular signaling pathways critical for cancer formation and proliferation. Down-regulation of SRC-3 can simultaneously inhibit multiple signaling pathways to sensitize tyrosine kinase inhibitors to overcome drug resistance. Furthermore, down-regulation of SRC-3 can significantly reduce cancer cell proliferation, motility, and metastasis. In contrast, inhibition of SRC-3 in normal cells does not influence adult mice life span as demonstrated in a SRC-3 knockout mouse model. Due to the many important features of SRC-3, we will explore this coactivator as a novel target to develop a shRNA based novel cancer therapeutic agent. Development of non-viral gene delivery carriers has recently attracted tremendous attention. Non-viral gene delivery has many advantages over viral delivery, including low immunogenicity and cost-effective scalability. Our group recently developed a star-shaped amine modified polyaspartic acid based polymer (SP) for gene delivery. SP can achieve higher cell transfection efficiency than lipofectamine (50% vs 30%) in vitro. Remarkably, intravenous injection of nanoparticles formed by SP and plasmid DNA encoding luciferase could achieve bioluminescence specifically in the lung with minimal expression from other organs. Therefore, we will take advantage of SP to deliver plasmids encoding shRNA against oncogenes in order to overcome TKI resistance and inhibit metastasis in advanced NSCLC. Contributors: Song, Xianzhou; Feng, Fude; Wang, Jin 376 2014 GRADUATE STUDENT SYMPOSIUM A MATERNAL-ZYGOTIC MODULE STABILIZES F-ACTIN TO PROMOTE ROBUST MORPHOGENESIS Liuliu Zheng Department of Biochemistry & Molecular Biology Advisor: Anna Sokac, Ph.D.-Department of Biochemistry & Molecular Biology Robustness is a property built into biological systems to ensure that the outcome of an event is reliable even when inputs, like gene dosage and environment, vary. During development, robustness safeguards embryos against mistakes that underlie structural and functional birth defects. Yet, our knowledge of how robustness is achieved during mechanical events like embryonic morphogenesis is very limited. Here we show that the tissue-building event of Drosophila cellularization is made robust by mechanisms targeting F-actin. We find that F-actin levels are significantly reduced in cellularizing embryos upon high temperature perturbation. This F-actin reduction leads to failures in cellularization and embryos that do not hatch. Using a genetic strategy, we identified two novel members of the Vinculin/α-Catenin Superfamily that work together to reinforce F-actin against such perturbations (Zheng et al., 2013). We find that zygotically-expressed serendipity-α (sry-α) and maternally-loaded spitting image (spt) are paralogs that share an overlapping, actin-stabilizing activity during cellularization. spt alone is sufficient for cellularization at optimal conditions, but both spt plus sry-α are required at high temperature and when the actin cytoskeleton is genetically compromised by a reduced dose of the actin accessory protein Profilin. Strikingly, we find that Sry-α and Spt expression is precisely controlled, and that there is crosstalk between them. Specifically, Spt provides pre-cellularization functions that Sry-α cannot provide; and expressing elevated levels of Spt alone is not an option because it induces gain-of-function phenotypes. In addition, reducing maternal spt dosage triggers a compensatory up-regulation of zygotic sry-α transcription. We suggest that Sry-α and Spt represent a robustness-promoting module for cellularization: Cross-talk between maternal spt and zygotic sry-α demonstrates that “paralog interactions” can span between maternal and zygotic genomes to promote robustness. In addition, we show for the first time that specific cytoskeletal-based mechanisms promote robust morphogenesis. Since all morphogenesis depends on cytoskeletal remodeling, in embryos and adults, robustness-promoting mechanisms aimed at F-actin could be effective at all life stages. Contributors: Zheng, Liuliu; Xu, Heng; Sepúlveda, Leonardo A.; Lua, Rhonald C.; Lichtarge, Olivier; Golding, Ido; Sokac, Anna Marie 377 BAYLOR COLLEGE OF MEDICINE BAYESIAN NETWORK ANALYSIS OF HIGH-DIMENSIONAL BIOLOGICAL DATA Quan Zhou Program in Structural and Computational Biology and Molecular Biophysics Advisor: Yongtao Guan, Ph.D.-Department of Pediatrics Biological studies often involve complex systems consisting of a large number of molecules, proteins or genes, to which traditional statistical methods are insensitive or even inapplicable. In this era of system biology, network analysis has become a rising trend. The overall goal of this project is to develop a new computational tool for inferring interactions from high-dimensional biological data via Bayesian network (BN). A BN is a directed acyclic graph where the nodes are the variables being investigated and the directed edges represent the causalities. In the past decade, BNs have been widely used in the inference of gene regulation/protein signaling pathways, the modeling of cellular and neural networks, and various genetic association studies. Nevertheless, these applications were seriously limited by the huge computational burden caused by high dimensionality. In this project, we equipped our tool with a sophisticated Markov Chain Monte Carlo sampling algorithm that enabled it to handle 30-100 variables. The program is near completion. Now we are looking for different types of suitable biological data to demonstrate its efficiency in learning from high-dimensional experimental data and ability to perform large-scale integrative analysis of heterogenous data. We hope this study may uncover the magnificent prospect of the BN applications in biological research. Contributors: Guan, Yongtao 378 2014 GRADUATE STUDENT SYMPOSIUM THE ROLE OF VITAMIN D AND VITAMIN D RECEPTOR IN ENDOPLASMIC RETICULUM STRESS RESOLUTION Ying Zhou Integrative Program in Molecular and Biomedical Sciences Advisor: David Moore, Ph.D.-Department of Molecular & Cellular Biology Karl-Dimiter Bissig, M.D./Ph.D.-Department of Molecular & Cellular Biology Vitamin D is synthesized predominantly in the liver and functions as an important secosteroid hormone with pleiotropic effects. While its key regulatory role in calcium and bone homeostasis is well established, recently there is increasing recognition that vitamin D also regulates cell proliferation and differentiation, and has immunomodulatory, anti-inflammatory and anti-fibrotic properties. Vitamin D plays a function mainly through vitamin D-binding protein and vitamin D receptor (VDR), which belongs to the nuclear receptor family. A low vitamin D status is common and may well affect one-third of the global population. Similarly, obesity and its health consequences are a worldwide increasing health burden. The two disorders are closely linked: human obesity is characterized by low serum 25-hydroxyvitamin D3 levels. It’s crucial to find out the potential mechanism of causality between vitamin D deficiency and obesity. The endoplasmic reticulum (ER) stress response, also commonly known as the unfolded protein response (UPR), is an adaptive response used to align ER functional capacity with demand. It is activated in various tissues under conditions related to obesity and type 2 diabetes. Hepatic ER stress contributes to the development of steatosis and insulin resistance, and components of the UPR regulate liver lipid metabolism. Pervious studies show that D signaling is involved in protecting several tissues from stressinduced deteriorating effects. To address the association between vitamin D/VDR signaling and pathology of diabetes, we used pharmacologic ER stress mouse model to test its essential role in ER stress resolution. Tunicamycin is a widely used chemical inducer for ER stress. Both wild type and VDR knockout mice were challenged with TM by intraperitioneal injection. Upon TM injection, VDRKO mice exhibited more severe hepatic steatosis and inflammation than WT mice in 24 hours. VDRKO mice showed more increase in hepatic triglycerides (TG) and cholesterol content. While WT mice almost recovered from the challenge in 72 hours, VDRKO mice still showed severe lipid accumulation in the liver, which suggested a disregulated response to ER stress. We also found that one of unfolded protein response (UPR) branches can’t be activated in VDR knockout mice after TM challenge. And VDR is a potential upstream regulator for XBP1 pathway. Our findings suggest that VD/VDR signaling potentially attenuates the progress of obesity and diabetes by promoting hepatic ER stress elimination. Contributors: Wu Nan; Mamrosh Jennifer; Moore David 379 BAYLOR COLLEGE OF MEDICINE ROLE OF HYPERGLYCEMIA IN DIABETES-ASSOCIATED BREAST CANCER RISK AND PROGRESSION Liyuan Zhu Department of Molecular Virology & Microbiology Advisor: Yi Li, Ph.D.-Department of Molecular & Cellular Biology Background: Epidemiological studies have shown that diabetes increases breast cancer risk and mortality. This correlation may result from hyperinsulinemia and hyperglycemia induced by diabetes. However, most previous studies have focused on the role of insulin and insulin-like growth factor (IGF) pathway while the way hyperglycemia influences breast cancer risk remains unknown. In the past decade, several cohort studies have shown increased breast cancer incidence and mortality rate in people with elevated fasting glucose levels or mild hyperglycemia. Therefore, I hypothesize that hyperglycemia is a key mediator of increased breast cancer risk in diabetic patients. Experimental design and methods: we use hyperglycemic MMTV-Wnt-1 transgenic female mice as our mouse model. Hyperglycemia is achieved through injecting low dose of streptozotocin (STZ) to kill the insulin-producing β-cells in pancreas. We randomized mice into two groups and injected with either STZ or vehicle. Then we palpated the mice for tumor and measured the size of tumor after its appearance three times a week. When the tumor reaches a diameter of 2.0 cm, we euthanize the tumor and collect tumor and lung for further investigation. We also looked at the early lesions development in hyperglycemic MMTV-Wnt-1 mice. Other mouse model, RCAS-neu-injected MA mice are also used to study the influence of hyperglycemia on breast cancer risk and mortality. Results: We observed significantly decreased tumor latency in the hyperglycemic group of mice comparing to the vehicle group. We confirmed that this accelerated tumorigenesis was not because of induction of Wnt-1 oncogene by performing qPCR for Wnt1 and downstream targets in both tumors and early lesions. The tumor growth rates after tumor appearance were similar, supported by the fact that proliferation rate and apoptosis level of tumor cells were not affected. RPPA result indicated that mTOR signaling pathway might involve. We also observed increased number of lung metastatic foci in STZ group of mice, with more larger foci (>25mm2). Conclusion: Hyperglycemia in diabetic mice accelerated tumorigenesis and lung metastasis formation. Hyperglycemia in diabetic patient might cause increased breast cancer risk and mortality. Contributors: Zhu, Liyuan; Bu, Wen; Faust, Claire; Chan, Lawrence; Li, Yi 380 2014 GRADUATE STUDENT SYMPOSIUM DAAM2 PROMOTES TUMOR PROLIFERATION AND MIGRATION IN GLIOBLASTOMA Wenyi Zhu Integrative Program in Molecular and Biomedical Sciences Advisor: Benjamin Deneen, Ph.D.-Department of Neuroscience Glioblastoma multiforme (GBM) is one of the most common and aggressively malignant high-grade gliomas in humans. GBM tumor as are comprised of astrocytes and oligodendrocytes. Genes involved in the development of these glial cells have been liked to GBM tumorigenesis. Considering tumorigenesis is currently viewed as a convergence of genetic mutation and developmental context, we want to understand how the mechanisms which govern glial cell fate decisions contribute to GBM formation. The Wnt signaling pathway has well defined functions in progenitors self-renewal during development. This function is dysregulated in several cancers, yet surprisingly poorly defined in GBM. Given our finding that Daam2 plays a key role in Wnt signaling in the developing and CNS, we hypothesize that Daam2 is a key component of the Wnt signaling pathway which is dis-regulated in GBM. Indeed, we found that Daam2 is highly expressed in GBM. Subsequent analysis of Daam2 function in GBM revealed that it promotes tumor proliferation, and migration. Currently, I am working to precisely define its molecular mechanism. We surprisingly find that Daam2 is not only involved in Wnt signaling pathway in GBM, but also interactions with PI3K. These studies represent the first characterization of Daam2/Wnt signaling/ PI3K function in GBM. Contributors: Zhu, Wenyi; Lee, Hyun-Kyoung; Loturco, Joseph; Mohila, Carrie; Deneen, Benjamin 381 BAYLOR COLLEGE OF MEDICINE IN VIVO TWO-PHOTON CALCIUM IMAGING OF SUBCELLULAR INPUT RETINOTOPY IN AN IDENTIFIED VISUAL INTERNEURON Ying Zhu Program in Structural and Computational Biology and Molecular Biophysics Advisor: Fabrizio Gabbiani, Ph.D.-Department of Neuroscience The Lobula Giant Movement Detector (LGMD) is an identified visual interneuron in the lobula neuropil of the locust optic lobe that responds preferentially to objects approaching on a collision course. The LGMD receives excitatory input from the entire visual hemifield sampled by one eye onto one of its dendritic fields, called dendritic field A. Calcium imaging of dendritic field A using a CCD camera has documented synaptic calcium signals in response to localized visual stimuli that preserve retinotopy down to the level of a single facet on the compound eye. Because single photon imaging with CCD sensors has a relatively low penetration depth and exhibits strong scattering, previous work could not investigate this retinotopic mapping at the level of individual thin dendritic branches. Our current work employs a custom-built two-photon microscope with sub-micron resolution in conjunction with an Organic Light-Emitting Diode (OLED) microdisplay that provides visual stimuli to the locust compound eye adequate to explore this retinotopy at the finest level. We find that adjacent facets on the compound eye have overlapped mappings on the LGMD’s excitatory dendritic branches. When the size of the visual stimuli decreases while their center location is kept unchanged, the overlap of the mapping and the number of activated branches also decrease. Therefore, in vivo two-photon calcium imaging is a suitable tool to understand the subcellular retinotopy in this neuron. Contributors: Zhu, Ying; Gabbiani, Fabrizio 382 2014 GRADUATE STUDENT SYMPOSIUM LOSS OF PALS1 IN SCHWANN CELLS LEADS TO RADIAL SORTING DEFECTS Daniel Richard Zollinger Department of Neuroscience Advisor: Matthew Rasband, Ph.D.-Department of Neuroscience Schwann cells in the peripheral nervous system (PNS) and oligodendrocytes in the central nervous system (CNS) surround
Similar documents
Graham Scallan Leading Business Counselor in Wexford
An effective and experienced business mentor can help to take your business to top of the roof. The Graham Scallan is the best leading business counselor in Wexford. Get advice from him to make your business successful. For More Info : http://www.grahamscallan.com/
More information