PROGRAM BOOK

Transcription

PROGRAM BOOK
2016 IETS
42nd ANNUAL CONFERENCE
PROGRAM
BOOK
Program Book
42nd Annual Conference of the
International Embryo Transfer Society
Embryo Resilience
in the Face of Biotechnology
Galt House Hotel
Louisville, Kentucky
January 23–26, 2016
Scientific Program Co-Chairs:
Marcelo Bertolini and François-Xavier Grand
Table of Contents
Preface and Acknowledgments......................................................................................................... 1
2016 Recipient of the IETS Pioneer Award...................................................................................... 3
Map of the Venue.............................................................................................................................. 4
General Information.......................................................................................................................... 6
Program............................................................................................................................................. 8
Section Editors and Manuscript and Abstract Reviewers............................................................... 15
Poster Session Information............................................................................................................. 17
Poster Session Order by Topic........................................................................................................ 18
Oral Session Order by Topic........................................................................................................... 36
Author Index................................................................................................................................... 38
2016 Recipients of the IETS Distinguished Service Award........................................................... 46
Special Events................................................................................................................................. 49
Exhibit Hall Layout......................................................................................................................... 52
Exhibit Directory............................................................................................................................ 53
Preconference Symposium on Biomaterials Repositories.............................................................. 63
2015 IETS Board of Governors
Gregg P. Adams, Immediate Past President
Patrick Blondin, President
Christine Wrenzycki, Vice President
Barbara S. Durrant, Treasurer
Fulvio Gandolfi, Governor
Irina A. Polejaeva, Governor
Daniel F. Salamone, Governor
Ann Van Soom, Governor
Preface and Acknowledgments
Greetings!
The 42nd Annual Conference of the International Embryo Transfer Society will be held at the Galt House in
Louisville, Kentucky, USA, from January 23 to 26, 2016. When we defined this year’s main theme, “Embryo
resilience in the face of biotechnology,” the goal was to address how the embryos were “adapting” to biotechnologies, using a complementary basic and applied approach. Our scientific community makes progress faster
every day. There is not a week without significant new developments or applications. This week, as an example,
the birth of IVF puppy dogs has just been published. In this context, there is a need to gather all expertise
and disciplines in order to investigate the extent of the short- and long-term effects of biotechnologies on the
embryo, fetus, and offspring development. There will be five plenary sessions featuring 10 invited speakers,
who will address topics on the effect of biotechnologies on gamete competence and embryo development, differentiation pathways and early embryo developmental competence, effect of events between ET and pregnancy
recognition on pregnancy outcome or early embryo mortality, decisive conceptus–endometrial interactions from
pregnancy recognition to fetal phase through implantation, and long-term consequences of early developmental
events. George Seidel will enlighten us as the keynote speaker. We thank all the speakers for their great insights,
outstanding knowledge, and high quality of the scientific material shared with us, along with their kind patience
and willingness to work hard to meet our short deadlines.
In addition to the regular format of our society meeting, the program this year will include two concurrent
forums: a DABE forum (Domestic Animal Biomedical Embryology), organized by Carol Keefer, and a practitioners forum, organized by Kevin Lindel and Peter Hansen. To extend the applied scope of this meeting, there
will also be three concurrent applied sessions on bovine reproduction, male fertility, and equine reproduction
organized by Gabriel Bó, Erdogan Memili, and Barry Ball, respectively. Finally, we will also have a session on
selected invited abstracts for oral presentations.
This year, 275 abstracts were submitted and 256 were accepted for the journal. Overall, over one hundred colleagues reviewed manuscripts and abstracts this year. These reviewers represent a wide range of geographic
area over the world, which serves to bring a broad set of perspectives to our society. We would like to thank
all the Session Editors and reviewers for their time and effort in the review process. We would like to take this
opportunity to thank the Morulas, for working with us in selecting candidate students to co-chair the sessions,
and the senior co-chairs, for volunteering to host our invited speakers. Also, we received 57 submissions for the
student competition, for which we thank Charles Rosenkrans for his leading role in selecting the finalists. We
also send our appreciation to the IETS Board of Governors and Executive Board for their support in preparing
for the 2015 Annual Conference. Enough thanks cannot be expressed to Debi Seymour, our IETS Executive
Secretary, for her invaluable guidance, advices and assistance in every single step of the organization of this
meeting. Our sincere appreciation is extended to Dr. Graeme Martin, Editor-in-Chief, and Sussan Au, publisher,
of Reproduction, Fertility and Development for their help in the production of the conference booklets and
proceedings.
Finally, we thank you all for attending the conference. Your participation in the scientific program has been the
very soul of our conferences. Every single successful cooking recipe has its own magic ingredient: your contribution to the event will be the magic ingredient! We thank YOU for your faithful support of our society and
for attending the meetings. It has been a great pleasure and a privilege to serve you and our society as program
co-chairs. We deeply encourage you to initiate, stimulate, and challenge discussions. Interactions are what will
always keep our society on the path of success.
Hope to see you all in Kentucky!
Marcelo Bertolini and François-Xavier Grand
2016 IETS Program Co-Chairs
International Embryo Transfer Society
1
2
42nd Annual Conference
2016 Recipient of the IETS Pioneer Award
Charles Earle Pope
Award Presentation: Tuesday, January 26, at 9:30
Previous Recipients
K. H. S. Campbell (2015)
B. Brackett (2004)
R. L. Brinster (1992)
J.-P. Renard (2015)
K. Betteridge (2003)
A. K. Tarkowski (1991)
W. W. Thatcher (2014)
R. H. Foote (2002)
J. D. Biggers (1990)
J. Hahn (2013)
P. J. Dziuk (2001)
C. Thibault (1989)
O. J. Ginther (2012)
R. Yanagimachi (2000)
A. L. McLaren and D. Michie (1988)
I. Wilmut (2011)
R. M. Moor (1999)
E. J. C. Polge (1987)
R. J. Mapletoft (2010)
I. Gordon (1998)
T. M. Sugie (1986)
S. P. Leibo (2009)
S. Wintenberger-Torres (1997)
L. E. A. Rowson (1985)
G. Seidel Jr. (2008)
W. K. Whitten (1996)
L. E. Casida (1984)
A. Iritani (2007)
C. R. Austin (1995)
M. C. Chang (1983)
D. Kraemer (2006)
N. W. Moore (1994)
R. O. Berry (1982)
S. Willadsen (2005)
R. G. Edwards (1993)
International Embryo Transfer Society
3
Map of the Venue
Galt House Hotel
140 N 4th Street, Louisville, Kentucky 40202
Second Floor (Ballroom Level)
4
42nd Annual Conference
Map of the Venue
Galt House Hotel
140 N 4th Street, Louisville, Kentucky 40202
Third Floor (Conservatory Skywalk Level)
International Embryo Transfer Society
5
General Information
Meeting Room Directory
Main Conference Sessions
Grand Ballroom C; Concurrent Sessions: Ballroom C, Jones, and Nunn
Exhibits
Grand Ballroom A, B
Poster Displays
Grand Ballroom A, B
Please see the Scientific Program on page 8 for additional room assignments.
Registration Desk Hours
The registration desk is located at GHE 2nd floor.
Pick up of preregistration packets
Friday, January 22
16:00–19:00
On-site registration hours
Saturday, January 23
07:00–18:00
Sunday, January 24
07:00–18:00
Monday, January 25
07:30–16:00
Tuesday, January 26
08:00–15:00
Exhibit Information
Grand Ballroom A, B
Setup
Saturday, January 23
09:00–17:00
Exhibits open
Sunday, January 24
09:00–19:00
18:00–19:00 (Reception)
Monday, January 25
08:00–17:00
Tuesday, January 26
08:30–13:00
Teardown
Tuesday, January 26
13:00–17:00
Details on the exhibitors can be found in the Exhibit Directory on page 53.
Badges
As a security requirement, we request that all participants wear their conference name badges to all sessions and
social functions.
6
42nd Annual Conference
Certificates of Attendance and Presentation
A Certificate of Attendance will be included in your badge packet.
Currency
The dollar is the legal tender in the United States. Should you need to exchange your local currency, you will be
able to make exchanges at the larger airports, Denver, Los Angeles, Atlanta, Dallas, or Houston, and at Louisville
International Airport. On the upper level, you will find a full-service bank, Fifth Third Bank that offers currency
exchange.
Messages
Any messages received for conference delegates will be posted on the message board located near the registration desk.
Refreshments
Morning and afternoon refreshments are included in your registration fee and are provided during the scheduled
break times in the exhibit area located in Grand Ballroom A, B.
Dining and Entertainment
There are six restaurants and lounges located in the Galt House Hotel.
RIVUE Restaurant and Lounge has it all—looks, style, and taste. Voted one of Louisville’s best restaurants,
RIVUE, our rooftop restaurant, features expansive views of downtown and the waterfront.
Café Magnolia is the perfect place to dine casually and enjoy breakfast, lunch, or dinner while savoring delectable
eats.
Al J’s at the Conservatory is in the center of it all and voted best place to “sit a drink.” Grab a bite or beverage at
the 30-foot aquarium bar, offering sweeping views of downtown and the waterfront.
Jockey Silks Bourbon Bar offers a place to relax and unwind while choosing from more than 150 Kentucky bourbons. The cozy venue was named one of Whisky Magazine’s “Best Bourbon Bars in the World.”
Thelma’s at the Conservatory is a 24-hour deli for that caffeine fix whenever the desire hits. Select from the wide
assortment of deli sandwiches, salads, specialty drinks, and more.
Jeff Ruby’s Steakhouse is a New York–style steakhouse specializing in steak, seafood, and sushi and featuring a
dynamic atmosphere with live entertainment.
Starving for something new? Exciting? Original? You’ll find it in Louisville’s restaurants, diners, taverns, and takeouts, where the selection ranges from perennial regional favorites to international standards of taste and refinement.
Louisville restaurants blend traditional Kentucky cooking with a cosmopolitan mix of eclectic international cuisine
including French, Italian, and Mediterranean fare; Mexican and American Southwest influences; as well as classic
barbecue. All can be found within the downtown area a short walk from the Galt House Hotel.
Services and Amenities
The Galt House Hotel services and amenities include a state-of-the-art rooftop fitness center, in-room safes, security,
business center, six restaurants and lounges, massages, valet parking, and many more services and amenities that will
make your stay feel as if you were in the comfort of your home. The Galt House Hotel offers complimentary Wi-Fi
in all public areas and wireless Internet in the guest rooms.
There is a shopping shuttle service for the guests to the new Outlet Shoppes of the Bluegrass, Wednesday to Sunday,
Departing the Galt House Hotel at 10:00 and departing the Outlet Shoppes of the Bluegrass at 14:00 (estimated
travel is 25 minutes). In addition to the complimentary round-trip shuttle, each guest will receive a shopping coupon booklet valued at
$250.
International Embryo Transfer Society
7
Program
Thursday, January 21
08:00–17:00 IETS Board of Governors meeting (Willis)
Friday, January 22
08:00–15:00
09:00–18:00
13:00–18:00
16:00–19:00
IETS Board of Governors meeting (Willis)
HASAC Research Subcommittee meeting (Nunn)
W2171 Research Committee (Collins)
Registration (GHE 2nd Floor Registration A, B, C)
Saturday, January 23
07:00–18:00
08:00–17:30
09:00–17:00
13:30–17:00
Registration (GHE 2nd Floor Registration A, B, C)
CANDES Preconference Symposium (Jones)
Commercial Exhibit Setup and Poster Setup (Grand Ballroom A, B)
IETS Foundation Board of Trustees Meeting (Willis)
Morulas Preconference Workshop—Epigenetics and embryo technology (Nunn)
Sponsored by CSIRO
14:00–14:50 Session I: Introduction to epigenetics and bovine embryo research
Pablo Ross, University of California
14:50–15:40 Session II: The analysis of DNA methylation in bovine embryos and sperm—
Prospects and challenges
Marc-André Sirard, Université Laval
15:40–16:00Break
Sponsored by CSIRO
16:00–16:50 Session III: Research in developmental epigenetics and laboratory techniques
Jason Knott, Michigan State University
16:50–17:00 Discussion and Closing Remarks
Sunday, January 24
06:30–08:00
07:00–08:25
07:00–18:00
07:00–08:30
07:00–08:30
Poster Setup (Grand Ballroom A, B)
HASAC Forms and Certificates Subcommittee Meeting (Breathitt)
Registration (GHE 2nd Floor Registration A, B, C)
Past-Presidents’ Breakfast (Brown)
Graduate and Undergraduate Student Competition Presenters Breakfast, with IETS Foundation
Education Committee Chair (Clements)
07:00–18:00 Affiliates Lounge (Wilson)
09:00–19:00 Commercial Exhibition (Grand Ballroom A, B)
08:45–09:00 Opening and Welcome—Marcelo Bertolini, François-Xavier Grand, and Edward Squires (Ballroom C)
8
42nd Annual Conference
Embryo Resilience in the Face of Biotechnology
Session I: Effect of the Environment on Gamete Competence and Embryo Development (Ballroom C)
Session co-chairs: Dimitrios Rizos, Instituto Nacional de Investigación y Tecnologia Agraria y Alimentaria, and
Paula Tribulo, University of Florida
09:00–09:45 Causes and consequences of oxidative stress in spermatozoa
John Aitken, University of Newcastle, Australia
09:45–10:30 Fertility and genomics: Comparison of gene expression in contrasting reproductive tissues in female
cattle
Alex Evans, University College Dublin, Ireland
10:30–11:00 Refreshment Break and Poster Exhibit and Exhibition (Grand Ballroom A, B)
IETS Foundation Student Competition Presentations (Ballroom C)
Session chair: Charles F. Rosenkrans, University of Arkansas
11:00
Generation of a stable transgenic swine model for cell tracking and chromosome dynamic studies
R. Sper, S. Simpson, X. Zhang, B. Collins, and J. Piedrahita (Abstract 1)
11:15
Circulating microRNA signatures associated with early bovine pregnancy
J. Ioannidis and X. Donadeu (Abstract 2)
11:30
Role of β-defensin 126 in promoting sperm motility in cattle
B. Fernandez-Fuertes, F. Narciandi, K. G. Meade, C. O’Farrelly, S. Fair, and P. Lonergan (Abstract 3)
11:45
Suppression of ASH2L alters DNA methylation and histone patterns during bovine embryonic
development
M. D. Snyder, J. H. Pryor, M. D. Peoples, G. L. Williamson, M. C. Golding, M. E. Westhusin, and
C. R. Long (Abstract 4)
12:00
12:15
A single nucleotide polymorphism in COQ9 affects mitochondrial function, body weight change after
calving, and fertility in Holstein cows
M. S. Ortega, S. Wohlgemuth, D. J. Null, J. B. Cole, and P. J. Hansen (Abstract 5)
The effects of depleting donor cell mitochondrial DNA on cattle embryos derived from somatic cell
nuclear transfer
K. Srirattana and J. C. St. John (Abstract 6)
12:30–14:00
12:30–14:00
12:30–14:00
12:30–14:00
Lunch Break
IETS Board Luncheon with Affiliate Society Representatives (Willis)
HASAC Emerging Technologies (Breathitt)
Morulas and Mentor Luncheon (Clements)
Sponsored by Vétoquinol
Session II: Differentiation Pathways and Early Embryo Developmental Competence (Ballroom C)
Session co-chairs: Bianca Gasparrini, University of Naples Federico II, and Carina Blaschka, Justus-Liebig-University
14:00–14:45 Mechanisms of epigenetic remodeling during preimplantation development
Pablo Ross, University of California, USA
14:45–15:30 Measuring embryo metabolism to predict embryo quality
Jeremy Thompson, University of Adelaide, Australia
15:30–16:00 Refreshment Break and Poster Exhibit and Exhibition (Grand Ballroom A, B)
International Embryo Transfer Society
9
Concurrent Forum
16:00–18:00 Practitioners’ Forum (Ballroom C)
Co-chairs: Peter J. Hansen, University of Florida, and Kevin A. Lindell, Tufts University
Sponsored by Partnar Animal Health Inc.
Research focus on recipient synchronization
Pietro Baruselli, Universidade de São Paulo, and Milo Wiltbank, University of Wisconsin
Panel discussion on practical aspects of handling IVP embryos and semen for IVF
Jeremy Block, Ovatech; Bill Croushore, White Oak Veterinary Clinic; Shantille Kruse, Boviteq USA;
and Hong Wei, Transova
Selected short presentations from the audience (limited to 2 slides total per person). Selection will be
based on relevance.
Concurrent Forum
16:00–18:00 Domestic Animal Biomedical Embryology (DABE) Forum (Nunn)
Chair: Carol Keefer, University of Maryland
Sponsored by Hamilton Thorne Inc.
Contributions from new investigators
Use of transwell cell culture and 3-dimensional printing technology to develop an in vitro bovine
oviduct (Abstract 52)
Marcia de Almeida Monteiro Melo Ferraz, doctoral candidate, Utrecht University, Utrecht,
the Netherlands
Effects of microvesicles secreted from equine amniotic-derived progenitor cells on in vitro lipopolysaccharide-treated tendon and endometrial cells (Abstract 226)
Anna Lange-Consiglio, PhD, Reproduction Unit, Large Animal Hospital, University of Milan, Italy
Generation of interspecies chimeras between primate induced pluripotent stem cells and porcine
parthenogenetic embryos (Abstract 223)
Monika Nowak-Imialek, DMV, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut,
Mariensee, Hannover, Germany
Efficient edition of the bovine PRNP prion gene in somatic cells and IVF embryos using the clustered
regularly interspaced short palindromic repeats (CRISPR)/CAS9 system (Abstract 243)
Romina Bevacqua, PhD assistant researcher, Animal Biotechnology Laboratory, Buenos Aires
University, Argentina
18:00–19:00 Welcome Reception (Grand Ballroom A, B)
Sponsored by Professional Embryo Transfer Supply Inc. (PETS)
Monday, January 25
07:30–16:00
07:00–18:00
07:30–08:30
08:00–17:00
10
Registration (GHE 2nd Floor Registration A, B, C)
Affiliates Lounge (Wilson)
HASAC Regulatory Subcommittee Meeting (Breathitt)
Commercial Exhibits (Grand Ballroom A, B)
42nd Annual Conference
Concurrent Session
08:00–09:30 Bovine reproduction: Donor selection and treatments to improve oocyte collection and in vitro
embryo production in cattle (Ballroom C)
Chair: Gabriel Bó, Universidad Nacional de Villa María
08:00–08:15 Influence of breed and season on in vitro embryo production
Gabriel Bó, Universidad Nacional de Villa María, Argentina
08:15–08:30 The importance of follicular differentiation to obtain fully competent bovine oocytes
Marc-André Sirard, Université Laval, Canada
08:30–08:45 Superstimulation strategies for ovum pickup in Holstein donors
Lais Vieira and Pietro Baruselli, Universidade de São Paulo, Brazil
08:45–09:00 Optimizing donor efficiency with follicle stimulating hormone superstimulation in a commercial in
vitro fertilization program in Holsteins
François-Xavier Grand, Boviteq, Canada
09:00–09:30 Roundtable Discussion
Concurrent Session
08:00–09:30 Equine reproduction: Pre- and post-ovulatory influences on fertility (Nunn)
Chair: Barry Ball, University of Kentucky
08:00–08:30 Anti-Müllerian hormone: Implications for follicular reserve, follicular function, and fertility in the mare
Barry Ball, Gluck Equine Research Institute; University of Kentucky, USA
08:30–09:00 Factors influencing the success of equine intracytoplasmic sperm injection in a clinical program
Katrin Hinrichs, Texas A&M University, USA
09:00–09:15 Effects of low circulating progesterone concentrations during early diestrus on the endometrial
transcriptome of the mare
Alex Esteller-Vico, University of Kentucky, USA
09:15–09:30 The mare model for luteinized unruptured follicle syndrome: Intrafollicular endocrine milieu
Eduardo Gastal, Southern Illinois University, USA
Concurrent Session
08:00–09:30 From bench to barn: Effects of bull fertility or sperm factors on embryo development and
cattle agriculture (Jones)
Chair: Erdoğan Memili, Mississippi State University
08:00–08:20 Systems biology of sperm and bull fertility
Erdoğan Memili, Mississippi State University, USA
08:20–08:40 Seminal plasma proteins: Functional attributes and potential markers of fertility
Arlindo A. Moura, Federal University of Ceará, Brazil
08:40–09:00 Genomic evaluation of fertility traits and discovery of haplotypes that affect fertility of U.S. dairy cattle
George R. Wiggans, Agricultural Research Service, USDA, USA
09:00–09:20 Prospects for improving semen quality and bull fertility: An industry perspective
Nancy Tisch, International Center for Biotechnology, Cooperative Resources International, USA
09:20–09:30Discussion
Abdullah Kaya, Selcuk University, Turkey
09:30–10:00 Distinguished Service Awards (Ballroom C)
International Embryo Transfer Society
11
10:00–10:45 Invited Abstract Oral Presentations (Ballroom C)
Chair: Matthew Wheeler, University of Illinois
Genome editing of somatic cell nuclear transfer derived zygotes by clustered regularly interspaced
short palindromic repeats (CRISPR) CAS9 guide rna injection
K. M. Whitworth*, S. L. Murphy, J. A. Benne, L. D. Spate, E. Walters, R. Hickey, S. L. Nyberg,
K. D. Wells, and R. S. Prather (Abstract 25)
Unveiling the role of lipids in organogesis: Molecular anatomy by desorption electrospray ionization
mass spectrometry imaging mass spectrometry
V. Pirro*, P. O. Favaron, C. R. Ferreira, L. S. Eberlin, R. S. Barreto, R. G. Cooks, and M. A. Miglino
(Abstract 61)
Interception of exosomal messages between the oviduct and the embryo: What are they tweeting about?
C. Almiñana*, E. Corbin, G. Harichaux, V. Labas, G. Tsikis, C. Soleilhavoup, K. Reynaud, X. Druart,
and P. Mermillod (Abstract 78)
Follicular fluid microRNA sequences as biomarkers of competent oocytes in cattle
R. Pasquariello*, N. Fiandanese, A. Viglino, P. Pocar, J.L. Williams, and F. Gandolfi (Abstract 148)
Inhibition of 5α-reductase during late gestation in the mare
M. Wynn*, E. Legacki, A. Conley, S. Loux, A. Esteller-Vico, S. Stanley, E. Squires, M. Troedsson, and
B. Ball (Abstract 67)
10:45–11:15
10:45–12:45
12:45–14:00
12:45–14:00
12:45–14:00
12:45–14:00
12:45–14:00
Refreshment Break/Poster Exhibit and Exhibition (Grand Ballroom A, B)
Poster Session I (Grand Ballroom A, B)
Lunch Break
Exhibitors' Lunch with IETS Board of Governors (Brown)
IETS Data Retrieval Committee Meeting (Willis)
HASAC Manual Meeting (Breathitt)
Morulas Career Luncheon (Clements)
Sponsored by CSIRO
Session III: Between ET and Pregnancy Recognition Through Elongation: Impact on Pregnancy
Outcome and/or Early Embryo Mortality (Ballroom C)
Session co-chairs: Michael Hōelker, University of Bonn, and Jacob Stewart, University of Illinois
14:00–14:45 Determinant molecular markers for peri-gastrulating bovine embryo development
Isabelle Hue, INRA, France
14:45–15:30 The role of progesterone in embryo development in cattle
Patrick Lonergan, University College Dublin, Ireland
15:30–16:00
16:00–17:00
17:00–18:00
Poster Exhibit and Exhibition and Break (Grand Ballroom A, B)
IETS Business Meeting (Ballroom C)
Peter Farin Trainee Award Winners Presentations (Ballroom C)
Co-chairs: Pablo Ross, University of California, and Jennifer Barfield, Colorado State University
Cumulus-oocyte-complex secretions from the first 8 hours of in vitro maturation affect oocyte
developmental competence
Karen Uhde, Utrecht University (Abstract 66)
Long-term propagation and cryopreservation of cat spermatogonial stem cells
Lindsey Vansandt, Cincinnati Zoo and Botanical Garden (Abstract 229)
12
42nd Annual Conference
Association of G6PDH-activity with oocyte developmental competence and molecular characteristics
in goats
Mia Yang, Utah State University
A metabolic approach towards optimizing developmental competency of in vitro matured equine
oocytes
Niamh Lewis, University of Liverpool
Immunolocalization of steroid sulfatase (STS) and the estrogen-specific sulfotransferase (SULT1E1)
in bovine follicles
Carina Blaschka, Justus-Liebig-University (Abstract 54)
Consequences of in vitro production of embryos with or without colony-stimulating factor 2 in
culture medium on morphometric features of the bovine conceptus at day 86 of gestation
Luiz Gustavo Bruno Siqueria, University of Florida (Abstract 175)
18:00–19:00 HASAC Open Meeting (Nunn)
18:00–19:00 Morulas Forum (Ballroom C)
19:30
Morulas Student Mixer (Galt House Hotel, Al J's at the Conservatory)
Tuesday, January 26
07:00–08:30
07:00–18:00
08:00–15:00
08:00–13:00
Organizational Meeting of the IETS Board of Governors (Willis)
Affiliates Lounge (Wilson)
Registration (GHE 2nd Floor Registration A, B, C)
Commercial Exhibits (Grand Ballroom A, B)
Session IV: From Pregnancy Recognition to Fetal Phase Through Implantation: Decisive ConceptusEndometrial Interactions (Ballroom C)
Session co-chairs: Barbara Durrant, San Diego Zoo Institute for Conservation Research, and Niamh Lewis,
University of Liverpool
08:00–08:45 Adaptability and potential for treatment of placental functions to improve embryonic development
and postnatal health
James C. Cross, University of Calgary, Canada
08:45–09:30 Pregnancy losses in cattle: Potential for improvement
Michael G. Diskin, Animal and Grassland Research and Innovation Centre, Ireland
09:30–10:00 Pioneer Award (Ballroom C)
10:00–10:30Break
10:00–12:00 Poster Session II and Exhibition (Grand Ballroom A, B)
12:00–12:30 14th Annual IETS Running Competition, Louisville Waterfront Park
12:00–13:30 Lunch Break
12:00–13:30 Organizational Lunch Meeting of the IETS Foundation (Willis)
12:00–13:30 2016, 2017, 2018 IETS Program Committee Lunch (Breathitt)
13:00–17:00 Commercial Exhibit and Poster Takedown (Grand Ballroom A,B)
Session V: Long-Term Outcomes of Early Developmental Events (Ballroom C)
Session co-chairs: Hosup Shim, Dankook University, and Steve Yang, University of Saskatchewan
13:30–14:15 Breeding animals for quality products: Not only genetics
Pascale Chavatte-Palmer, INRA, France
International Embryo Transfer Society
13
14:15–15:00 Livestock in biomedical research: History, current status and future prospective
Irina Polejaeva, Utah State University, USA
Session VI: Keynote Address (Ballroom C)
Session chair: Matthew Wheeler, University of Illinois
15:00–15:45 Assisted reproduction with gametes and embryo: What research is needed and fundable?
George Seidel, Colorado State University, USA
Awards Presentations and Updates (Ballroom C)
15:45–16:15 IETS Foundation Student Competition Awards, CANDES, DABE, and HASAC Updates
16:15–16:30 Closing Ceremony (Ballroom C)
18:30–22:30 Closing Party (Kentucky Derby Museum)
14
42nd Annual Conference
Section Editors and Manuscript and Abstract Reviewers
The Program Co-Chairs Acknowledge and Thank the Following People
Section Editors
Charles Rosenkrans, Student Competition
João Batista Borges, Artificial Insemination
Daniel Salamone, Cloning and Nuclear Transfer
Pierre Comizzoli, Cryopreservation and Cryobiology
Charles Long, Developmental Biology
Pat Lonergan, Early Pregnancy and Pregnancy
Recognition
Melanie McDowall, Embryo Culture
David Wells, Embryo Manipulation
Jeremy Block, Embryo Transfer
Ann Van Soom, Epidemiology and Diseases
Barbara Durrant, Exotic Species
Fulvio Gandolfi, Folliculogenesis and Oogenesis
Christine Wrenzycki, Gene Expression
Hiroaki Funahashi, IVF and IVP
John Kastelic, Male Physiology
Trudee Fair, Oocyte Activation
Dulama Richani, Oocyte Maturation
Ulrike Taylor, Sexing
Cesare Galli, Sperm Injection
Tiziana Brevini, Stem Cells
Osamu Dochi, Superovulation
Carol Keefer, Transgenesis
Charles Rosenkrans, Undergraduate Poster Competition
Manuscript and Abstract Reviewers
Tomas J. Acosta
Gregg Adams
Paulo L. Aguiar
Ramiro Albeiro
Fernanda D. Lamdim
Alvarenga
Gary Anderson
Hernan Baldassarre
Barry Ball
Stephan Barcikowski
Jennifer Barfield
Pawel M. Bartlewski
Pietro Baruselli
Andrea Cristina Basso
Don Bergfelt
Ann Sofi Bergqvist
Michael Bertoldo
Jeff Betthauser
Romina Bevacqua
Maajid Hassan Bhat
Andrezj Bielanski
Mario Binelli
Terry Blanchard
Gabriel Bo
Ken Bondioli
Vilceu Bordignon
Angie Boyer
Maria Julia Bragado
Steven Brinsko
Leo Brito
Jose Buratini
Henrik Callesen
Diogo Camara
Fernando Campos-Chillon
Luis Sergio Almeida
Camargo
Prithviraj Chakravarty
Pascale Chavatte-Palmer
Silvia Colleoni
Pierre Comizzoli
Anna Lange Consiglio
Martina Crispo
Xiang-Shun Cui
Dinesh Dadarwal
Bradford Daigneault
Philip Damiani
Sebastian E. Demyda
Anna Denicol
Catharina Deschauwer
Andras Dinnyes
Margo Dode
Xavier Donadeu
José Eduardo dos Santos
Kylie Dunning
Veronique Duranthon
Barbara Durrant
Alan Ealy
Judith Eckert
Sean Fair
Trudee Fair
Laura Falchi
Charlotte Farin
Richard Fayrer-Hosken
Roseli Fernandes Gonzalez
Mariana Fernandes
Machado
Manoel Sa Filho
Niamh Forde
Hiroaki Funahashi
Cesare Galli
Andres Gambini
Luis Garcia-Marin
Bianca Gasparrini
John Gibbons
Michael Golding
Martha Gomez
Karen Goodrowe
Andres Gonzalez Serrano
Hanna Grothmann
Alfonso Gutiérrez-Adán
Mary Hagedorn
International Embryo Transfer Society
Johannes Handler
Carol Hanna
Maria Ines Hiriart
Michael Höelker
Harlan Howard
Poul Hyttel
Sang-Hwan Hyun
Eleonora Iacono
Kei Imai
Yubyeol Jeon
K. L. Jones
Poothappillai Kasinathan
John Kastelic
Carol Keefer
Kazuhiro Kikuchi
I. H. Kim
Rebecca Krisher
Wilfried Kues
Giovanna Lazzari
Tamara Leahy
Daniel Le Bourhis
Rejean Lefebvre
Khomotso Lekola
Valentina Lodde
Pat Lonergan
Charles Long
Charles Looney
15
Barbara Loureiro
Charles Love
Alberto Luciano
Mahdi Mahdipour
Michael Mahony
Satoko Matoba
Melanie McDowall
Serge McGraw
Flavio Vieira Meirelles
Erdoğan Memili
Alberto Menchaca
Evelyne Meyer
Maria Angelica Miglino
Marta Munoz
Bruce Murphy
Taku Nagai
Lucky Nedambale
Amir Niasari-Naslaji
Heiner Niemann
Monika Nomm
16
Bjorn Oback
Lynne O’Shea
John Parrish
Georgia Pennarossa
Jorge Piedrahita
Bill Plummer
Irina Polejaeva
Jose Henrique Pontes
Earle Pope
Randall Prather
Christopher Price
Detlef Rath
Eduardo Ribeiro
Dulama Richani
Dimitrios Rizos
Sangho Roh
Sergio Romero
Rick Rorie
Ryan Rose
Charles Rosenkrans
Pablo Ross
Terri Roth
Tricia Rowlison
Rainer Saner
Marina Sansinena
Roberto Sartori
Carey Satterfield
Dragos Scarlet
Hosup Shim
Kevin Sinclair
Jaswant Singh
Luiz Siqueira
Lawrence Smith
Katrien Smits
Nucharin Songsasen
Jacob W. Stewart
Roger Sturmey
Ali Reza Talebi
Ali Cihan Taskin
Ulrike Taylor
42nd Annual Conference
Bhanu Telugu
Dawit Tesfaye
William Thatcher
Michel Thibier
Jeremy Thompson
Gabor Vajta
Ann Van Soom
Paul J. Verma
Rajneesh Verma
Takuya Wakai
David Wells
Mark Westhusin
Brett White
Eckhard Wolf
Christine Wrenzycki
Tim Xiang
Jianbo Yao
Jimena Yapura
Agca Yuksel
Hashiyada Yutaka
Poster Session Information
Location
Posters are located in Grand Ballroom A, B of the Galt House Hotel on the second floor of the Suite Tower (see map
on page 4).
Poster Numbers
Posters are identified by the number corresponding to the abstract number in Reproduction, Fertility and
Development 2016; 28 (1). Numbering of the posters begin at 1 and ends at 245.
Setup
Posters can be put up from 08:00 to 17:00 on Saturday, January 23, 2016, and from 06:30 to 08:00 on Sunday,
January 24, 2016. All posters must remain up throughout the meeting. Authors of posters not put up by 08:00 on
Sunday will be reported to the IETS President for possible disciplinary action.
Poster Session I
Presentations by authors of odd-numbered abstracts (e.g., 7, 9, 11) in Reproduction, Fertility and Development
2016; 28 (1), and the Student Competition finalist and Undergraduate finalist poster presentations will take place
Monday, January 25, 2016, from 10:45 to 12:45.
Poster Session II
Presentations by authors of even-numbered abstracts (e.g., 8, 10, 12) in Reproduction, Fertility and Development
2016; 28 (1), will take place Tuesday, January 26, 2016, from 10:00 to 12:00.
Teardown
Poster teardown will take place from 13:00 to 17:00 Tuesday, January 26, 2016. Posters that are not taken down by
17:00 on Tuesday will be taken down and discarded.
International Embryo Transfer Society
17
Poster Session Order by Topic
Poster number = abstract number in Reproduction, Fertility and Development 2016; 28 (1)
Student Competition
1
Generation of a stable transgenic swine model for cell tracking and chromosome dynamic studies
R. Sper, S. Simpson, X. Zhang, B. Collins, and J. Piedrahita
2
Circulating microRNA signatures associated with early bovine pregnancy
J. Ioannidis and X. Donadeu
3
Role of β-defensin 126 in promoting sperm motility in cattle
B. Fernandez-Fuertes, F. Narciandi, K. G. Meade, C. O’Farrelly, S. Fair, and P. Lonergan
4
Suppression of ASH2L alters DNA methylation and histone patterns during bovine embryonic
development
M. D. Snyder, J. H. Pryor, M. D. Peoples, G. L. Williamson, M. C. Golding, M. E. Westhusin, and
C. R. Long
5
6
A single nucleotide polymorphism in COQ9 affects mitochondrial function, body weight change after
calving, and fertility in Holstein cows
M. S. Ortega, S. Wohlgemuth, D. J. Null, J. B. Cole, and P. J. Hansen
The effects of depleting donor cell mitochondrial DNA on cattle embryos derived from somatic cell
nuclear transfer
K. Srirattana and J. C. St. John
Artificial Insemination
7
Kisspeptin slowly administered during proestrus improves follicle growth and ovulation in anestrous
cows
G. G. Macedo, R. V. Sala, L. F. Martins, and P. S. Baruselli
8
Response of communal cows to oestrus synchronization and timed artificial insemination
Z. C. Raphalalani, K. A. Nephawe, M. L. Mphaphathi, F. V. Ramukhithi, M. M. Seshoka,
M. Nkadimeng, A. Maqhashu, M. A. Bopape, L. F. Seolwana, M. H. Mapeka, N. L. Kanuya,
and T. L. Nedambale
9
Endoscopy-mediated intratubal insemination in the cow—A preliminary report about the application
of a novel minimally invasive insemination technique
K. Stein, V. Havlicek, S. Papp, F. Palm, G. Brem, and U. Besenfelder
10
11
12
18
Effect of estradiol benzoate or hCG on day 22 after timed artificial insemination on the corpus luteum
size, progesterone concentration, and pregnancy rate in resynchronized beef heifers
J. B. S. Borges, H. L. D. Neri, M. R. Almeida, E. P. Silva, and A. Bilha
Comparison of controlled internal drug-release insert–based and progesterone-free methods for
ovulation synchronization and timed artificial insemination of goats
A. Llanes, W. B. Knox, and C. E. Farin
Initial gonadotropin-releasing hormone treatment increased pregnancy per timed insemination only
in acyclic beef heifers subjected to a 5-day Co-synch protocol
I. López-Helguera, P. Whittaker, A. Behrouzi, and M. G. Colazo
42nd Annual Conference
13
Effect of 6 times reusing of controlled internal drug release (CIDR) for short term (6 days) on progesterone level and reproductive performance of Awassi ewes
A. Swelum, A. Mouamen, and A. Alowaimer
14
Epigenetic changes in equine spermatozoa during cryopreservation: A preliminary study
C. Aurich, B. Schreiner, N. Ille, M. Alvarenga, and D. Scarlet
Cloning and Nuclear Transfer
15
16
Histone acetylation profile of porcine embryos produced by 2 cloning methods with or without in
vitro culture
Y. Liu, A. Lucas-Hahn, B. Petersen, R. Li, D. Hermann, P. Hassel, M. Ziegler, J. Li, K. Larsen,
H. Niemann, and H. Callesen
Histone deacetylase inhibitors PCI-24781 and quisinostat improve the in vitro developmental
competence of pig somatic cell nuclear transfer embryos
L. Jin, H.-Y. Zhu, Q. Guo, Y.-C. Zhang, X.-C. Li, J.-D. Kang, and X.-J. Yin
Treatment with MGCD 0103 improves the in vitro development of porcine embryos derived from
somatic cell nuclear transfer
H.-Y. Zhu, L. Jin, Q. Guo, Y.-C. Zhang, X.-C. Li, J.-D. Kang, and X.-J. Yin
18
Embryo aggregation in pig improves cloning efficiency and embryo quality
C. Buemo, A. Gambini, L. Moro, R. F. Y. Martin, and D. Salamone
19
Direct introduction of gene constructs into the pronucleus-like structure of cloned embryos: A new
strategy for the generation of genetically modified pigs
M. Kurome, S. Leuchs, B. Kessler, E. Kemter, E. Jemiller, V. Zakhartchenko, and E. Wolf
20
Sialyltransferase gene expression in GGTA1 knockout pigs
G. A. Kim, J.-X. Jin, S. Lee, A. Oh, and B. C. Lee
21
Generation of GGTA1 and CMAH biallelic knockout porcine fibroblast cell lines by transcription
activator-like effector nuclease and clustered regularly interspaced short palindromic repeats
(CRISPR)/Cas9
J.-D. Kang, S.-M. Ryu, H.-Y. Zhu, L. Jin, W.-X. Li, C.-D. Cui, J.-S. Kim, and X.-J. Yin
17
22
23
24
25
26
Production of porcine transgenic cell line inserted with SV40LT, EGFRvIII gene, and inducible
CreERT system
S. U. Hwang, J. D. Yoon, K. Y. Eun, H. G. Kim, and S. H. Hyun
Reduction of mitochondrial function, proliferation, and gene expression in fibroblast donor cells for
use in somatic cell nuclear transfer by CPI-613 and PS48
B. R. Mordhorst, S. L. Murphy, L. D. Spate, R. M. Ross, K. D. Wells, J. A. Green, and R. S. Prather
Effect of donor cell type on in vitro and in vivo developmental competence of cloned buffalo
(Bubalus bubalis) embryos
N. L. Selokar, P. Sharma, D. Kumar, R. K. Sharma, and P. S. Yadav
Genome editing of somatic cell nuclear transfer derived zygotes by clustered regularly interspaced
short palindromic repeats (CRISPR)/Cas9 guide RNA injection
K. M. Whitworth, S. L. Murphy, J. A. Benne, L. D. Spate, E. Walters, R. Hickey, S. L. Nyberg,
K. D. Wells, and R. S. Prather
Pilot study: Yolk sac VEGF expression in bovine embryos from reproductive techniques
A. M. Mess, A. C. O. Carreira, C. Marinovic de Oliveira, P. Fratini, P. O. Favaron, R. S. N. Barreto,
F. V. Meirelles, and M. A. Miglino
International Embryo Transfer Society
19
27
An improved large animal model for the study of adult stem cells
S. Simpson, L. Gonzalez, J. Chung, A. Blikslager, S. Magness, and J. Piedrahita
28
Doubling oocyte cytoplasm volume increases blastocyst quality following interspecies somatic cell
nuclear transfer in Argali sheep (Ovis ammon)
A. L. Green, F. C. Oback, J. E. Oliver, L. Popovic, L. T. McGowan, S. J. Appleby, F. Meng,
D. L. Hyndman, D. Carson, and D. N. Wells
Cryopreservation and Cryobiology
29
30
The novel cryoprotective agent carboxylated ε-poly-l-lysine is effective for vitrification of
pre-implantation mouse embryos at the different stages
J. Ito, Y. Kawasaki, Y. Shibao, K. Matsumura, S.-H. Hyon, and N. Kashiwazaki
Effect of the repeated use of open system vitrification devices on mii stage and cleavage rates of
bovine cumulus–oocyte complexes
F. A. Diaz, E. J. Gutierrez, B. A. Foster, P. T. Hardin, and K. R. Bondioli
31
Intrafollicular transfer of fresh and vitrified immature bovine oocytes: An option for embryo production
J. F. W. Spricigo, S. B. S. Netto, C. V. Muterlle, S. A. D. Rodrigues, L. O. Leme, A. L. Guimaraes,
F. Caixeta, I. Pivato, and M. A. N. Dode
32
Effects of high hydrostatic pressure on expression profiles of in vitro-produced, vitrified bovine
blastocysts
Z. Jiang, P. Harrington, M. Zhang, S. Marjani, L. Kuo, C. Pribenszky, and X. Tian
33
34
35
36
Post-thaw viability of bovine embryos produced in vitro following treatment with ascorbic acid,
dithiothreitol, and caspase-3 inhibitor during cryopreservation
E. L. Carrascal-Triana, A. M. Zolini, A. Ruiz, J. M. Penitente-Filho, C. A. A. Torres, and J. Block
Effects of l-carnitine and trans-10,cis-12 conjugated linoleic acid supplementation during maturation
on development and cryotolerance of bovine embryos produced in vitro
J. Block, A. M. Zolini, E. Carrascal-Triana, A. Ruiz, P. J. Hansen, and C. A. A. Torres
Effects of thawing temperature of frozen semen on viability of refrozen and thawed Chickso (Korean
brindle cattle) and Korean albino cattle spermatozoa
S. W. Kim, C. Y. Choe, D. K. Kim, A. R. Choi, and H. H. Seong
Effect of cAMP modulators during oocyte in vitro maturation on gap junctional activity of vitrified
bovine oocytes
C. A. S. Monteiro, G. R. Leal, H. F. R. A. Saraiva, A. J. R. Camargo, P. M. S. Rosa, A. L. R. Rodrigues,
R. V. Serapião, L. A. G. Nogueira, L. S. A. Camargo, J. M. Garcia, and C. S. Oliveira
37
Double freezing and thawing of Nguni bull semen
M. L. Mphaphathi, M. M. Seshoka, T. R. Netshirovha, Z. C. Raphalalani, T. C. Chokoe,
M. Nkadimeng, N. L. Kanuya, J. P. C. Greyling, and T. L. Nedambale
38
Quail egg yolk in citrate extender is suitable for cryopreservation of Nguni bull semen
M. M. Seshoka, M. L. Mphaphathi, K. S. Mafolo, M. Nkadimeng, Z. C. Raphalalani, N. L. Kanuya,
and T. L. Nedambale
39
Fertility potential of frozen-thawed wood bison semen using extender without exogenous protein
S. X. Yang, G. P. Adams, J. M. Palomino, and M. Anzar
40
Cholesterol supplementation reduces cryocapacitation damages in buffalo (Bubalus bubalis) sperm
V. Longobardi, G. Albero, A. Salzano, G. Zullo, G. Bifulco, C. De Canditiis, and B. Gasparrini
20
42nd Annual Conference
41
42
Developmental potential of buffalo oocytes vitrified at the germinal vesicle stage: Effects of different
cryoprotectant combinations and cryodevices
A. S. El-Shalofy, A. R. Moawad, G. M. Darwish, S. T. Ismail, and A. B. Badawy
Diffusion of the cryoprotectants ethylene glycol and glycerol into day 9 to 11 equine embryos:
Preliminary results
T. Chenier, B. Foster, L. Gonzalez, and M. Schlaf
43
Mass vitrification of germinal-vesicle stage equine oocytes
H. S. Canesin, I. Ortiz, J. G. Brom-de-Luna, Y. H. Choi, and K. Hinrichs
44
Effect of cryoprotectant agents in equine ovarian biopsy fragments
G. D. A. Gastal, B. G. Alves, S. O. Paiva, K. A. Alves, S. G. S. de Tarso, G. Ishak, S. T. Bashir,
and E. L. Gastal
45
Acrosome reaction and heterologous zona binding assay of frozen stallion sperm after hyperactivation
M. A. Lagares, H. S. Martins, M. R. Souza, C. F. A. M. Penna, F. O. P. Leme, G. C. Silva, S. F. Cortes,
and R. Stahlberg
46
Vitrification at the germinal vesicle stage triggers precocious meiotic resumption but does not affect
cytoplasmic maturation in cumulus-enclosed porcine oocytes during in vitro maturation
T. Somfai, N. T. Men, H. Kaneko, J. Noguchi, S. Haraguchi, E. C. da Silva Santos, T. Nagai,
and K. Kikuchi
47
48
49
Effect of l-carnitine treatment during oocyte maturation on the post-thaw development of porcine
embryos vitrified at the pronuclear stage
C. G. Grupen, T. Somfai, and K. Kikuchi
Carboxylated ε-poly-l-lysine (COOH-PLL) improves developmental ability and reduces
mitochondrial damage of vitrified porcine embryos at the pronuclear stage
M. Kamoshita, K. Fujiwara, K. Matsumura, S.-H. Hyon, J. Ito, and N. Kashiwazaki
Effect of adding Trolox C and ascorbic acid to ram sperm before cryopreservation on the motility and
binding capability
J. Costa, W. Lima, E. Moraes, P. Sousa, L. Ramon, D. Lima, and V. Coelho
50
Estimation of chromatin abnormality of Ogye rooster semen with Diff-Quik staining
S. W. Kim, A. R. Choi, C. Y. Choe, D. K. Kim, H. H. Seong, and J. D. Kim
51
Optimization of vitrification parameters for rhesus macaque blastocysts
C. Ramsey, C. Hanna, and J. Hennebold
Developmental Biology
52
Use of transwell cell culture and 3-dimensional printing technology to develop an in vitro bovine oviduct
M. A. M. M. Ferraz, H. H. W. Henning, K. M. A. Van Dorenmalen, P. L. A. M. Vos, T. A. E. Stout,
P. F. Costa, J. Malda, and B. M. Gadella
53
Targeted screen for amino acids that regulate bovine inner cell mass development
V. Najafzadeh, R. Martinus, and B. Oback
54
Immunolocalization of steroid sulfatase and estrogen-specific sulfotransferase in bovine follicles
C. Blaschka, G. Schuler, and C. Wrenzycki
55
Effect of dimethyl sulfoxide supplementation on bovine in vitro embryo development
J. Stöhr, H. Grothmann, and C. Wrenzycki
International Embryo Transfer Society
21
56
The effects of serum level of androgens, luteinizing hormone, and insulin-like growth factor 1 in
early follicular phase on ovarian follicular growth parameters and pregnancy rate
Z. Raoofi, F. Hosseini, S. Pegah Parvar, and S. Paniz Parvar
57
Developmental characteristics of later-stage porcine embryos produced in vivo or in vitro
H. Callesen and P. Holm
58
Effect of lysophosphatidic acid on porcine oocyte in vitro maturation and subsequent embryonic
development after parthenogenetic activation and IVF
K.-J. Kim, E. Lee, and S.-H. Hyun
59
Lipid dynamics in the early embryonic development of zebrafish by desorption electrospray
ionization mass spectrometry imaging and nanoelectrospray mass spectrometry
A. K. Jarmusch, C. T. Mahapatra, V. Pirro, C. R. Ferreira, and M. S. Sepúlveda
60
Effects of AY9944 A-7 on meiotic resumption of porcine oocytes and cumulus cell expansion
S. Lee, C. Khoirinaya, J.-X. Jin, G. A. Kim, and B.-C. Lee
61
Unveiling the role of lipids in organogesis: Molecular anatomy by desorption electrospray ionization
mass spectrometry imaging mass spectrometry
V. Pirro, P. O. Favaron, C. R. Ferreira, L. S. Eberlin, R. S. Barreto, R. G. Cooks, and M. A. Miglino
62
Effect of progesterone on calcium regulation during differentiation of mouse embryonic stem cells
into cardiomyocytes
Y.-K. Choi, H. Y. Kang, J.-U. Hwang, T. D. Nam, and E.-B. Jeung
63
Equine follicles modulate cortisol levels and capability of oocytes to adapt to stress situations
D. Scarlet, N. Ille, G. D. A. Gastal, B. G. Alves, S. O. Paiva, M. O. Gastal, E. L. Gastal, and C. Aurich
64
Epithelial mesenchymal transition and differentiation of steroidogenic factor 1 mouse embryonic
stem cells into the steroidogenic cells
H. Y. Kang, Y.-K. Choi, J.-U. Hwang, and E.-B. Jeung
65
66
Effect of trichostatin A and 5-aza-2′-deoxycytidine treatment of donor cells, fused embryos, or both
on the developmental competence, quality, and epigenetic status of cloned buffalo (Bubalus bubalis)
embryos
M. Saini, N. L. Selokar, H. Agrawal, S. K. Singla, M. S. Chauhan, R. S. Manik, and P. Palta
Cumulus-oocyte-complex secretions from the first 8 hours of in vitro maturation affect oocyte
developmental competence
K. Uhde, H. T. A. van Tol, T. A. E. Stout, and B. A. J. Roelen
67
Inhibition of 5α-reductase during late gestation in the mare
M. Wynn, E. Legacki, A. Conley, S. Loux, A. Esteller-Vico, S. Stanley, E. Squires, M. Troedsson,
and B. Ball
68
Steroidogenic control of intrauterine sexual differentiation in Spix’s yellow-toothed cavy, Galea spixii
A. C. Santos, D. C. Viana, F. D. Oliveira, M. F. Oliveira, and A. C. Assis-Neto
69
Age at puberty and reproductive development of lambs fed sunflower cake
K. L. Da Silva, N. G. Alves, I. J. Ascari, D. R. Da Silva, J. P. A. Campos, D. M. Costa, A. C. Costa,
F. J. Barbosa, I. F. F. Garcia, and R. Ribeiro de Lima
70
Gonadal development in guinea pig males (Cavia porcellus)
F. Oliveira, A. Santos, and A. A. Neto
22
42nd Annual Conference
Early Pregnancy
71
Circulating microRNAs as potential biomarkers of early pregnancy in high-producing dairy cows
N. Fiandanese, A. Viglino, F. Strozzi, A. Stella, J. L. Williams, P. Lonergan, N. Forde, and
D. Iamartino
72
Preliminary data on the presence of bacteria in the uterus of pregnant cows
H. G. Pedersen, L. R. V. Knudsen, J. S. Agerholm, T. K. Jensen, K. S. Klitgaard, and
C. C. Karstrup
73
Proteomic analysis of uterine luminal fluid on day 7 of pregnancy in cattle
C. Passaro, N. Forde, T. E. Spencer, and P. Lonergan
74
Analysis of steroid hormones in bovine oviductal fluid by gas chromatography coupled with tandem
mass spectrometry
J. Lamy, P. Liere, P. Mermillod, and M. Saint-Dizier
75
Expression of growth factor genes in in vitro–produced blastocyst changes after uterine passage, but
endometrial expression is unaffected by the presence of embryos
M. Muñoz, S. Carrocera, D. Martin, N. Peynot, C. Giraud-Delville, E. Correia, O. Sandra,
V. Duranthon, and E. Gómez
76
Effects of reduced environmental light on the in vitro maturation of pig oocytes
L. Y. Parra-Forero, A. Góngora, S. Romo-García, E. P. López Damian, G. D. Mendoza,
J. A. Guevara, and A. García-Contreras
77
Use of seminal plasma to improve reproductive performance in alpacas (Vicugna pacos) following
natural mounting
W. Huanca, J. Turin, C. Mamani, R. Sanchez, W. F. Huanca, and T. Huanca
78
Interception of exosomal messages between the oviduct and the embryo: What are they tweeting
about?
C. Almiñana, E. Corbin, G. Harichaux, V. Labas, G. Tsikis, C. Soleilhavoup, K. Reynaud, X. Druart,
and P. Mermillod
Embryo Culture
79
Reactive oxygen species level in cultured porcine embryos after high hydrostatic pressure
M. Romek, M. Kucia, B. Gajda, and Z. Smorag
80
Reactive oxygen species level in pig embryos cultured in presence of hyaluronan
B. Gajda, M. Kucia, Z. Smorag, and M. Romek
81
Altered protein composition of porcine follicular fluid due to a high-fibre diet and the potential for
optimisation of in vitro culture media
S. Jarrett, A. C. Gill, D. Kurian, E. M. Ferguson, and C. J. Ashworth
82
Early porcine embryo energy preference and subsequent development
L. D. Spate, B. K. Redel, and R. S. Prather
83
In low oxygen culture, is hypotaurine necessary for in vitro development of porcine embryos?
J. A. Benne, L. D. Spate, B. M. Elliott, and R. S. Prather
84
Porcine embryos utilize small amounts of pyruvate, lactate, and glucose in vitro
B. R. Redel, L. D. Spate, B. Elliott, M. Paczkowski, R. L. Krisher, and R. S. Prather
International Embryo Transfer Society
23
85
Carboxyethyl germanium sesquioxide confers antioxidative protection for porcine in vitro–fertilized
embryos
E. Kim and S.-H. Hyun
86
Improved bovine embryo production using novel in vitro culture systems
J. H. Pryor, J. F. Hasler, L. Strøbech, B. Avery, N. Hashem, S. Menges, C. R. Long, G. Shewfelt,
and C. R. Looney
87
Coagulansin-A via heat shock protein 70 induction shows beneficial effects on the development of
bovine embryos in vitro
I. Khan, K.-L. Lee, A.-N. Ha, P.-R. Park, S.-H. Song, M. M. R. Chowdury, L. Xu, M.-D. Joo,
and I.-K. Kong
88
Enrichment of culture medium with crocetin improves in vitro embryo development in cattle
G. Zullo, J. E. Tamayo Palacio, C. De Canditiis, V. Longobardi, A. Salzano, G. Albero,
and B. Gasparrini
89
Effect of media, metabolic regulators, and stage of development on lipid content and mitochondrial
polarity of in vitro–produced Holstein embryos
M. A. Roberts, L. F. Campos-Chillon, M. Barceló-Fimbres, and J. L. Altermatt
Lipid content of in vivo– and in vitro–produced Jersey and Holstein cattle embryos and the effect of
forskolin on embryo lipid reduction
K. Rhodes-Long, L. F. Campos-Chillon, M. Barceló-Fimbres, and J. L. Altermatt
91
Embryo sugar consumption during in vitro development
M. Rubessa, A. Ambrosi, K. M. Polkoff, S. E. Denmark, and M. B. Wheeler
92
Insulin-like growth factor-1 protects bovine pre-implantation embryos produced in vitro from
anti-developmental actions of menadione
N. A. S. Rocha-Frigoni, B. C. S. Leão, P. C. Dall’Acqua, M. Ambrogi, and G. Z. Mingoti
90
93
Development of bovine pre-implantation embryos is altered by addition of activin and connective
tissue growth factor from days 5 to 7 after fertilization
J. Kannampuzha-Francis and P. J. Hansen
Effects of 6- or 12-hour culture in a micro q straw incubator on development of in vitro–produced
bovine embryos
C. R. Looney, J. H. Pryor, M. Snyder, A. Ilercil, and C. R. Long
95
Role of polyamines in bovine pre-implantation development
J. Herrick, A. Greene, W. Schoolcraft, and R. Krisher
96
Efficient production of monozygotic twin bovine embryos using blastomere separation technique
with commercial well of the well culture dish
Y. Hashiyada, Y. Aikawa, S. Sugimura, H. Matsuda, M. Ohtake, T. Yamanouchi, Y. Goto,
S. Kobayashi, and K. Imai
94
97
98
24
One-day protein-free culture selects for bovine blastocysts with improved long-term viability after
vitrification
E. Gomez, S. Carrocera, A. Murillo, V. Maillo, A. Gutiérrez-Adan, D. Martín, and M. Muñoz
Experimental transfer of bovine IVF-derived 32-cell stage embryos into the uterus: Environmental
effects on developmental characteristics
M. Hoelker, D. Salilew-Wondim, F. Rings, D. Tesfaye, and K. Schellander
42nd Annual Conference
99
Extracellular vesicles of bovine oviductal fluid modify the gene expression on bovine in vitro–
derived embryos
R. Lopera-Vasquez, M. Hamdi, V. Maillo, C. Nunez, M. Yanez-Mo, M. A. Ramirez, A. Gutierrez-Adan,
P. Bermejo-Alvarez, and D. Rizos
100
Sequential versus single-step medium for rhesus macaque embryo culture
C. Hanna, C. Ramsey, and J. Hennebold
101
Towards optimal in vitro culture conditions for pig-monkey aggregation chimeras
B. Burchardt, A. Lucas-Hahn, P. Hassel, M. Ziegler, G. Neuhaus, S. Wunderlich, S. Petkov, U. Martin,
and H. Niemann
102
Effects of linoleic acid/linolenic acid ratio on oocyte maturation and fertilization in prepubertal sheep
V. Ghaffarilaleh, R. Ghaffarilaleh, and G. Ghaffarilaleh
103
Ovarian stimulation of anestrous cats with follicle-stimulating hormone improves oocyte quality and
developmental capability after parthenogenetic activation
D. Veraguas, P. Gallegos, A. E. Velasquez, F. O. Castro, and L. Rodriguez-Alvarez
Embryo Manipulation
104
105
Embryo manipulation techniques alter pre-implantation development and gene expression in mouse
embryos
D. Jones, M. Paczkowski, and T. Kuehl
Bovine demi-blastocysts are able to expand to a size similar to intact blastocysts until at least day 13
of in vitro culture
A. E. Velasquez, J. Manríquez, D. Veraguas, F. O. Castro, and L. I. Rodríguez-Alvarez
Embryo Transfer
106
Comparison of in vitro embryo production in Panama using Holstein oocytes with Bos indicus sexed
semen from donors in different locations: Field data
S. J. R. Rodriguez, Y. E. Ramirez, E. Gomes Jr., L. F. Nasser, J. H. F. Pontes, M. Rubessa,
and M. B. Wheeler
107
Factors that influence fertility in an IVF embryo transfer program in dairy heifers
L. C. Carrenho-Sala, R. V. Sala, M. Fosado, D. C. Pereira, S. Garcia, A. Lopez, J. F. Moreno, A.
Garcia-Guerra, and M. C. Wiltbank
108
Factors affecting pregnancy rates in recipients receiving in vitro–produced embryos by fixed time
embryo transfer
M. Pelizzari, A. Tribulo, J. Garzon, B. Bernal, R. Tribulo, H. Tribulo, and G. A. Bó
109
Comparison of methods for synchronizing recipients of in vitro–produced embryos
R. V. Sala, L. C. Carrenho-Sala, M. Fosado, L. C. C. Tosta, R. D. Tosta, M. Stoll, J. F. Moreno,
B. M. Monteiro, P. S. Baruselli, A. Garcia-Guera, and M. C. Wiltbank
110
Treatment with GnRH on day 5 reduces pregnancy loss in heifers receiving in vitro–produced
expanded blastocysts
A. Garcia-Guerra, R. V. Sala, G. M. Baez, M. Fosado, L. F. Melo, J. C. L. Motta, L. Leffers,
E. A. Walleser, J. C. Ochoa, J. F. Moreno, and M. C. Wiltbank
International Embryo Transfer Society
25
Epidemiology/Diseases
111
Risk of Chlamydia abortus transmission via embryo transfer using in vitro early bovine embryos
F. Fieni, M. Oseikria, K. Laroucau, F. Vorimore, D. Tainturier, S. Destrumelle, and J. L. Pellerin
112
Presence of bacteria in the endometrium and oviduct of cows with pyometra as detected by
fluorescence in situ hybridization
C. C. Karstrup, L. Knudsen, T. K. Jensen, K. Schou, J. S. Agerholm, and H. G Pedersen
113
Expression of beta-oxidation-related genes in preeclampsia-like model under hypoxic condition
in vivo and in vitro
M. H. Lee, E.-K. Shin, H. Y. Kang, J.-U. Hwang, and E.-B. Jeung
114
Endoplasmic reticulum (er) stress in hypoxia-induced diabetes mellitus model
C. Ahn, D. Lee, K. P. Kim, M. H. Lee, and E.-B. Jeung
Exotic Species
115
Characterizing neutrophil profiles in horses for rhinoceros capture
R. A. Fayrer-Hosken, M. Kruger, M. Vandenplas, S. Giguere, and P. Buss
116
Factors impacting the success of postmortem sperm recovery and cryopreservation in the rhinoceros
T. L. Roth, M. A. Stoops, T. R. Robeck, and J. K. O’Brien
117
Development of a noninvasive, fecal protein pregnancy test for polar bears
C. DeLorenzo, B. Lynch, T. Roth, K. Petren, and E. Curry,
118
A rapid, minimally invasive method of collecting semen from polar bears
E. Curry and T. L. Roth,
119
Hormone-assisted reproduction in a captive mountain yellow-legged frog population
N. E. Calatayud, D. M. Shier, R. R. Swaisgood, and B. S. Durrant
120
Impact of season on seminal characteristics and sperm cryopreservation in the tufted deer (Elaphodus
cephalophus)
S. Panyaboriban, N. Songsasen, R. P. Singh, L. Padilla, J. Brown, D. Reed, M. Techakumphu,
and B. Pukazhenthi
121
Fixed-time laparoscopic oviductal artificial insemination (LO-AI) with frozen-thawed semen in
Pallas’ cats (Otocolobus manul)
W. F. Swanson, H. L. Bateman, J. R. Herrick, L. A. Lyons, and L. M. Vansandt
122
Timed artificial insemination in wood bison using frozen-thawed semen
G. P. Adams, S. X. Yang, J. M. Palomino, and M. Anzar
Folliculogenesis and Oogenesis
123
The mare model to study how ovarian dynamics affect preantral follicle features
M. O. Gastal, K. A. Alves, B. G. Alves, G. D. A. Gastal, S. G. S. de Tarso, J. R. Figueiredo,
M. L. Gambarini, and E. L. Gastal
124
Comparison of two embedding systems for follicular culture
K. M. Polkoff, M. Rubessa, R. Winters, N. A. Lopez, and M. B. Wheeler
125
Changes in intrafollicular concentrations of free insulin-like growth factor-1, activin-A, inhibin-A,
vascular endothelial growth factor, estradiol, and prl before ovulation in mares
S. T. Bashir, G. M. Ishak, M. O. Gastal, J. F. Roser, and E. L. Gastal
26
42nd Annual Conference
126
Effective method for in vitro culture of cryopreserved ovine ovarian tissue
A. Seisenbayeva, Y. Toishibekov, U. Iglmanov, B. Valiyeva, and B. Katubayeva
127
Thyroid stimulating hormone level change during controlled ovarian stimulation
Z. Raoofi and F. Razaghian
128
The role of endothelins in regulating bovine granulosa cells steroidogenesis
L. F. Schütz, J. E. Ervin, L. Zhang, C. Robinson, M. Totty, M. Albonico, F. Caloni, and L. J. Spicer
129
MicroRNA-183~96~182 cluster promote bovine granulosa cell proliferation through coordinated
regulation of FOXO1
S. Gebremedhn, D. Salilew-Wondim, M. Hoelker, F. Rings, C. Neuhoff, E. Tholen, C. Looft,
K. Schellander, and D. Tesfaye
130
Antral follicle counts, vulva width, and serum anti-Mullerian hormone concentrations in bovine
females of the Tabapuã breed
J. C. Souza, R. Maculan, R. G. Rosa, L. F. Smith, P. S. Casas, B. R. M. Faria, N. Alves,
and T. M. Gonçalves
131
Retrospective data analysis of Holstein heifer growth in embryo production centers
C. Baron, F. Charreaux, M. Rea, A. Morel, and H. Quinton
132
Blastocyst production from bovine ovarian cortex fragments xenografted under the back skin of mice
T. J. M. Alves, M. T. Dias, C. M. Assuncao, L. S. A. Camargo, J. H. M. Viana, and
P. H. A. Campos Junior
133
Antral follicle count, vulva width, and fertility traits in Bos taurus indicus cattle (Tabapuã)
R. Maculan, R. G. Rosa, R. R. Bonfim, G. L. Vasconcelos, T. L. C. Pinto, G. M. Moreira, J. A. S. Viafara,
T. M. Gonçalves, and J. C. Souza
134
Follicular dynamics of Gyr and Holstein oocyte donors kept under tropical conditions
L. P. Dias, P. M. S. Rosa, A. J. R. Camargo, R. V. Serapião, L. S. A. Camargo, and C. S. Oliveira
135
Short-term ovarian effects of unilateral ovariectomy in cows
R. Carrasco, J. Singh, and G. P. Adams
136
Comparison of NCSU-23 and alpha-minimal essential media in the development of isolated porcine
preantral follicles in vitro
M. Rubessa, R. Rocha, L. Lima, R. Winters, J. R. Figueiredo, and M. B. Wheeler
137
Dynamic shift in cytoplasmic lipids in cat oocytes during ovarian follicle development
C. Thongkittidilok, Y. Li, D. Wildt, and N. Songsasen
Gene Expression
138
Liver receptor homolog 1 influences blastocyst hatching in porcine
J. Guo, T. Kim, N.-H. Kim, and X.-S. Cui
139
Sex-specific gene expression in porcine pre-implantation embryos
D. Kradolfer, J. Knubben, V. Flöter, J. Bick, S. Bauersachs, and S. E. Ulbrich
140
Expression of perilipin in porcine oocytes and cumulus cells at different stages of in vitro maturation
A. Oh, J.-X. Jin, S. Lee, G. A. Kim, and B. C. Lee
141
Expression status of aquaporins 3, 7, and 9 in bovine pre-implantation embryos
T. Fujii, H. Hirayama, S. Kageyama, A. Naito, S. Fukuda, S. Moriyasu, and K. Sawai
International Embryo Transfer Society
27
142
MicroRNA-17-92 cluster regulates bovine granulosa cell function by targeting BMPR2 and PTEN genes
E. Andreas, D. Salilew-Wondim, M. Hoelker, C. Neuhoff, E. Tholen, C. Looft, K. Schellander,
and D. Tesfaye
143
Genomic assessment associations of the paternal effect for daughter fertility traits in Holstein cattle
A. A. Sermyagin, E. A. Gladyr, O. S. Romanenkova, A. N. Ermilov, G. Brem, and N. A. Zinovieva
144
Dosage compensation and X-linked gene expression in bovine in vivo and in vitro embryos
J. E. Duan, N. K. Jue, Z. Jiang, R. O’Neill, E. Wolf, L. A. Blomberg, H. Dong, X. Zheng, J. Chen, and
X. C. Tian
145
Oviductal expression of genes involved in growth factor, cytokine, hormone, and WNT signaling
during the early estrous cycle of the cow
P. Tribulo, L. G. Siqueira, and P. J. Hansen
146
Sperm storage: Expression of progesterone receptors, structural proteins, and heat shock proteins in
the avian oviduct
C. Riou, M. Saint-Dizier, and N. Gerard
147
Effect of ovulatory follicle diameter on the oocyte transcriptome in beef cows
S. E. Dickinson, J. A. Green, T. W. Geary, K. G. Pohler, G. A. Bridges, and M. F. Smith
148
Follicular fluid microRNA sequences as biomarkers of competent oocytes in cattle
R. Pasquariello, N. Fiandanese, A. Viglino, P. Pocar, J. L. Williams, and F. Gandolfi
149
Thyroid hormone marker deiodinase 1 induced by 2,4,6-tribromophenol in GH3 pituitary cells
D. Lee, J.-U. Hwang, H. Y. Kang, and E.-B. Jeung
150
In vitro model for effect of endocrine-disrupting chemicals on placental cation transporter channel
J.-H. Lee, M. H. Lee, M. J. Lee, and E.-B. Jeung
151
Comparative analysis of fresh and cryopreserved boar spermatozoa using RNA sequencing
J. Feugang, S. Liao, W. Sanders, J. Lu, M. Crenshaw, S. Willard, and P. Ryan
152
Proliferation- and senescence-related gene expression in canine fibroblasts derived from dogs of
different age
Y. B. Choi, G. A. Kim, H. J. Oh, M. J. Kim, Y. K. Jo, E. M. N. Setyawan, S. H. Lee, and B. C. Lee
153
Different molecular mechanisms for histone deacetylase inhibitor-induced apoptosis in dog
fibroblasts and mesenchymal stem cells
M. J. Kim, H. J. Oh, G. A. Kim, Y. K. Jo, Y. B. Choi, E. M. N. Setyawan, S. H. Lee, and B. C. Lee
154
Molecular characterization of cat primordial germ cells
J. Galiguis, C. E. Pope, C. Dumas, G. Wang, R. A. MacLean, and M. C. Gómez
155
Comparison of circulating microRNAs between pregnant and nonpregnant mares
S. C. Loux, K. E. Scoggin, J. E. Bruemmer, I. F. Canisso, M. H. Troedsson, E. L. Squires, and B. A. Ball
156
Effect of 2-methoxyestradiol (2-ME) on human uterine leiomyosarcoma
K. P. Kim, J.-S. Lee, C. Ahn, Y.-K. Choi, and E.-B. Jeung
IVF and IVP
157
28
Effect of melatonin on embryo quality of bovine oocytes subjected to heat shock
N. G. Alves, I. J. Ascari, L. S. A. Camargo, J. Jasmin, C. C. R. Quintão, J. A. S. Oliveira, E. D. Souza,
C. M. A. Assunção, V. R. A. Mendes, and R. R. Lima
42nd Annual Conference
158
Impact of matured cattle oocytes at higher incubation temperature on in vitro embryo production
M. Nkadimeng, E. van Marle-Koster, K .P. M. Lekola, M. L. Mphaphathi, M. M. Seshoka,
Z. C. Raphalalani, N. L. Kanuya, and T. L. Nedambale
159
In vitro embryo production from Holstein calf oocytes recovered by laparoscopic ovum pickup
H. Baldassarre, L. Currin, L. Michalovic, W. Glanzner, K. Gutierrez, R. Bohrer, P. da Rosa,
M. De Cesaro, N. Dicks, Y. Schuermann, and V. Bordignon
160
Lipidome signatures in early bovine embryo development
M. J. Sudano, T. D. S. Rascado, A. Tata, K. R. A. Belaz, V. G. Santos, R. S. Valente, F. S. Mesquita,
C. R. Ferreira, J. P. Araujo, M. N. Eberlin, and F. D. C. Landim-Alvarenga
161
Effect of oviductal fluid on bovine oocyte zona pellucida hardening and sperm binding, and on early
embryo development
P. Hugon, J. Lamy, E. Corbin, P. Mermillod, and M. Saint-Dizier
162
Effects of prolactin and dithiothreitol on the quality and developmental capacity of in vitro–matured
bovine oocytes
G. Singina, I. Lebedeva, E. Shedova, and N. Zinovieva
163
Effects of serum and l-carnitine on development and cryotolerance of bovine embryos produced in vitro
A. Zolini, E. L. Carrascal-Triana, A. Ruiz, J. M. Penitente-Filho, P. J. Hansen, C. A. Alves Torres,
and J. Block
164
Effects of kinetics and morphology on early embryonic development in bovine OPU-IVF embryos
M. Takayama, O. Dochi, and K. Imai
165
Cryopreservation of in vitro produced bovine embryos after lipid decrease with forskolin
D. Paschoal, M. Sudano, R. Maziero, M. Guastali, L. Magalhães, F. Landim-Alvarenga, A. Martins Jr.,
and C. Leal
166
The effect of temperature during storage of in vitro–matured bovine oocytes in a HEPES-buffered
medium on developmental competence
T. Suttirojpattana, T. Somfai, S. Matoba, T. Nagai, R. Parnpai, and M. Geshi
167
168
169
170
171
Effect of DNA methylation inhibitor on heterochromatin in bovine embryos derived from heatshocked oocytes
T. D. Araujo, J. Jasmin, C. C. R. Quintao, E. D. Souza, J. H. M. Viana, and L. S. A. Camargo
Effect of cyclic adenosine monophosphate modulators during oocyte in vitro maturation on bovine
embryos (Gyr × Holstein) quality
G. R. Leal, C. A. S. Monteiro, H. F. R. A. Saraiva, A. J. R. Camargo, P. M. S. Rosa, A. L. R. Rodrigues,
C. O. P. Vasconcelos, L. A. G. Nogueira, C. S. Oliveira, and R. V. Serapião
Ovum pick-up, in vitro embryo production, and embryo transfer in cattle in tropical and subtropical
Mexico
S. Castañeda, S. Romo, and M. E. Kjelland
In vitro embryo production and apoptosis detection using bovine oocytes matured with IGF-I or
IGF-LongR3
P. L. M. De Chico, E. C. S. Recalde, T. L. Ikeda, M. J. Sudano, and F. Landim-Alvarenga
Normal versus low anti-Müllerian hormone in patients with advanced maternal age: In vitro fertilization outcomes and morphokinetic parameters
A. Kotlyar, R. Flyckt, and N. Desai
International Embryo Transfer Society
29
172
Antiviral action of propolis aqueous extract during maturation of infected bovine oocytes
E. G. Palazzi, D. Hansen, M. F. Alves, A. H. C. Nogueira, R. A. Ogata, J. G. Bersano, L. P. Pacheco,
E. de Stefano, L. H. Okuda, R. S. Jordão, and E. M. Pituco
173
In vitro embryo production from oocytes fertilised with unsorted or X-sorted sperm and issued
from subfertile high genetic merit cows submitted or not to a 48-h coasting period following FSH
stimulation
G. Gamarra, S. Lacaze, C. Ponsart, M. Mouneyres, and B. Le Guienne
174
Development and in vitro production of prepubertal Nelore heifers long-acting progesterone exposed
R. Corrêa, J. R. Maio, and J. Garcia
175
Consequences of in vitro production of embryos with or without colony-stimulating factor 2 in
culture medium on morphometric features of the bovine conceptus at day 86 of gestation
L. G. B. Siqueira, P. Tribulo, A. C. Denicol, M. S. Ortega, V. M. Negrón-Pérez,
J. Kannampuzha-Francis, and P. J. Hansen
176
Serum anti-Müllerian hormone concentrations and in vitro embryo production in beef cattle
G. A. Bó, I. Anduaga, B. Bernal, G. Moreira, A. Cedeño, J. Barajas, M. Pelizzari, S. Andrada,
and A. Tribulo
177
Performance of Gyr prepubertal heifers in in vitro embryo production
P. M. S. Rosa, A. J. R. Camargo, R. V. Serapião, L. S. A. Camargo, and C. S. Oliveira
178
First commercial cattle in vitro embryo production and pregnancy rates of both fresh and frozen in
vitro embryos in the north coast of Peru
H. W. Vivanco-Mackie, R. D. Navarro, M. D. P. Salazar, E. A. Aguirre, G. B. Saldaña, and
I. M. Lizaraso
179
180
181
182
Relationship between gene expression in individual blastomeres of 2-cell stage bovine embryos and
the normality of first cleavage
S. Matoba, M. Kaneda, T. Somfai, T. Nagai, and M. Geshi
Assessment of bull semen quality loaded in new Sensitemp straws using semen and in vitro
production technologies
D. Le Bourhis, S. Camugli, P. Salvetti, L. Schibler, and E. Schmitt
In vitro fertilization (IVF) using semi-defined culture conditions from low or high antral follicle
count pubertal beef heifers
C. C. Chase Jr., R. A. Cushman, A. K. McNeel, O. L. Amundson, G. A. Perry, S. C. Tenley, J. R. Wood,
A. S. Cupp, J. L. Vallet, and J. R. Miles
Effects of IGF-1 or IGF-1 LongR3 on cellular and molecular aspects of cumulus–oocyte complexes
during in vitro oocyte maturation in cattle
M. S. Araujo, M. D. Guastali, A. C. S. Castilho, and F. Landim-Alvarenga
183
In vitro embryo production: A tool to preserve the threatened wood bison (Bison bison athabascae)
M. P. Cervantes, J. M. Palomino, M. Anzar, R. J. Mapletoft, G. Mastromonaco, and G. P. Adams
184
Preliminary findings on carbohydrate metabolism of intact equine cumulus–oocyte complexes during
in vitro maturation
N. Lewis, K. Hinrichs, D. Brison, R. Sturmey, D. Grove-White, K. Schnauffer, and
C. McGregor-Argo
30
42nd Annual Conference
Male Physiology
185
186
187
Transient receptor potential channel-2 (TRPP2) regulates motility and intracellular calcium of
porcine sperm
B. W. Daigneault and D. J. Miller
Quantitative echogenicity and echotexture analysis of the accessory sex glands of pubertal and
mature Dorper rams
E. S. C. Camela, R. P. Nociti, V. J. C. Santos, B. I. Macente, G. S. Maciel, M. A. R. Feliciano,
W. R. R. Vicente, P. M. Bartlewski, and M. E. F. Oliveira
Exploitation of in vitro capacitation for nanoparticle incorporation within mammalian spermatozoa
L. Myles, C. Durfey, P. Ryan, S. Willard, and J. Feugang
Oocyte Activation
188
Haploid activation of bovine oocytes with ionomycin and single or combined activating agents
M. Suvá, N. G. Canel, and D. F. Salamone
189
Characterization of the first sperm-induced calcium transient in pig oocytes
C. Wang and Z. Machaty
190
Spindlin 1 is required for metaphase II arrest in porcine oocytes
J.-W. Choi, T. Kim, N.-H. Kim, and X.-S. Cui
Oocyte Maturation
191
Searching for the in vivo transcriptome blueprint of competent bovine oocytes
V. Bocchi, M. G. Stillacci, A. Zecconi, C. Galli, G. Stadaioli, T. A. L. Brevini, A. Bagnato,
and F. Gandolfi
192
Prematuration of bovine cumulus–oocyte complexes with cyclic adenosine monophosphate
modulators affects both oocyte and blastocyst ultrastructure
E. Razza, H. Pedersen, L. Stroebech, M. Machado, M. Nogueira, H. Kadarmideen, H. Callesen,
and P. Hyttel
193
Improved post-thaw survival of bovine embryos produced in serum-free in vitro production system
M. Nõmm, E. Mark, O. Sarv, S. Kõks, and Ü. Jaakma
194
Effect of corpus luteum presence for the duration of follicular growth on bovine oocyte developmental
competence
T. Yamanouchi, H. Matsuda, M. Ohtake, Y. Aikawa, Y. Goto, S. Kobayashi, and Y. Hashiyada
195
Heat stress alters the transcriptome of maturing bovine oocytes
L. A. Rispoli, R. R. Payton, C. Gondro, A. M. Saxton, and J. L. Edwards
196
Elevated temperature during bovine oocyte maturation alters mitochondrial activity
R. R. Payton, L. A. Rispoli, K. A. Nagle, A. M. Saxton, and J. L. Edwards
197
Effects of pre-in vitro maturation with caffeine on bovine oocyte developmental capacity
S. M. B. Ulloa, J. Heinzmann, D. Herrmann, U. Baulain, K.-G. Hadeler, P. Aldag, A. Lucas-Hahn,
and H. Niemann
198
Resveratrol supplementation during in vitro maturation and fertilisation enhances developmental
competence of bovine oocytes
P. Kordowitzki, S. M. Bernal, D. Herrmann, P. Aldag, and H. Niemann
International Embryo Transfer Society
31
199
200
201
Effect of urokinase type plasminogen activator during in vitro maturation of bovine oocytes and early
embryo production
M. Roldán-Olarte, V. Maillo, M. J. Sánchez-Calabuig, A. Tío-Castro, P. Beltrán, D. C. Miceli,
D. Rizos, and A. Gutiérrez-Adán
Effects of melatonin on reactive oxygen species generation and acquisition of embryonic developmental
competence of bovine oocytes matured in vitro
P. C. Dall’Acqua, B. C. S. Leão, N. A. S. Rocha-Frigoni, M. Ambrogi, and G. Z. Mingoti
Role of the nitric oxide system in effects of prolactin and growth hormone on metaphase-II
chromosomes in bovine oocytes aging in vitro
I. Lebedeva, G. Singina, E. Shedova, A. Lopukhov, and N. Zinovieva
202
Characterization of bovine oocyte cytoplasmic maturation with common in vitro maturation protocols
B. A. Foster, F. A. Diaz, P. T. Hardin, E. J. Gutierrez, and K. R. Bondioli
203
Supplementation with linolenic acid, l-carnitine, alone or associated, during IVM resulted in
decrease of ros levels and apoptotic index of bovine in vitro produced embryos
B. C. S. Leão, N. A. S. R. Frigoni, P. C. Dall’Acqua, M. Ambrogi, and G. Z. Mingoti
204
205
206
207
Inadequacy of vascular endothelial growth factor in culture medium reduces the viability of cumulus
cells and prevents in vitro maturation of porcine oocytes
T. T. M. Bui, P. P. Ferré, M. T. Tran, T. Wakai, and H. Funahashi
Holding pig oocytes at 24°C prior to in vitro maturation alters the developmental capacity after in
vitro fertilisation but not parthenogenetic activation
C. Quadalti, I. Lagutina, G. Lazzari, and C. Galli
Effect of addition follicular fluid or growth differentiation factor-9 on in vitro maturation of porcine
oocytes denuded 20 h after the start of in vitro maturation
P. Ferré, T. T. M. Bui, M. T. Tran, T. Wakai, and H. Funahashi
Ganglioside improves the meiotic maturation and anti-apoptotic effects during in vitro maturation of
porcine oocytes
J.-W. Kim, H.-J. Park, S.-K. Chae, J.-H. Ahn, G.-Y. Do, J.-Y. Park, S.-G. Yang, and D.-B. Koo
Effect of GDF8 and SB431542 on porcine oocyte during in vitro maturation and subsequent
embryonic development
J. D. Yoon, E. Lee, and S.-H. Hyun
209
Maturation kinetics after holding equine oocytes in embryo holding medium
P. Dini, O. Bogado, K. Smits, A. VanSoom, and P. Daels
210
Post-translational modification of histone h4 acetylation during in vitro maturation of canine oocytes
T. F. Motheo, D. R. Arnold, W. R. R. Vicente, B. I. Macente, F. G. F. Filgueira, D. G. Chung,
V. T. Almeida, R. A. R. Uscategui, and F. L. Lopes
211
Production of progesterone from canine cumulus cells matured in vitro with steroids and
gonadotropin
M. Apparicio, G. J. Covizzi, A. E. Alves, E. A. Pires-Butler, T. F. Motheo, B. I. Macente,
G. P. Nogueira, and W. R. R. Vicente
208
212
32
Effect of canine oviduct cells and cumulus cells co-culture on in vitro maturation of porcine oocytes
and embryo development
S. H. Lee, H. J. Oh, G. A. Kim, M. J. Kim, Y. B. Choi, Y. Kwang Jo, E. Nugraha Setyawan, and B. C. Lee
42nd Annual Conference
213
Maturation of oocytes with follicular fluid from gilts consuming high fat and fructose affects
subsequent embryo development
R. Poole, V. McCracken, M. Rhoads, and K. Lee
Sexing
214
Pre-implantation embryo sex determination by amplification of amelogenin gene in water buffaloes
(Bubalus bubalis)
J.-H. Shang, C.-Y. Yang, H.-Y. Zheng, J. Qin, Y.-P. Gu, and H.-Z. Liu
215
Production of progeny with sexed white Hanwoo (Korean native albino cattle) semen
J. S. Choi, C. Y. Choe, D. K. Kim, H. H. Seong, and S. W. Kim
Sperm Injection
216
217
218
219
220
Effect of sperm pretreatment with lysolecithin and Triton X-100 on pronuclear formation and quality
of bovine embryos produced by intracytoplasmic sperm injection
F. Zambrano, M. E. Arias, T. Vargas, L. Aguila, and R. Felmer
Improvement of intracytoplasmic sperm injection embryo development in bovine using high
cysteamine concentration during in vitro maturation and sperm co-culture with cumulus–oocyte
complexes
N. G. Canel, M. Suvá, R. J. Bevacqua, and D. F. Salamone
An evaluation of in vitro maturation oocyte activation methods for improving ovine intracytoplasmic
sperm injection efficiency
J. E. Hernández, Y. Ducolomb, S. Romo, R. Fierro, M. E. Kjelland, and M. Betancourt
Sorting of equine sperm using a microfluidic device as a method of sperm selection for in vitro fertilization and intracytoplasmic sperm injection
R. Gonzalez and E. Carnevale
Effect of zinc supplementation during in vitro maturation on equine blastocyst rates after intracytoplasmic sperm injection
Y. H. Choi, J. R. Gibbons, H. S. Canesin, and K. Hinrichs
Stem Cells
221
Derivation of insulin-producing cells from human dermal fibroblasts at intermediate phase of
reprogramming
J. H. Kang, S. M. Park, S. Y. Heo, J. Kang, S. Y. Lee, K. S. Ahn, S. G. Lee, and H. Shim
222
Incomplete reprogramming of induced pluripotent stem cells derived from porcine fetal fibroblasts
K.-H. Choi, D. Son, D.-K. Lee, J.-N. Oh, S.-H. Kim, T.-Y. Park, and C.-K. Lee
223
Generation of interspecies chimeras between primate induced pluripotent stem cells and porcine
parthenogenetic embryos
M. Nowak-Imialek, S. Wunderlich, D. Herrmann, S. Klein, U. Baulain, A. Lucas-Hahn, S. Petkov,
E. Mall, B. Petersen, U. Martin, and H. Niemann
224
Characterization of equine endometrial-derived mesenchymal stromal cells
E. Rink, H. French, E. Watson, C. Aurich, and F. X. Donadeu
225
Autologous transplantation of mesenchymal stem cells derived from adipose tissue in animal
H. Malik, V. Sharma, S. Saini, S. Guha, and D. Malakar
International Embryo Transfer Society
33
226
Effects of microvesicles secreted from equine amniotic-derived progenitor cells on in vitro lipopolysaccharide-treated tendon and endometrial cells
A. Lange-Consiglio, C. Perrini, P. Esposti, M. C. Deregibus, G. Camussi, L. Pascucci, M. G. Marini,
B. Corradetti, D. Bizzaro, and F. Cremonesi
227
Mesenchymal stem cell isolation and culture from adipose tissue of a dead dog
S. Saini, V. Sharma, H. N. Malik, S. K. Guha, and D. Malakar
228
WNT signalling pathway activation in cat embryonic stem like-cells and its role in maintaining
pluripotency
M. Duque, E. S. Zanneti, J. H. Galiguis, C. E. Pope, and M. C. Gomez
229
Long-term propagation and cryopreservation of cat spermatogonial stem cells
L. M. Vansandt, M. Dickson, R. Zhou, L. Li, B. S. Pukazhenthi, and C. L. Keefer
230
Effect of different concentrations of glial cell line-derived neurotrophic factor on expression of
self-renewal related genes in goat (Capra hircus) spermatogonial stem cells
A. Sharma, S. M. Shah, N. Saini, M. K. Singh, S. K. Singla, P. Palta, R. S. Manik, and M. S. Chauhan
231
Use of soft substrates to promote and maintain OCT4 expression in epigenetically erased fibroblasts
G. Pennarossa, R. Santoro, M. Pesce, F. Gandolfi, and T. A. L. Brevini
Superovulation
232
233
234
235
236
237
Effect of superovulation pretreatment on developmental characteristics of in vitro–fertilized bovine
embryos transferred to the oviduct-uterus environment
V. Havlicek, A. Gad, S. Papp, K. Stein, F. Palm, D. Tesfaye, M. Hoelker, and U. Besenfelder
Anti-Müllerian hormone as a predictive endocrine marker for superovulatory response and embryo
production in beef cattle
K. Center, D. Dixon, and R. Rorie
Hormonal follicle stimulation in Holstein cows for in vitro embryo production using sperm sorted by
flow cytometry
L. Ferré, Y. Bogliotti, J. Chitwood, M. Kjelland, and P. Ross
Administration of levothyroxine improves the proportion of transferable embryos following
superovulation in Japanese Black cattle
N. Tominaga, M. Tomomatsu, M. Nakamura, A. Ideta, M. Konishi, and Y. Aoyagi
Superovulation in sheep with reduced injection FSH/eCG protocols followed by transfer of fresh or
vitrified embryos
R. C. Fry, R. Mapletroft, G. A. Bó, M. M. Izzo, and M. A. Humphris
Effect of a PTEN inhibitor on ovarian contents of apoptosis-related proteins and anti-Müllerian
hormone (AMH) in A/J mice
O. Suzuki
Transgenesis
238
Production of cleavage-resistant phytase transgenic pigs by handmade cloning
M. Zhang, S. Chen, X. Chen, Y. Huang, L. Wei, B. Chen, Y. Wu, Q. Jiang, Y. Guo, Y. Wei, X. Lin,
G. Lan, and H. Jiang
34
42nd Annual Conference
239
240
241
MAC-T cells as a tool to examine genome editing using clustered regularly interspaced short
palindromic repeats (CRISPR)
S. N. Lotti, I. Tasan, H. Zhao, and M. B. Wheeler
Precise genome editing of PDX1 by direct injection of transcription activator-like effector nucleases
(TALENS) into parthenogenetic pig embryos
C. de Frutos, D. Webster, S. C. Fahrenkrug, and D. F. Carlson
Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 associated with transient
depletion of non-homologous end-joining pathway increased gene-targeting efficiency in goat
fibroblasts
K. C. S. Tavares, C. R. Lazzarotto, S. G. Neto, L. T. Martins, L. H. Aguiar, C. E. M. Calderón,
L. P. R. Teixeira, F. E. M. Lopes, M. B. Wheeler, C. R. Long, B. Whitelaw, M. Bertolini, and
L. R. Bertolini
242
Highly efficient Sleeping Beauty transposon-mediated transgenesis in bovine fetal fibroblasts
A. E. Fili, A. P. Alessio, W. Garrels, D. O. Forcato, M. F. Olmos Nicotra, A. C. Liaudat,
R. J. Bevacqua, V. Savy, M. I. Hiriart, N. Rodriguez, T. R. Talluri, Z. Ivics, D. F. Salamone,
W. A. Kues, P. Bosch
243
Efficient edition of the bovine PRNP prion gene in somatic cells and IVF embryos using the clustered
regularly interspaced short palindromic repeats (CRISPR)/Cas9 system
R. J. Bevacqua, R. Fernandez-Martín, V. Savy, N. G. Canel, M. I. Gismondi, W. Kues, D. F. Carlson,
S. C. Fahrenkrug, H. Niemann, O. A. Taboga, S. Ferraris, and D. F. Salamone
Undergraduate Poster Competition
244
245
Chondrogenic pellet cultures for cartilage tissue engineering grow by deposition of matrix and not by
cellular proliferation
S. Johnson, D. Milner, H. Lopez-Lake, and M. Wheeler
Risk of transmission of bovine leukosis virus (BLV) using seropositive bulls for in vitro fertilization
embryo production
J. Stewart, M. Rubessa, K. Polkoff, S. Lotti, and M. Wheeler
International Embryo Transfer Society
35
Oral Session Order by Topic
Donor Selection and Treatments to Improve Oocyte Collection and In Vitro Embryo
Production in Cattle
246
Influence of breed and season on in vitro embryo production
B. Bernal, J. Revol, J. M. Oviedo, A. Tribulo, H. Tribulo, G. Moreria, and G. A. Bó
247
The importance of follicular differentiation to obtain fully competent bovine oocytes
M. A. Sirard
248
Superstimulation strategies for ovum pickup in Holstein donors
L. M. Vieira, G. A. Bó, and R. J. Mapletoft
249
Optimizing donor efficiency with follicle stimulating hormone superstimulation in a commercial in
vitro fertilization program in Holsteins
F. X. Grand, C. Vigneault, and P. Blondin
Equine Reproduction: Pre- and Post-Ovulatory Influences on Fertility
250
Anti-Müllerian hormone: Implications for follicular reserve, follicular function, and fertility in the
mare
B. A. Ball, A. Claes, T. E. Curry Jr., M. H. T. Troedsson, E. L. Squires, and K. E. Scoggin
251
Factors influencing the success of equine intracytoplasmic sperm injection in a clinical program
K. Hinrichs and Y. H. Choi
252
Effects of low circulating progesterone concentrations during early diestrus on the endometrial
transcriptome of the mare
A. Esteller-Vico, J. N. Macleod, D. E. Graugnard, K. E. Scoggin, E. L. Squires, M. H. Troedsson,
and B. A. Ball
253
The mare model for luteinized unruptured follicle syndrome: Intrafollicular endocrine milieu
E. L. Gastal, S. T. Bashir, M. O. Gastal, S. G. S. de Tarso, S. P. Tazawa, J. Cuervo-Arango,
A. R. Baerwald, and D. B. Hales
From Bench to Barn: Effects of Bull Fertility or Sperm Factors on Embryo
Development and Cattle Agriculture
254
Systems biology of sperm and bull fertility
E. Memili
255
Seminal plasma proteins: Functional attributes and potential markers of fertility
A. A. Moura
256
Genomic evaluation of fertility traits and discovery of haplotypes that affect fertility of US dairy cattle
G. R. Wiggans, D. J. Null, J. B. Cole, and H. D. Norman
36
42nd Annual Conference
International Embryo Transfer Society
37
Author Index
Author, Poster = abstract number in Reproduction, Fertility and Development 2015; 27(1)
Adams, G. P., 39, 122, 135, 183
Agerholm, J. S., 72, 112
Agrawal, H., 65
Aguiar, L. H., 241
Aguila, L., 216
Aguirre, E. A., 178
Ahn, C., 114, 156
Ahn, J.-H., 207
Ahn, K. S., 221
Aikawa, Y., 96, 194
Albero, G., 40, 88
Albonico, M., 128
Aldag, P., 197, 198
Alessio, A. P., 242
Almeida, M. R., 10
Almeida, V. T., 210
Almiñana, C., 78
Alowaimer, A., 13
Altermatt, J. L., 89, 90
Alvarenga, M., 14
Alves, A. E., 211
Alves, B. G., 44, 63, 123
Alves, K. A., 44, 123
Alves, M. F., 172
Alves, N., 130
Alves, N. G., 69, 157
Alves, T. J. M., 132
Ambrogi, M., 92, 200, 203
Ambrosi, A., 91
Amundson, O. L., 181
Andrada, S., 176
Andreas, E., 142
Anduaga, I., 176
Anzar, M., 39, 122, 183
Aoyagi, Y., 235
Apparicio, M., 211
Appleby, S. J., 28
Araujo, J. P., 160
Araujo, M. S., 182
Araujo, T. D., 167
Arias, M. E., 216
Arnold, D. R., 210
Ascari, I. J., 69, 157
Ashworth, C. J., 81
Assis-Neto, A. C., 68
Assuncao, C. M., 132
38
Assunção, C. M. A., 157
Aurich, C., 14, 63, 224
Avery, B., 86
Badawy, A. B., 41
Baerwald, A. R., 253
Baez, G. M., 110
Bagnato, A., 191
Baldassarre, H., 159
Ball, B., 67
Ball, B. A., 155, 250, 252
Barajas, J., 176
Barbosa, F. J., 69
Barceló-Fimbres, M., 89, 90
Baron, C., 131
Barreto, R. S., 61
Barreto, R. S. N., 26
Bartlewski, P. M., 186
Baruselli, P. S., 7, 109
Bashir, S. T., 44, 125, 253
Bateman, H. L., 121
Bauersachs, S., 139
Baulain, U., 197, 223
Behrouzi, A., 12
Belaz, K. R. A., 160
Beltrán, P., 199
Benne, J. A., 25, 83
Bermejo-Alvarez, P., 99
Bernal, B., 108, 176, 246
Bernal, S. M., 198
Bersano, J. G., 172
Bertolini, L. R., 241
Bertolini, M., 241
Besenfelder, U., 9, 232
Betancourt, M., 218
Bevacqua, R. J., 217, 242, 243
Bick, J., 139
Bifulco, G., 40
Bilha, A., 10
Bizzaro, D., 226
Blaschka, C., 54
Blikslager, A., 27
Block, J., 33, 34, 163
Blomberg, L. A., 144
Blondin, P., 249
Bó, G. A., 108, 176, 236, 246, 248
Bocchi, V., 191
42nd Annual Conference
Bogado, O., 209
Bogliotti, Y., 234
Bohrer, R., 159
Bondioli, K. R., 30, 202
Bonfim, R. R., 133
Bopape, M. A., 8
Bordignon, V., 159
Borges, J. B. S., 10
Bosch, P., 242
Brem, G., 9, 143
Brevini, T. A. L., 191, 231
Bridges, G. A., 147
Brison, D., 184
Brom-de-Luna, J. G., 43
Brown, J., 120
Bruemmer, J. E., 155
Buemo, C., 18
Bui, T. T. M., 204, 206
Burchardt, B., 101
Buss, P., 115
Caixeta, F., 31
Calatayud, N. E., 119
Calderón, C. E. M., 241
Callesen, H., 15, 57, 192
Caloni, F., 128
Camargo, A. J. R., 36, 134, 168, 177
Camargo, L. S. A., 36, 132, 134, 157,
167, 177
Camela, E. S. C., 186
Campos, J. P. A., 69
Campos Junior, P. H. A., 132
Campos-Chillon, L. F., 89, 90
Camugli, S., 180
Camussi, G., 226
Canel, N. G., 188, 217, 243
Canesin, H. S., 43, 220
Canisso, I. F., 155
Carlson, D. F., 240, 243
Carnevale, E., 219
Carrascal-Triana, E., 34
Carrascal-Triana, E. L., 33, 163
Carrasco, R., 135
Carreira, A. C. O., 26
Carrenho-Sala, L. C., 107, 109
Carrocera, S., 75, 97
Carson, D., 28
Casas, P. S., 130
Castañeda, S., 169
Castilho, A. C. S., 182
Castro, F. O., 103, 105
Cedeño, A., 176
Center, K., 233
Cervantes, M. P., 183
Chae, S.-K., 207
Charreaux, F., 131
Chase, C. C., 181
Chauhan, M. S., 65, 230
Chen, B., 238
Chen, J., 144
Chen, S., 238
Chen, X., 238
Chenier, T., 42
Chitwood, J., 234
Choe, C. Y., 35, 50, 215
Choi, A. R., 35, 50
Choi, J. S., 215
Choi, J.-W., 190
Choi, K.-H., 222
Choi, Y. B., 152, 153, 212
Choi, Y. H., 43, 220, 251
Choi, Y.-K., 62, 64, 156
Chokoe, T. C., 37
Chowdury, M. M. R., 87
Chung, D. G., 210
Chung, J., 27
Claes, A., 250
Coelho, V., 49
Colazo, M. G., 12
Cole, J. B., 5, 256
Collins, B., 1
Conley, A., 67
Cooks, R. G., 61
Corbin, E., 78, 161
Corradetti, B., 226
Corrêa, R., 174
Correia, E., 75
Cortes, S. F., 45
Costa, A. C., 69
Costa, D. M., 69
Costa, J., 49
Costa, P. F., 52
Covizzi, G. J., 211
Cremonesi, F., 226
Crenshaw, M., 151
Cuervo-Arango, J., 253
Cui, C.-D., 21
Cui, X.-S., 138, 190
Cupp, A. S., 181
Currin, L., 159
Curry, E., 117, 118
Curry, T. E., 250
Cushman, R. A., 181
da Rosa, P., 159
Da Silva, D. R., 69
Da Silva, K. L., 69
da Silva Santos, E. C., 46
Daels, P., 209
Daigneault, B. W., 185
Dall’Acqua, P. C., 92, 200, 203
Darwish, G. M., 41
De Canditiis, C., 40, 88
De Cesaro, M., 159
De Chico, P. L. M., 170
de Frutos, C., 240
de Stefano, E., 172
de Tarso, S. G. S., 44, 123, 253
DeLorenzo, C., 117
Denicol, A. C., 175
Denmark, S. E., 91
Deregibus, M. C., 226
Desai, N., 171
Destrumelle, S., 111
Dias, L. P., 134
Dias, M. T., 132
Diaz, F. A., 30, 202
Dickinson, S. E., 147
Dicks, N., 159
Dickson, M., 229
Dini, P., 209
Dixon, D., 233
Do, G.-Y., 207
Dochi, O., 164
Dode, M. A. N., 31
Donadeu, F. X., 224
Donadeu, X., 2
Dong, H., 144
Druart, X., 78
Duan, J. E., 144
Ducolomb, Y., 218
Dumas, C., 154
Duque, M., 228
Duranthon, V., 75
Durfey, C., 187
Durrant, B. S., 119
Eberlin, L. S., 61
Eberlin, M. N., 160
International Embryo Transfer Society
Edwards, J. L., 195, 196
Elliott, B., 84
Elliott, B. M., 83
El-Shalofy, A. S., 41
Ermilov, A. N., 143
Ervin, J. E., 128
Esposti, P., 226
Esteller-Vico, A., 67, 252
Eun, K. Y., 22
Fahrenkrug, S. C., 240, 243
Fair, S., 3
Faria, B. R. M., 130
Farin, C. E., 11
Favaron, P. O., 26, 61
Fayrer-Hosken, R. A., 115
Feliciano, M. A. R., 186
Felmer, R., 216
Ferguson, E. M., 81
Fernandez-Fuertes, B., 3
Fernandez-Martín, R., 243
Ferraris, S., 243
Ferraz, M. A. M. M., 52
Ferré, L., 234
Ferré, P., 206
Ferré, P. P., 204
Ferreira, C. R., 59, 61, 160
Feugang, J., 151, 187
Fiandanese, N., 71, 148
Fieni, F., 111
Fierro, R., 218
Figueiredo, J. R., 123, 136
Filgueira, F. G. F., 210
Fili, A. E., 242
Flöter, V., 139
Flyckt, R., 171
Forcato, D. O., 242
Forde, N., 71, 73
Fosado, M., 107, 109, 110
Foster, B., 42
Foster, B. A., 30, 202
Fratini, P., 26
French, H., 224
Frigoni, N. A. S. R., 203
Fry, R. C., 236
Fujii, T., 141
Fujiwara, K., 48
Fukuda, S., 141
Funahashi, H., 204, 206
Gad, A., 232
Gadella, B. M., 52
39
Gajda, B., 79, 80
Galiguis, J., 154
Galiguis, J. H., 228
Gallegos, P., 103
Galli, C., 191, 205
Gamarra, G., 173
Gambarini, M. L., 123
Gambini, A., 18
Gandolfi, F., 148, 191, 231
Garcia, I. F. F., 69
Garcia, J., 174
Garcia, J. M., 36
Garcia, S., 107
García-Contreras, A., 76
Garcia-Guera, A., 109
Garcia-Guerra, A., 107, 110
Garrels, W., 242
Garzon, J., 108
Gasparrini, B., 40, 88
Gastal, E. L., 44, 63, 123, 125, 253
Gastal, G. D. A., 44, 63, 123
Gastal, M. O., 63, 123, 125, 253
Geary, T. W., 147
Gebremedhn, S., 129
Gerard, N., 146
Geshi, M., 166, 179
Ghaffarilaleh, G., 102
Ghaffarilaleh, R., 102
Ghaffarilaleh, V., 102
Gibbons, J. R., 220
Giguere, S., 115
Gill, A. C., 81
Giraud-Delville, C., 75
Gismondi, M. I., 243
Gladyr, E. A., 143
Glanzner, W., 159
Golding, M. C., 4
Gomes, E., 106
Gómez, E., 75, 97
Gómez, M. C., 154, 228
Gonçalves, T. M., 130, 133
Gondro, C., 195
Góngora, A., 76
Gonzalez, L., 27, 42
Gonzalez, R., 219
Goto, Y., 96, 194
Grand, F. X., 249
Graugnard, D. E., 252
Green, A. L., 28
Green, J. A., 23, 147
40
Greene, A., 95
Greyling, J. P. C., 37
Grothmann, H., 55
Grove-White, D., 184
Grupen, C. G., 47
Gu, Y.-P., 214
Guastali, M., 165
Guastali, M. D., 182
Guevara, J. A., 76
Guha, S., 225
Guha, S. K., 227
Guimaraes, A. L., 31
Guo, J., 138
Guo, Q., 16, 17
Guo, Y., 238
Gutierrez, E. J., 30, 202
Gutierrez, K., 159
Gutiérrez-Adán, A., 97, 99, 199
Ha, A.-N., 87
Hadeler, K.-G., 197
Hales, D. B., 253
Hamdi, M., 99
Hanna, C., 51, 100
Hansen, D., 172
Hansen, P. J., 5, 34, 93, 145, 163,
175
Haraguchi, S., 46
Hardin, P. T., 30, 202
Harichaux, G., 78
Harrington, P., 32
Hashem, N., 86
Hashiyada, Y., 96, 194
Hasler, J. F., 86
Hassel, P., 15, 101
Havlicek, V., 9, 232
Heinzmann, J., 197
Hennebold, J., 51, 100
Henning, H. H. W., 52
Heo, S. Y., 221
Hermann, D., 15
Hernández, J. E., 218
Herrick, J., 95
Herrick, J. R., 121
Herrmann, D., 197, 198, 223
Hickey, R., 25
Hinrichs, K., 43, 184, 220, 251
Hirayama, H., 141
Hiriart, M. I., 242
Hoelker, M., 98, 129, 142, 232
Holm, P., 57
42nd Annual Conference
Hosseini, F., 56
Huanca, T., 77
Huanca, W., 77
Huanca, W. F., 77
Huang, Y., 238
Hugon, P., 161
Humphris, M. A., 236
Hwang, J.-U., 62, 64, 113, 149
Hwang, S. U., 22
Hyndman, D. L., 28
Hyon, S.-H., 29, 48
Hyttel, P., 192
Hyun, S. H., 22
Hyun, S.-H., 58, 85, 208
Iamartino, D., 71
Ideta, A., 235
Iglmanov, U., 126
Ikeda, T. L., 170
Ilercil, A., 94
Ille, N., 14, 63
Imai, K., 96, 164
Ioannidis, J., 2
Ishak, G., 44
Ishak, G. M., 125
Ismail, S. T., 41
Ito, J., 29, 48
Ivics, Z., 242
Izzo, M. M., 236
Jaakma, Ü., 193
Jarmusch, A. K., 59
Jarrett, S., 81
Jasmin, J., 157, 167
Jemiller, E., 19
Jensen, T. K., 72, 112
Jeung, E.-B., 62, 64, 113, 114, 149,
150, 156
Jiang, H., 238
Jiang, Q., 238
Jiang, Z., 32, 144
Jin, J.-X., 20, 60, 140
Jin, L., 16, 17, 21
Jo, Y. K., 152, 153
Johnson, S., 244
Jones, D., 104
Joo, M.-D., 87
Jordão, R. S., 172
Jue, N. K., 144
Kadarmideen, H., 192
Kageyama, S., 141
Kamoshita, M., 48
Kaneda, M., 179
Kaneko, H., 46
Kang, H. Y., 62, 64, 113, 149
Kang, J., 221
Kang, J. H., 221
Kang, J.-D., 16, 17, 21
Kannampuzha-Francis, J., 93, 175
Kanuya, N. L., 8, 37, 38, 158
Karstrup, C. C., 72, 112
Kashiwazaki, N., 29, 48
Katubayeva, B., 126
Kawasaki, Y., 29
Keefer, C. L., 229
Kemter, E., 19
Kessler, B., 19
Khan, I., 87
Khoirinaya, C., 60
Kikuchi, K., 46, 47
Kim, D. K., 35, 50, 215
Kim, E., 85
Kim, G. A., 20, 60, 140, 152, 153,
212
Kim, H. G., 22
Kim, J. D., 50
Kim, J.-S., 21
Kim, J.-W., 207
Kim, K. P., 114, 156
Kim, K.-J., 58
Kim, M. J., 152, 153, 212
Kim, N.-H., 138, 190
Kim, S. W., 35, 50, 215
Kim, S.-H., 222
Kim, T., 138, 190
Kjelland, M., 234
Kjelland, M. E., 169, 218
Klein, S., 223
Klitgaard, K. S., 72
Knox, W. B., 11
Knubben, J., 139
Knudsen, L., 112
Knudsen, L. R. V., 72
Kobayashi, S., 96, 194
Kõks, S., 193
Kong, I.-K., 87
Konishi, M., 235
Koo, D.-B., 207
Kordowitzki, P., 198
Kotlyar, A., 171
Kradolfer, D., 139
Krisher, R., 95
Krisher, R. L., 84
Kruger, M., 115
Kucia, M., 79, 80
Kuehl, T., 104
Kues, W., 243
Kues, W. A., 242
Kumar, D., 24
Kuo, L., 32
Kurian, D., 81
Kurome, M., 19
Kwang Jo, Y., 212
Labas, V., 78
Lacaze, S., 173
Lagares, M. A., 45
Lagutina, I., 205
Lamy, J., 74, 161
Lan, G., 238
Landim-Alvarenga, F., 165, 170, 182
Landim-Alvarenga, F. D. C., 160
Lange-Consiglio, A., 226
Laroucau, K., 111
Larsen, K., 15
Lazzari, G., 205
Lazzarotto, C. R., 241
International Embryo Transfer Society
Le Bourhis, D., 180
Le Guienne, B., 173
Leal, C., 165
Leal, G. R., 36, 168
Leão, B. C. S., 92, 200, 203
Lebedeva, I., 162, 201
Lee, B. C., 20, 140, 152, 153, 212
Lee, B.-C., 60
Lee, C.-K., 222
Lee, D., 114, 149
Lee, D.-K., 222
Lee, E., 58, 208
Lee, J.-H., 150
Lee, J.-S., 156
Lee, K., 213
Lee, K.-L., 87
Lee, M. H., 113, 114, 150
Lee, M. J., 150
Lee, S., 20, 60, 140
Lee, S. G., 221
Lee, S. H., 152, 153, 212
Lee, S. Y., 221
Leffers, L., 110
Legacki, E., 67
41
Lekola, K .P. M., 158
Leme, F. O. P., 45
Leme, L. O., 31
Leuchs, S., 19
Lewis, N., 184
Li, J., 15
Li, L., 229
Li, R., 15
Li, W.-X., 21
Li, X.-C., 16, 17
Li, Y., 137
Liao, S., 151
Liaudat, A. C., 242
Liere, P., 74
Lima, D., 49
Lima, L., 136
Lima, R. R., 157
Lima, W., 49
Lin, X., 238
Liu, H.-Z., 214
Liu, Y., 15
Lizaraso, I. M., 178
Llanes, A., 11
Lonergan, P., 3, 71, 73
Long, C. R., 4, 86, 94, 241
Longobardi, V., 40, 88
Looft, C., 129, 142
Looney, C. R., 86, 94
Lopera-Vasquez, R., 99
Lopes, F. E. M., 241
Lopes, F. L., 210
Lopez, A., 107
Lopez, N. A., 124
López Damian, E. P., 76
López-Helguera, I., 12
Lopez-Lake, H., 244
Lopukhov, A., 201
Lotti, S., 245
Lotti, S. N., 239
Loux, S., 67
Loux, S. C., 155
Lu, J., 151
Lucas-Hahn, A., 15, 101, 197, 223
Lynch, B., 117
Lyons, L. A., 121
Macedo, G. G., 7
Macente, B. I., 186, 210, 211
Machado, M., 192
Machaty, Z., 189
Maciel, G. S., 186
42
MacLean, R. A., 154
Macleod, J. N., 252
Maculan, R., 130, 133
Mafolo, K. S., 38
Magalhães, L., 165
Magness, S., 27
Mahapatra, C. T., 59
Maillo, V., 97, 99, 199
Maio, J. R., 174
Malakar, D., 225, 227
Malda, J., 52
Malik, H., 225
Malik, H. N., 227
Mall, E., 223
Mamani, C., 77
Manik, R. S., 65, 230
Manríquez, J., 105
Mapeka, M. H., 8
Mapletoft, R. J., 183, 248
Mapletroft, R., 236
Maqhashu, A., 8
Marini, MG, 226
Marinovic de Oliveira, C., 26
Marjani, S., 32
Mark, E., 193
Martin, D., 75
Martin, R. F. Y., 18
Martin, U., 101, 223
Martín, D., 97
Martins, A., 165
Martins, H. S., 45
Martins, L. F., 7
Martins, L. T., 241
Martinus, R., 53
Mastromonaco, G., 183
Matoba, S., 166, 179
Matsuda, H., 96, 194
Matsumura, K., 29, 48
Maziero, R., 165
McCracken, V., 213
McGowan, L. T., 28
McGregor-Argo, C., 184
McNeel, A. K., 181
Meade, K. G., 3
Meirelles, F. V., 26
Melo, L. F., 110
Memili, E., 254
Men, N. T., 46
Mendes, V. R. A., 157
Mendoza, G. D., 76
42nd Annual Conference
Meng, F., 28
Menges, S., 86
Mermillod, P., 74, 78, 161
Mesquita, F. S., 160
Mess, A. M., 26
Miceli, D. C., 199
Michalovic, L., 159
Miglino, M. A., 26, 61
Miles, J. R., 181
Miller, D. J., 185
Milner, D., 244
Mingoti, G. Z., 92, 200, 203
Moawad, A. R., 41
Monteiro, B. M., 109
Monteiro, C. A. S., 36, 168
Moraes, E., 49
Mordhorst, B. R., 23
Moreira, G., 176
Moreira, G. M., 133
Morel, A., 131
Moreno, J. F., 107, 109, 110
Moreria, G., 246
Moriyasu, S., 141
Moro, L., 18
Motheo, T. F., 210, 211
Motta, J. C. L., 110
Mouamen, A., 13
Mouneyres, M., 173
Moura, A. A., 255
Mphaphathi, M. L., 8, 37, 38, 158
Muñoz, M., 75, 97
Murillo, A., 97
Murphy, S. L., 23, 25
Muterlle, C. V., 31
Myles, L., 187
Nagai, T., 46, 166, 179
Nagle, K. A., 196
Naito, A., 141
Najafzadeh, V., 53
Nakamura, M., 235
Nam, T. D., 62
Narciandi, F., 3
Nasser, L. F., 106
Navarro, R. D., 178
Nedambale, T. L., 8, 37, 38, 158
Negrón-Pérez, V. M., 175
Nephawe, K. A., 8
Neri, H. L. D., 10
Neto, A. A., 70
Neto, S. G., 241
Netshirovha, T. R., 37
Netto, S. B. S., 31
Neuhaus, G., 101
Neuhoff, C., 129, 142
Niemann, H., 15, 101, 197, 198,
223, 243
Nkadimeng, M., 8, 37, 38, 158
Nociti, R. P., 186
Noguchi, J., 46
Nogueira, A. H. C., 172
Nogueira, G. P., 211
Nogueira, L. A. G., 36, 168
Nogueira, M., 192
Nõmm, M., 193
Norman, H. D., 256
Nowak-Imialek, M., 223
Nugraha Setyawan, E., 212
Null, D. J., 5, 256
Nunez, C., 99
Nyberg, S. L., 25
Oback, B., 53
Oback, F. C., 28
O’Brien, J. K., 116
Ochoa, J. C., 110
O’Farrelly, C., 3
Ogata, R. A., 172
Oh, A., 20, 140
Oh, H. J., 152, 153, 212
Oh, J.-N., 222
Ohtake, M., 96, 194
Okuda, L. H., 172
Oliveira, C. S., 36, 134, 168, 177
Oliveira, F., 70
Oliveira, F. D., 68
Oliveira, J. A. S., 157
Oliveira, M. E. F., 186
Oliveira, M. F., 68
Oliver, J. E., 28
Olmos Nicotra, M. F., 242
O’Neill, R., 144
Ortega, M. S., 5, 175
Ortiz, I., 43
Oseikria, M., 111
Oviedo, J. M., 246
Pacheco, L. P., 172
Paczkowski, M., 84, 104
Padilla, L., 120
Paiva, S. O., 44, 63
Palazzi, E. G., 172
Palm, F., 9, 232
Palomino, J. M., 39, 122, 183
Palta, P., 65, 230
Paniz Parvar, S., 56
Panyaboriban, S., 120
Papp, S., 9, 232
Park, H.-J., 207
Park, J.-Y., 207
Park, P.-R., 87
Park, S. M., 221
Park, T.-Y., 222
Parnpai, R., 166
Parra-Forero, L. Y., 76
Paschoal, D., 165
Pascucci, L., 226
Pasquariello, R., 148
Passaro, C., 73
Payton, R. R., 195, 196
Pedersen, H., 192
Pedersen, H. G, 112, 72
Pegah Parvar, S., 56
Pelizzari, M., 108, 176
Pellerin, J. L., 111
Penitente-Filho, J. M., 33, 163
Penna, C. F. A. M., 45
Pennarossa, G., 231
Peoples, M. D., 4
Pereira, D. C., 107
Perrini, C., 226
Perry, G. A., 181
Pesce, M., 231
Petersen, B., 15, 223
Petkov, S., 101, 223
Petren, K., 117
Peynot, N., 75
Piedrahita, J., 1, 27
Pinto, T. L. C., 133
Pires-Butler, E. A., 211
Pirro, V., 59, 61
Pituco, E. M., 172
Pivato, I., 31
Pocar, P., 148
Pohler, K. G., 147
Polkoff, K., 245
Polkoff, K. M., 91, 124
Ponsart, C., 173
Pontes, J. H. F., 106
Poole, R., 213
Pope, C. E., 154, 228
Popovic, L., 28
Prather, R. S., 23, 25, 82, 83, 84
International Embryo Transfer Society
Pribenszky, C., 32
Pryor, J. H., 4, 86, 94
Pukazhenthi, B., 120
Pukazhenthi, B. S., 229
Qin, J., 214
Quadalti, C., 205
Quintão, C. C. R., 157, 167
Quinton, H., 131
Ramirez, M. A., 99
Ramirez, Y. E., 106
Ramon, L., 49
Ramsey, C., 51, 100
Ramukhithi, F. V., 8
Raoofi, Z., 56, 127
Raphalalani, Z. C., 8, 37, 38, 158
Rascado, T. D. S., 160
Razaghian, F., 127
Razza, E., 192
Rea, M., 131
Recalde, E. C. S., 170
Redel, B. K., 82
Redel, B. R., 84
Reed, D., 120
Revol, J., 246
Reynaud, K., 78
Rhoads, M., 213
Rhodes-Long, K., 90
Ribeiro de Lima, R., 69
Rings, F., 98, 129
Rink, E., 224
Riou, C., 146
Rispoli, L. A., 195, 196
Rizos, D., 99, 199
Robeck, T. R., 116
Roberts, M. A., 89
Robinson, C., 128
Rocha, R., 136
Rocha-Frigoni, N. A. S., 92, 200
Rodrigues, A. L. R., 36, 168
Rodrigues, S. A. D., 31
Rodriguez, N., 242
Rodriguez, S. J. R., 106
Rodríguez-Alvarez, L. I., 103, 105
Roelen, B. A. J., 66
Roldán-Olarte, M., 199
Romanenkova, O. S., 143
Romek, M., 79, 80
Romo, S., 169, 218
Romo-García, S., 76
Rorie, R., 233
43
Rosa, P. M. S., 36, 134, 168, 177
Rosa, R. G., 130, 133
Roser, J. F., 125
Ross, P., 234
Ross, R. M., 23
Roth, T., 117
Roth, T. L., 116, 118
Rubessa, M., 91, 106, 124, 136, 245
Ruiz, A., 33, 34, 163
Ryan, P., 151, 187
Ryu, S.-M., 21
Saini, M., 65
Saini, N., 230
Saini, S., 225, 227
Saint-Dizier, M., 74, 146, 161
Sala, R. V., 7, 107, 109, 110
Salamone, D., 18
Salamone, D. F., 188, 217, 242, 243
Salazar, M. D. P., 178
Saldaña, G. B., 178
Salilew-Wondim, D., 98, 129, 142
Salvetti, P., 180
Salzano, A., 40, 88
Sanchez, R., 77
Sánchez-Calabuig, M. J., 199
Sanders, W., 151
Sandra, O., 75
Santoro, R., 231
Santos, A., 70
Santos, A. C., 68
Santos, V. G., 160
Santos, V. J. C., 186
Saraiva, H. F. R. A., 36, 168
Sarv, O., 193
Savy, V., 242, 243
Sawai, K., 141
Saxton, A. M., 195, 196
Scarlet, D., 14, 63
Schellander, K., 98, 129, 142
Schibler, L., 180
Schlaf, M., 42
Schmitt, E., 180
Schnauffer, K., 184
Schoolcraft, W., 95
Schou, K., 112
Schreiner, B., 14
Schuermann, Y., 159
Schuler, G., 54
Schütz, L. F., 128
Scoggin, K. E., 155, 250, 252
44
Seisenbayeva, A., 126
Selokar, N. L., 24, 65
Seolwana, L. F., 8
Seong, H. H., 35, 50, 215
Sepúlveda, M. S., 59
Serapião, R. V., 36, 134, 168, 177
Sermyagin, A. A., 143
Seshoka, M. M., 8, 37, 38, 158
Setyawan, E. M. N., 152, 153
Shah, S. M., 230
Shang, J.-H., 214
Sharma, A., 230
Sharma, P., 24
Sharma, R. K., 24
Sharma, V., 225, 227
Shedova, E., 162, 201
Shewfelt, G., 86
Shibao, Y., 29
Shier, D. M., 119
Shim, H., 221
Shin, E.-K., 113
Silva, E. P., 10
Silva, G. C., 45
Simpson, S., 1, 27
Singh, J., 135
Singh, M. K., 230
Singh, R. P., 120
Singina, G., 162, 201
Singla, S. K., 65, 230
Siqueira, L. G., 145
Siqueira, L. G. B., 175
Sirard, M. A., 247
Smith, L. F., 130
Smith, M. F., 147
Smits, K., 209
Smorag, Z., 79, 80
Snyder, M., 94
Snyder, M. D., 4
Soleilhavoup, C., 78
Somfai, T., 46, 47, 166, 179
Son, D., 222
Song, S.-H., 87
Songsasen, N., 120, 137
Sousa, P., 49
Souza, E. D., 157, 167
Souza, J. C., 130, 133
Souza, M. R., 45
Spate, L. D., 23, 25, 82, 83, 84
Spencer, T. E., 73
Sper, R., 1
42nd Annual Conference
Spicer, L. J., 128
Spricigo, J. F. W., 31
Squires, E., 67
Squires, E. L., 155, 250, 252
Srirattana, K., 6
St. John, J. C., 6
Stadaioli, G., 191
Stahlberg, R., 45
Stanley, S., 67
Stein, K., 9, 232
Stella, A., 71
Stewart, J., 245
Stillacci, M. G., 191
Stöhr, J., 55
Stoll, M., 109
Stoops, M. A., 116
Stout, T. A. E., 52, 66
Strøbech, L., 86
Stroebech, L., 192
Strozzi, F., 71
Sturmey, R., 184
Sudano, M., 165
Sudano, M. J., 160, 170
Sugimura, S., 96
Suttirojpattana, T., 166
Suvá, M., 188, 217
Suzuki, O., 237
Swaisgood, R. R., 119
Swanson, W. F., 121
Swelum, A., 13
Taboga, O. A., 243
Tainturier, D., 111
Takayama, M., 164
Talluri, T. R., 242
Tamayo Palacio, J. E., 88
Tasan, I., 239
Tata, A., 160
Tavares, K. C. S., 241
Tazawa, S. P., 253
Techakumphu, M., 120
Teixeira, L. P. R., 241
Tenley, S. C., 181
Tesfaye, D., 98, 129, 142, 232
Tholen, E., 129, 142
Thongkittidilok, C., 137
Tian, X., 32
Tian, X. C., 144
Tío-Castro, A., 199
Toishibekov, Y., 126
Tominaga, N., 235
Tomomatsu, M., 235
Torres, C. A. A., 33, 34, 163
Tosta, L. C. C., 109
Tosta, R. D., 109
Totty, M., 128
Tran, M. T., 204, 206
Tribulo, A., 108, 176, 246
Tribulo, H., 108, 246
Tribulo, P., 145, 175
Tribulo, R., 108
Troedsson, M., 67
Troedsson, M. H., 155, 252
Troedsson, M. H. T., 250
Tsikis, G., 78
Turin, J., 77
Uhde, K., 66
Ulbrich, S. E., 139
Ulloa, S. M. B., 197
Uscategui, R. A. R., 210
Valente, R. S., 160
Valiyeva, B., 126
Vallet, J. L., 181
Van Dorenmalen, K. M. A., 52
van Marle-Koster, E., 158
van Tol, H. T. A., 66
Vandenplas, M., 115
Vansandt, L. M., 121, 229
VanSoom, A., 209
Vargas, T., 216
Vasconcelos, C. O. P., 168
Vasconcelos, G. L., 133
Velasquez, A. E., 103, 105
Veraguas, D., 103, 105
Viafara, J. A. S., 133
Viana, D. C., 68
Viana, J. H. M., 132, 167
Vicente, W. R. R., 186, 210, 211
Vieira, L. M., 248
Viglino, A., 71, 148
Vigneault, C., 249
Vivanco-Mackie, H. W., 178
Vorimore, F., 111
Vos, P. L. A. M., 52
Wakai, T., 204, 206
Walleser, E. A., 110
Walters, E., 25
Wang, C., 189
Wang, G., 154
Watson, E., 224
Webster, D., 240
Wei, L., 238
Wei, Y., 238
Wells, D. N., 28
Wells, K. D., 23, 25
Westhusin, M. E., 4
Wheeler, M., 244, 245
Wheeler, M. B., 91, 106, 124, 136,
239, 241
Whitelaw, B., 241
Whittaker, P., 12
Whitworth, K. M., 25
Wiggans, G. R., 256
Wildt, D., 137
Willard, S., 151, 187
Williams, J. L., 71, 148
Williamson, G. L., 4
Wiltbank, M. C., 107, 109, 110
Winters, R., 124, 136
Wohlgemuth, S., 5
Wolf, E., 19, 144
Wood, J. R., 181
Wrenzycki, C., 54, 55
Wu, Y., 238
Wunderlich, S., 101, 223
Wynn, M., 67
Xu, L., 87
Yadav, P. S., 24
Yamanouchi, T., 96, 194
Yanez-Mo, M., 99
Yang, C.-Y., 214
Yang, S. X., 39, 122
Yang, S.-G., 207
Yin, X.-J., 16, 17, 21
Yoon, J. D., 22, 208
Zakhartchenko, V., 19
Zambrano, F., 216
Zanneti, E. S., 228
Zecconi, A., 191
Zhang, L., 128
Zhang, M., 32, 238
Zhang, X., 1
Zhang, Y.-C., 16, 17
Zhao, H., 239
Zheng, H.-Y., 214
Zheng, X., 144
Zhou, R., 229
Zhu, H.-Y., 16, 17, 21
Ziegler, M., 15, 101
Zinovieva, N., 162, 201
Zinovieva, N. A., 143
International Embryo Transfer Society
Zolini, A., 163
Zolini, A. M., 33, 34
Zullo, G., 40, 88
45
2016 Recipients of the IETS Distinguished Service Award
Naida M. Loskutoff
The 2016 IETS Distinguished Service Award has been awarded posthumously to Naida Loskutoff in honor of her long-term support and significant contributions to the IETS.
Loskutoff made some important decisions early in her academic career
that helped forge her path in research and application of embryo technologies in wildlife species. Unknown to her at the time, her early mentors
included two innovative researchers who would later become recipients of
the IETS Pioneer Award. With a BS degree in zoology from the California
State Polytechnic University, Loskutoff continued on to obtain her MS
and PhD degrees from the Department of Veterinary Physiology and
Pharmacology, Texas A&M University. It was during her graduate studies
that she began focusing on in vitro embryo production and manipulation
techniques. Her PhD advisor, Duane Kramer (2006 IETS Pioneer Award),
became a long-time mentor whom she fondly referred to throughout her
career. She followed her PhD with a postdoctoral fellowship at the Animal
Biotechnology Embryo Laboratory, University of Guelph, under the supervision of Keith Betteridge (2003 IETS Pioneer Award) where she produced the world’s first identical quadruplet
bulls from a single four-cell embryo.
In 1992 Loskutoff began her professional career at Omaha’s Henry Doorly Zoo and Aquarium, Nebraska, where
she became Director of Reproductive Sciences. During her time in this position, she not only mentored numerous young students and scientists within the United States and around the world, but she was instrumental in
promoting the application of reproductive technologies in nondomestic and endangered species. Her novel
research resulted in significant accomplishments, including a patented sperm disinfection protocol and the first
gorilla birth from IVF-ET. Most importantly, she focused on developing methods that were transportable, allowing her to explore new species in their home environments, and as a result, spent much of her time bringing
wildlife reproductive science to Africa.
Loskutoff was a member of IETS for more than 30 years, having joined the society in 1982. During that time,
she was a consistent attendee at the annual conferences, as well as an active contributor to the society with
participation in the HASAC committee (2000–2015), Foundation (1993–1999), and Board of Governors, specifically in the roles of vice president (2006), president (2007), and immediate past president (2008). For the
2001 annual conference in Omaha, Nebraska, Loskutoff was the chair of the Local Organizing Committee.
In that same year, she had the vision to establish the Committee for Companion Animals, Non-Domestic and
Endangered Species (CANDES). As chair of CANDES for more than 10 years, she initiated discussions on
topics that were not even being considered at the time for nondomestic and endangered species, including biosecurity, regulatory requirements for import and export, and legalities of ownership. She put significant effort
into getting CANDES established and supported several symposiums over the years, both in Omaha and during
subsequent IETS annual conferences.
In her many roles within IETS, Loskutoff dedicated her time with pleasure and was a strong advocate of the
importance of IETS as a society and resource for veterinarians, reproductive biologists, technologists, and students. Her noteworthy accomplishments in wildlife reproductive science and within IETS make her a worthy
recipient of the Distinguished Service Award.
46
42nd Annual Conference
Steph Dieleman
P. L. A. M. Vos et al., Theriogenology 84 (2015) 321–322
Associate Professor Steph J. Dieleman was born in 1944 and grew up in
Twisk, a village located at the northwest of the Netherlands. After finishing secondary school he started his academic career at Utrecht University
as a student of the Chemistry Faculty and passed his majors in organic
chemistry and biochemistry. During his study he proved his first scientific
potency by receiving a prestigious prize for his research project performed
in the Shell/AKZO Company. In June 1969 he passed his final doctoral
exams and started his working career at the TNO Chemical Institute,
Utrecht. For a period of two years, he focused on a project about biochemical aspects of the resistance against specific fungi infections in apples.
In November 1971 he accepted a position and hence a new challenge at
Utrecht University at the Faculty of Veterinary Medicine, Department
Obstetrics, Gynaecology, and AI. This career switch was the start of
his research focus on the hormonal processes around parturition, and
Dieleman’s first challenge was to develop the Radio Immuno Assay (RIA) technology for hormonal (steroid)
analysis and to set up an endocrine laboratory for reproductive studies. In 1973 he became the head of this
endocrine biochemistry laboratory. During those days he went to Edinburgh, Scotland, for a sabbatical to learn
more about RIA, and indeed he developed the first oestradiol RIA in Utrecht using an antiserum that he received
as a present during this visit. In 1975 he started his PhD project on the endocrinology of the bovine oestrous
cycle, finally resulting in the acceptance of his PhD thesis titled “Steroids of preovulatory bovine follicles
relative to the peak of luteinizing hormone,” which was successfully defended at Utrecht University in 1984.
This study showed that concurrently with changes in the micromorphology of the follicular wall, the steroidal
microenvironment of the maturing bovine oocyte switches from predominantly oestrogenic at oestrus to one in
which progesterone is the major steroid present shortly before ovulation. Through this milestone Dieleman created a solid scientific basis for the introduction and further development of embryo technologies and hence a
better understanding of the endocrinology during the normal oestrus cycle and the follicular development during
superovulation in cattle. In this respect, the testing of the first monoclonal antibody in PMSG superstimulated
animals, called “anti-PMSG,” was performed for the company Intervet, the Netherlands.
Especially, his interest for follicular development in relation to (final) oocyte maturation has to be mentioned.
An impressive number of (EU) projects and in vivo experiments have been performed, mainly in cattle, to gain
a better understanding on the communication between the oocyte and its microenvironment. To this end many
laboratory tools have been developed and validated to support and prove the proposed experimental hypothesis:
from hormonal analysis, staining techniques, in vitro production of blastocysts to genomics and proteomics.
Steph Dieleman performed research in the context of reproductive physiology, hence he stimulated in vivo
research being the bridge between fundamental and the application in the field. To reach this goal, the multidisciplinary approach was an absolute prerequisite for him; he always strongly believed in his own original scientific concepts, ideas, and hypotheses, although those were always open for discussion and critical comments.
In the field of reproductive physiology, technology, and embryo production, the national and international
research achievements of Dieleman have been impressive: a very productive, worldwide-acknowledged, multifaceted research program in species such as cows, pigs, horses, elephants, dogs, dolphins, and ostriches.
Through his work in collaboration with many outstanding and acknowledged research colleagues and international groups, he has contributed to more than 300 book contributions, abstracts, and full papers in regular and
top scientific journals, including 38 refereed papers in Theriogenology.
The supervision and professional guidance of dozens of postdocs, PhDs, and research students was an important
achievement of Dieleman. Dieleman liked this work, as a perfectionist and hard worker; he was therefore very
stimulating for all his students and colleagues who have spent time at his laboratory. They have experienced the
laboratory of Steph Dieleman to be a pleasant and stimulating environment for performing excellent research,
with lots of fun and good team work in a warm atmosphere.
International Embryo Transfer Society
47
During his scientific career, Dieleman received much recognition for his contributions and achievements.
Over the years, he was awarded academic honors, and he was invited as keynote speaker and session chair at
many international scientific conferences and meetings. He has been a member of the Board of Governors and
secretary-treasurer and president of the International Embryo Transfer Society (IETS). In the IETS he was recognized for his financial expertise (“the master of finance”). For his active membership and contributions, the
AETE society awarded him with the Pioneer Award at the annual meeting of the AETE in Sardinia in 2007.
Moreover, Steph Dieleman was an expert in organizing scientific meeting programs, and as a result he organized
an impressive list of recognized scientific meetings worldwide and in the Netherlands. For example, he chaired
the IETS meeting in Maastricht (2000), which was also organized by him. Furthermore, he was president of
the International Congress on Animal Reproduction (ICAR) and organized the ICAR conference in The Hague
(1992). He also organized the European Embryo Transfer Society (AETE) meeting (2002) and the International
Conference on Pig Reproduction (ICPR) (2004), both organized in the ancient monastery Rolduc.
Finally, at the end of his impressive career, Dieleman organized the International Conference on Farm Animal
Reproduction (ICFAR), again in Rolduc, the Netherlands (June 2007), which was his final international recognition to his scientific colleagues and friends that were gathered together from all over the world to contribute to
this special and memorable scientific meeting. To this end he was special editor of the proceedings that appeared
as a special issue in Theriogenology [1], containing 34 excellent contributions covering several main topics in
the field of theriogenology. This special issue not only was a concise update of the state of the art in research
“from egg to embryo,” but also exemplified the broad and multidisciplinary approach Steph Dieleman added
during his scientific career to this exciting field.
For more than 30 years Steph Dieleman performed interdisciplinary, clinically orientated research and education in the field of reproduction. He was a true and driven scientist who exposed a serious attitude, but he also
very much enjoyed the Burgundy lifestyle, which he continued after his retirement. One of the last photographs
that was taken of him, at his 70th birthday, is characteristic and gives an honored remembrance of our friend
and colleague Steph Dieleman. He is enjoying his glass of wine with his closest friends during a boot trip in his
favorite waterside landscape at Loosdrecht. Steph Dieleman passed away February 15, 2015. We will remember
Dieleman as a warm person, an excellent scientist, and a dear colleague and friend.
Reference
[1] S. J. Dieleman (guest editor). Proceedings of the International Conference on Farm Animal Reproduction.
Kerkrade, Limburg, the Netherlands, May 27–31, 2007. Theriogenology 2006, 68 (Suppl. 1), pp. S1–S228.
48
42nd Annual Conference
Special Events
Morulas’ Preconference Workshop
Epigenetics and Embryo Technologies
Saturday, January 23
14:00–17:00
Nunn
Sponsored by CSIRO
This preconference symposium organized by the IETS Morulas Board of Governors explores the growing field of
epigenetics as it relates to gamete and embryo technologies. Trainees should plan to arrive one day early and take
advantage of this great opportunity by hearing from excellent speakers who are leading researchers in the field of
epigenetics including Pablo Ross, Marc-André Sirard, and Jason Knott. All IETS members are welcome to this threehour event that will cover the basics of epigenetics while incorporating new techniques and real-life data examples
along with discussion from the audience. Trainees will have the opportunity to interact with each other and speakers
in a short discussion that will close the conference. Please take advantage of this wonderful opportunity at a fantastic
cost (registration required).
Affiliates Lounge
Sunday–Tuesday, January 24–26
Wilson
07:00–17:00, Daily
A room has been reserved for the society affiliates to meet and network for the duration of the meeting.
Morulas and Mentor Luncheon
Sunday, January 24
12:30–14:00
Clements
Sponsored by Vetoquinol
One of the main goals of the Morulas is to provide trainees opportunities to interact with the general membership of
the IETS. The Morulas and Mentors luncheon is designed to give trainees a chance to sit down with mentors in small
groups to develop meaningful connections with leaders in our field. Join a number of outstanding Mentors at this
annual event and choose from one of eight Mentors that you would like to dine with (ticket required).
Practitioners’ Forum
Sunday, January 24
16:00–18:00
Ballroom C
Sponsored by Partnar Animal Health Inc.
DABE Forum
Sunday, January 24
16:00–18:00
Nunn
Contributions from new investigators
Sponsored by Hamilton Thorne Inc.
Welcome Reception
Sunday, January 24
18:00–19:00
Grand Ballroom A, B
Sponsored by Professional Embryo Transfer Supple Inc. (PETS)
A welcome reception will be held in the Grand Ballroom A, B of the Galt House Hotel from 18:00 to 19:00. Meet the
exhibitors and renew old friendships. Wine, cocktails, and light hors d’oeuvres will be served.
International Embryo Transfer Society
49
Concurrent Session
Monday, January 25
08:00–09:30
Ballroom C
Bovine reproduction: Donor selection and treatments to improve oocyte collection and in vitro embryo production in
cattle
Concurrent Session
Monday, January 25
08:00–09:30
Nunn
Equine reproduction: Pre- and post-ovulatory influences on fertility
Concurrent Session
Monday, January 25
08:00–09:30
Jones
From bench to barn: Effects of bull fertility or sperm factors on embryo development and cattle agriculture
Morulas Career Luncheon
Monday, January 25
12:45–14:00
Clements
Sponsored by CSIRO Publishing
This year’s Career Luncheon will feature a talk by two speakers who will share unique perspectives from their
own personal career paths. Hear Jennifer Barfield of Colorado State University speak about her bison program and
Christian Vigneault from Boviteq in Québec speak about his research with bovine oocytes and embryos. This forum
provides a chance for trainees to gain perspective outside of their current work environment and meet others with
similar aspirations (ticket required).
Open Meeting of the Health and Safety Advisory Committee (HASAC)
Monday, January 25
18:00–19:00
Nunn
Morulas’ Trainee Forum
Monday, January 25
18:00–19:00
Ballroom C
All trainees are invited and encouraged to attend the Morulas Trainee Forum. The Board of Governors will be updating the membership on activities and attending to business matters. In addition we will welcome the new Morulas
president, recognize the 2015 Mentor of the Year Recipient, and discuss important events and opportunities for all
trainees. This is a great time to get involved and boost your international relations.
Morulas’ Student Mixer
Monday, January 25
19:30
Galt House Hotel, Al J’s at the Conservatory
After business comes fun! Shortly after the Morulas Trainee Forum, everyone is invited to gather with friends and
drinks for a social event. Hosted by IETS, this annual event is a fun time for all trainees and general members to
relax and enjoy the atmosphere. Take advantage of meeting new people and establish connections that last a lifetime.
50
42nd Annual Conference
The mixer will conveniently be located in the Atrium on the 2nd floor of the Galt Hotel. Registration and tickets are
NOT required.
14th Annual IETS Fun Run
Tuesday, January 26
12:30–13:00
Louisville Waterfront Park
Even if you do not participate, come and cheer on the runners in a magnificent landscape and even dress up!
Closing Party
Tuesday, January 26
18:30–22:30
Kentucky Derby Museum
Come and join us for the event of the week. Share a meal, enjoy the surroundings, and get reacquainted with old
friends. There will be music for your listening and dancing pleasure. You will also have a chance to tour the museum.
With two floors of interactive, family-friendly exhibits, the Kentucky Derby Museum takes visitors through every
stage of a Thoroughbred’s life, from birth to the first Saturday in May. Learn about all the great history of the race
dating back to the inaugural running in 1875 and about our most recent Kentucky Derby champion. Experience all
the traditions that have transformed the race into what it is today. You will also have a chance to view the new stateof-the-art 360° Greatest Race Experience. Don’t miss this event. Shuttle service to and from the Galt House to the
museum will be provided, beginning at 18:00, from the Galt House Hotel, outside of the Suite Tower, with return
service beginning at 19:30. Tickets are required for this event.
International Embryo Transfer Society
51
Exhibit Hall Layout
52
42nd Annual Conference
Exhibit Directory
Booth Listing by Number:
Booth #
Company
1 .......................................... ICPbio Reproduction
2 ......................................... American Embryo Transfer Association (AETA)
3 .......................................... MOFA Global
4 .......................................... Biogenics Inc.
5 & 6..................................... WTA Technologies LLC
7 .......................................... IVFtech ApS
8 .......................................... RI Life Sciences
9 .......................................... Misawa Medical Industry Co. Ltd.
10 & 11..................................Vetoquinol
12.......................................... Partnar Animal Health
13.......................................... IVF Bioscience
14 ......................................... Echo Control Medical
15.......................................... IMV Technologies
16.......................................... Professional Embryo Transfer Supply Inc. (PETS)
18.......................................... Agtech Inc.
19..........................................Boviteq
20.......................................... STEM ALPHA
International Embryo Transfer Society
53
Alphabetical Listing of Exhibitors
Agtech Inc.
Agtech Inc. is your source for the embryo transfer products you can count on for success. In all that we do,
from product development to assisting with your product needs, it is the drive to build on our customer’s success that keeps us moving forward.
We are driven to help our customers succeed, helping
them continue to be leaders in the industry and support
their goals.
We take great pride in our relationships with our customers, providing them with the products, support, and
attention to detail that they have come to rely on.
8801 Anderson Avenue
Manhattan, KS 66503-9612, USA
Phone: 800-367-4016
Fax: 785-776-4295
www.agtechinc.com
Booth: 18
In 2014, Biogenics released BioViatorTM, the world's
first portable, ambient compensating incubator/refrigerator that does not require a padded shipping carton
increasing its dimensional weight. Battery life is up to
72 hours at 38°C. Datalogging and ramp functions are
standard, as well as many other patent-pending features.
At IETS in 2016, Biogenics unveils BinucleateTM, a
faster, simpler, and more reliable method for seeding embryo straws when used in conjunction with our
C2060S cryochamber.
Biogenics' technical specialists cordially welcome your
visit to our stand, and appreciate your valuable time.
American Embryo Transfer Association (AETA)
The purpose of the American Embryo Transfer
Association is to unite those organizations and individuals in the United States engaged in the embryo transfer
industry into an affiliated federation operating under
self-imposed standards of performance and conduct
• to present a unified voice of the industry to promote
the mutual interests and ideals of its members;
• to protect the users of the embryo transfer industry to
the extent technically and ethically possible;
• to educate the public properly to the status and capability of the United States embryo transfer industry;
• and to encourage others to engage in the pursuit of this
industry.
1800 South Oak Street
Suite 100
Champaign, IL 61820, USA
Phone: 217-398-2217
http://www.aeta.org
Booth: 2
Biogenics Inc.
Biogenics has specialized in the focused niche of cryopreservation instruments and temperature control for
embryo transfer since 1990. In 2015, our 25th year in
business, we engineered and manufactured generation
two of our groundbreaking CRYSALYS® 9500 freezing
54
system, incorporating numerous changes to make these
systems more robust than ever. Freeze at rates as high as
70°C per minute or as low as 0.1°C per minute, and in
large or small volumes of straws.
2797 Napa Valley Corporate Dr.
Napa, CA 94558-6216, USA
Phone: 707-224-7024
http://www.biogenics.com
Booth: 4
Boviteq
A world leader in developing and implementing new
techniques for embryo transfer, Boviteq offers reproductive and genetic solutions to clients across North
America from its world-class in vitro–fertilization
(IVF) laboratories in Madison, Wisconsin, and SaintHyacinthe, Québec.
Working with accredited OPU (ovum pickup) centers, clients can take advantage of Boviteq’s innovative reproductive technologies. “Our goal is to make
available a range of options to enable breeders to
optimize the reproductive career of their elite animals
and manage the genetic advancement of their herds,”
explains Boviteq’s Director of Embryo Operations and
R&D, Patrick Blondin.
This network of accredited OPU centers and vets is
part of a unique, 360° IVF embryo solution, and as a
fully integrated semen and IVF embryo facility, Boviteq
is one of the industry’s only true genetic solutions
company.
6330 Copps Avenue, Suite B
Monona, WI 53716, USA
Phone: 608-210-4151
http://www.boviteq.com/us-home
Booth: 19
42nd Annual Conference
Echo Control Medical
The company ECM has been in the field of ultrasound
scanning for more than 30 years.
We design and produce a complete range of ultrasound
systems for reproduction diagnosis and ovary exam.
Come and see the Exago as well as the Exapad, which
can be equipped with on OPU guide and deliver an outstanding image quality for oocyte retrieval applications.
126 Bd De La Republique
F-16000 Angouleme, France
Phone: +33 5 45 92 03 57
http://www.ecmscan.com
Booth: 14
ICPbio Reproduction
ICPbio Reproduction is a global supplier of embryo
transfer and reproductive products, including flushing
and embryo handling media for the equine, bovine, and
ovine used by veterinarians and reproductive specialists.
ICPbio Reproduction also manufactures and distributes
the Ovagen brand FSH for superovulation of ovine and
bovine for embryo transfer procedures.
PO Box 39
303 South McKay Avenue
Spring Valley, WI 54767, USA
Phone: 877-978-5827
http://www.icpbiorepro.com
Booth: 1
IMV Technologies
The world’s leading player in the reproductive biotechnology market, IMV is present in 120 countries.
IMV designs and develops equipment, consumables, and
preservation media dedicated to animal reproduction,
bringing each idea from the drawing board to large-scale
production.
Today, IMV Technologies develops solutions designed
to simplify the processes involved in herd improvement
programs around five main areas of expertise:
• semen collection and analysis,
• sample preparation and dilution,
• packaging and cryopreservation,
• assisted insemination, and
• embryo transfer.
IMV Technologies offers the widest range of ET products, including collection and freezing media, filtration
devices, laboratory equipment, and embryo packaging
and transfer tools. See our complete range at www.imvtechnologies.com.
11725 95th Avenue North
Maple Grove, MN 55369, USA
http://www.imv-technologies.com
Booth: 15
IVF Bioscience
Bringing a new approach to the animal ART market,
IVF Bioscience’s new IVP media range for bovine,
caprine, and ovine will help to produce an increased
number of higher quality embryos. Alongside our readyto-use and serum-free media, we aim to provide exceptional service levels to help create a more productive
world. Join us at stand 13 to find out more about our
IVP media line.
Bickland Industrial Park
Falmouth, Cornwall TR11 4TA, United Kingdom
Phone: +441326 372 733
http://www.research-instruments.com
Booth: 13
IVFtech ApS
IVFtech is a company producing high quality, customizable equipment for IVF laboratories.
The art and science of assisted reproduction often
demands personalized solutions where strict considerations must be given to the culture conditions and the
growth environment of gametes and embryos. Key factors for success rely on providing a steady temperature
close to 37°C and securing an atmosphere with the right
humidity and CO2 concentration.
IVFtech knows that not all laboratories are the same,
that’s why IVFtech combines the bespoke nature of our
products with a high quality and service level.
Custom products and services are, by definition, unique.
Toppevadvej 34-38
DK-3660 Stenløse, Denmark
Phone: +45 3940 2565
Fax: +45 3940 2564
IVFtech ApS CVR no.: 20892307
e-mail:[email protected]
www.ivftech.dk
Booth: 7
Misawa Medical Industry Co. Ltd.
We are one of the leading manufacturers and distributors of disposable needles and cow ova vacuuming, ET
products for veterinary purpose in Japan. We established
a worldwide reputation for our reliability and expertise
based on experiences over half a century.
International Embryo Transfer Society
55
Our products for veterinary purposes, cow ova vacuuming needles, “mo-No.4” embryo transfer catheter, and
“mo-No.5” intrauterine horn catheter were developed
and manufactured with a special and innovative method.
Box 188
Canton, TX 75103, USA
www.pets-inc.com
Booth: 16
We offer these products of superior quality, all manufactured in Japan.
RI Life Sciences
RI Life Sciences (RI LS) specializes in micromanipulation, laser ablation, and cell handling technologies for
life sciences applications. RI LS is proud to be a division of Research Instruments Limited, a world leader in
the design and manufacture of specialist hardware and
consumables for the human ART field.
351 Asahi-machi
Kasama City, Ibaraki 309-1717, Japan
Phone: +81 296 77 8804
http://www.misawa-medical.co.jp/English/intro.html
Booth: 9
MOFA Global
MOFA Global, a subsidiary of CRI International, is a
leading provider of assisted reproduction technologies
for porcine, bovine, equine, and canine. We are a company active in research and product development and
have produced many products that are considered industry standards. Visit the MOFA Global booth to learn
more about the available products for artificial insemination, ovum pickup, in vitro fertilization, embryo
transfer, embryo cryopreservation, and embryo sexing
technologies.
419 Venture Ct.
Verona, WI 53593, USA
Phone: 800-646-4882
http://www.mofaglobal.com
Booth: 3
Partnar Animal Health
Partnar Animal Health is pleased to present its range
of embryo transfer and OPU products. We will present information on Stimufol (pFSH) and eMP3, our
own range of embryo flush, hold, and freeze media. For
OPU, we also have the Ibex Portable Ultrasound with
an OPU probe, aspiration pumps, and retrieval needles
and the MicroQ controlled temperature shipping device
for oocyte, embryo, and fresh extended sexed semen
transport.
2014 Holland Ave, Unit 227
Port Huron, MI 48060, USA
Phone: 519-666-0033
http://www.partnaranimalhealth.com
Booth: 12
Professional Embryo Transfer Supply Inc. (PETS)
PETS has been a world leading embryo transfer supply
company in the bovine and equine industries for almost
three decades. Our goal all this time has been your success, and we work every day to achieve this with quality
service and ET supplies from ICPbio, Vetoquinol, MAI,
SPI, Wesco, NovaVive, and more. Come visit with us
for more details.
56
Visit stand 8 to try the Saturn 5 Active Laser System,
offering a movable target that ablates cells with submicron accuracy. Demo the Integra 3 micromanipulator
featuring Thermosafe, which maintains sample temperature uniformly across the dish. See our extensive consumables range including glass micropipettes and the
EZ-Range of cell handling devices.
Bickland Industrial Park
Falmouth, Cornwall, United Kingdom TR11 4TA
Phone: +44(0)1326 372753
Fax: +44(0)1326 378783
www.research-instruments.com
Booth: 8
STEM ALPHA
Cryo.3 is an animal-component-free medium for freezing and thawing cells, designed and produced by STEM
ALPHA, a French company. Cryo.3 has been designed
for improving preservation in low temperature (−80°C
to −196°C). Cryo.3 provides a safe protective environment for cells and tissues during the freezing, storage,
and thawing processes. Cryo.3 is animal component free
and Dextran free.
La Chenevatière - RD389
69610 Saint Genis l’Argentière, France
http://www.stemalpha.com
Booth: 20
Vetoquinol
In April 2014 Bioniche Animal Health, the manufacturer
of Folltropin, became the newest division of Vetoquinol.
Vetoquinol is a family-owned, independent company
devoted exclusively to animal health. Our product
portfolio is divided between livestock and companion animals and includes most therapeutic categories.
Vetoquinol trades throughout Europe, the Americas,
Africa, the Middle East, and Asia Pacific. With the
acquisition of Bioniche Animal Health, Vetoquinol is
committed to servicing the assisted reproduction industry with its long-lasting tradition of excellence.
42nd Annual Conference
BP 189
70204 Lure Cedex, France
Phone: +33 3 84 62 55 55
Fax: +33 3 84 62 55 56
www.vetoquinol.com
Booth: 10 & 11
WTA sells throughout Brazil and in many countries,
being recognized as one of the leading companies in the
market.
WTA Technologies LLC
WTA, Watanabe Tecnologia Aplicada, is a Brazilian
technology-based company, focused on products for
animal assisted reproduction, offering high added value
solutions for ovum pickup (OPU), in vitro fertilization
(IVF), embryo transfer (ET), and artificial insemination
(AI). Our products are mainly focused on cattle, horses,
and small ruminants.
Each product is designed to provide security, economy,
and the very best result, always focusing on animal
health. Every piece has a precise design, quality materials, and fine workmanship.
WTA, Brazil: + 55 16 39518161
Sales, Brazil: + 66 16 99762-7777
Sales, USA: + (979) 324-6168
http://www.wtavet.com.br
Booth: 5 & 6
International Embryo Transfer Society
57
Thank You to Our Exhibitors
58
42nd Annual Conference
International Embryo Transfer Society
59
60
42nd Annual Conference
International Embryo Transfer Society
61
Don't miss out on this opportunity!
Come and join us for the event of the week. Share a meal, enjoy the surroundings, and get reacquainted with old
friends. There will be music for your listening and dancing pleasure. You will also have a chance to tour the museum.
With two floors of interactive, family-friendly exhibits, the Kentucky Derby Museum takes visitors through every
stage of a Thoroughbred’s life, from birth to the first Saturday in May. Learn about all the great history of the race
dating back to the inaugural running in 1875 and about our most recent Kentucky Derby champion. Experience all
the traditions that have transformed the race into what it is today. You will also have a chance to view the new stateof-the-art 360° Greatest Race Experience. Don’t miss this event. Shuttle service to and from the Galt House to the
museum will be provided, beginning at 18:00, from the Galt House Hotel, outside of the Suite Tower, with return
service beginning at 19:30. Tickets are required for this event.
62
42nd Annual Conference
Preconference Symposium
42nd Annual Conference of the IETS
Louisville, Kentucky
Biomaterials Repositories: The Science and Business of Biobanking
Dedicated to Naida Loskutoff for her tireless efforts and significant contribution to the advancement of
reproductive sciences in companion animals, non-domestic and endangered species
Saturday, January 23, 2016
Galt House Hotel (Jones)
Program Co-Chairs:
Nucharin Songsasen and Gabriela Mastromonaco
08:30–08:45 Opening and Welcome
Nucharin Songsasen and Gabriela Mastromonaco
08:45–09:30 Keynote Speaker
Cryobiology principles and species conservation: Benefits for coral reefs
Mary Hagedorn, Smithsonian Conservation Biology Institute, Hawaii Institute of Marine Biology,
Hawaii, USA
Session 1: Emerging Techniques for Germplasm Cryopreservation
Session chair: Ann Van Soom, Ghent University, Ghent, Belgium
09:30–09:55 Dry-state preservation of mammalian sperm
Takehito Kaneko, Graduate School of Medicine, Kyoto University, Kyoto, Japan
09:55–10:20 Cryopreservation of mouse oocytes and embryos by vitrification and laser-induced ultra-rapid warming
Peter Mazur, College of Arts and Sciences, University of Tennessee, Knoxville, USA
10:20–10:45 Micromanipulation of equine blastocysts to allow vitrification
Katrin Hinrichs, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University,
College Station, USA
10:45–11:05Break
Session 2: Biobanking in Practice
Session chair: Jason Herrick, National Foundation for Fertility Research, Colorado, USA
11:05–11:30
Oocyte banking
Zsolt Peter Nagy, Reproductive Biology Associates, My Egg Bank, Atlanta, USA
11:30–11:55
Establishing a biobank to support biomedical research at a veterinary hospital
Marta Castelhano, Cornell Veterinary Biobank, Cornell University, Ithaca, USA
11:55–12:20 Implementing the use of biobanks: Lessons learned from the endangered black-footed ferret
Rachel Santymire, Davee Center for Epidemiology and Endocrinology, Lincoln Park Zoo,
Chicago, USA
12:20–13:30 Lunch (on your own)
International Embryo Transfer Society
63
13:30–14:15 Keynote Speaker
Biobanking rare breeds: Challenges and implementation
Phillip H. Purdy, USDA-ARS National Animal Germplasm Program, Fort Collins, USA
Session 3: Unique Challenges to Cryopreservation
Session chair: William Swanson, Cincinnati Zoo and Botanical Garden, Cincinnati, USA
14:15–14:40 Repository development for germplasm of aquatic species
Terrence Tiersch, Aquatic Germplasm and Genetic Resources Center, Louisiana State University
Agricultural Center, Baton Rouge, USA
14:40–15:05 Cryopreservation and associated technologies for threatened amphibians and reptiles
John Clulow, School of Environmental and Life Sciences, University of Newcastle, New South Wales,
Australia
15:05–15:30 Sperm cryopreservation in wild ungulates
Budhan Pukazhenthi, Smithsonian Conservation Biology Institute, Front Royal, USA
15:30–15:50Break
Session 4: Management of Biospecimens
Session chair: Gabriela Mastromonaco, Toronto Zoo, Toronto, Canada
15:50–16:15 Implications of the Nagoya Protocol
Pierre Comizzoli, Smithsonian Conservation Biology Institute, Office of the Under-Secretary for
Science, Washington, DC
16:15–17:15 Roundtable Discussion
Future of biobanking: What should we become?
Z. Nagy, M. Castelhano, P. Purdy, B. Pukazhenthi, and P. Comizzoli
17:15–17:30Closing
64
42nd Annual Conference
Thank You to Our Sponsors
Platinum Level
International Embryo Transfer Society
65
Bronze Level
66
42nd Annual Conference
International Embryo Transfer Society
67
Bronze Level
(continued)
Friend Level
68
42nd Annual Conference