50th Annual Meeting

Transcription

50th Annual Meeting
ACNP
50TH
Annual
Meeting
December 4-8, 2011
Hilton Waikoloa Village
Waikoloa, Hawaii
President: Eric J. Nestler, M.D., Ph.D.
Program Committee Chair: William A. Carlezon, Ph.D.
Program Committee Co-Chair: Anissa Abi-Dargham, M.D.
ACNP
AMERICAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY
50th
ANNUAL MEETING
FINAL
PROGRAM
Waikoloa, Hawaii
Hilton Waikoloa Village
DECEMBER 4-8, 2011
Disclosures for 2011 speakers (panel, mini-panel, study group, and plenary) and poster
presenters may be found online at: www.acnp.org (click the Annual Meeting tab)
69' x 36'
Kohala 1
69' x 36'
Kohala 2
69' x 36'
Water’s Edge Ballroom
King's 3
BOARDROOM
ii
Lagoon
Lagoon
Lanai
35' x 55'
LOUNGE
Queen's 4
36' x 55'
Queen's 5
37' x 55'
Queen's 6
35' x 55'
Grand Promenade
95' x 134'
King's 1
36' x 55'
King's 2
37' x 55'
Monarchy
120' x 206'
Kohala 3
Grand Ballroom
69' x 36'
HILTON WAIKOLOA VILLAGE
Function and Meeting Space
Kohala 4
Kohala Ballroom
21' x 36'
28' x 36'
Kona 2 Kona 3
Waikoloa Suites
Waikoloa 1 Waikoloa 2 Waikoloa 3
31' x 23' 31' x 23' 31' x 23'
30' x 36'
Kona 1
80' x 36'
Kona 4
80' x 36'
Kona 5
Kona Ballroom
Grand
Staircase
up
To
Main Lobby
ACNP 50th Annual Meeting • Final Program
MEETING SPACE MAP
KONA POOL SLIDE
KOHALA TENNIS GARDEN
KAMEHAMEHA COURT
iii
GUEST PARKING
W A T E R ’ S
AD
MEN
CH
BEA
E
OON
E D G E
RO
ND P
GRA
LAGOON LANAI
WATER'S EDGE
BOARDROOM
A T
LAGOON
LAG
LAUNDERETTE
1st FLOOR
CAMP MENEHUNE
2nd FLOOR
TO WAIKOLOA BEACH GOLF COURSE
KINGS' SHOPS AND QUEENS’ MARKETPLACE
WEDDING
CHAPEL
GAZEBO
PALACE GARDENS
• Complimentary Golf Shuttle departs from the Lower Lobby.
• Complimentary Beach and Resort Shuttle departs from the Main Lobby.
• Please see your Concierge or call extension 53 for accessibility assistance
and stroller access around the resort.
TO WAIKOLOA KINGS'
GOLF COURSE
KOHALA POOL SLIDE
KOHALA POOL
KOHALA POOL BAR
KONA
KOHALA
MONARCHY
BALLROOMS
BALLROOM BALLROOMS
WAIKOLOA
QUEEN'S
KING'S
SUITES
BALLROOM
BALLROOM
GRAND
STAIRCASE
DOLPHIN
LEARNING
CENTER
LOWER LOBBY
(Downstairs from Main Lobby)
JAPANESE SERVICE DESK
WAIKOLOA BEACH DRIVE
MAIN LOBBY
REGISTRATION/CHECK-OUT
DISCOVERING HAWAII
BUDGET RENT-A-CAR
BUSINESS CENTER
LAUNDERETTE
4th FLOOR
THE GALLERY
SUNDRY SHOP
RETAIL SHOPPING
KOHALA
SPA
HILTON GRAND
VACATIONS
SALES GALLERY
TENNIS SHOP
KIMO BEAN
COFFEE BAR
LAGOON TOWER
OCEAN SPORTS
DOLPHIN QUEST VILLAGE
CHECKER BOARD
INTERACTIVE FOUNTAIN
CHILDREN'S SAND POOL
KONA POOL
OCEAN VIEW
TERRACE
JAPANESE STEAK HOUSE
PALACE
TOWER
SUNDRY SHOP
PALACE LAWN
OCEAN
TOWER
C
OU
RT
RESTAURANTS
POOLS
RECREATION DESK
TRAM STOP
BOAT STOP
WALKWAY
TRAM
BOATWAY
TOWERS
ELEVATORS
STAIRWAYS
RESTROOMS
CONCIERGE
2nd FLOOR
1st FLOOR
SUNDRY SHOP
KIMO BEAN COFFEE BAR
4_10
www.HiltonWaikoloaVillage.com
ISLAND ORIENTATION
4th FLOOR OCEAN TOWER
HILTON GRAND VACATIONS
SALES GALLERY
GUEST WELCOME CENTERS
DISCOVERING HAWAII
WAIKOLOA SUNSET
ATM
CONVENTION CENTER
RECREATION DESK
OCEAN TOWER POOL
D
FOO
KAMUELA PROVISION
COMPANY
KONA POOL BAR
WAIULUA BAY
BUDDHA POINT
ACNP 50th Annual Meeting • Final Program
PROPERTY MAP
ACNP 50th Annual Meeting • Final Program
Program at a Glance
Saturday, December 3, 2011
8:00 am – 3:00 pm
ACNP Council Meeting
Monday, December 5, 2011
Queen’s 4
1:30 pm – 3:00 pm Monarchy Ballroom
Distinguished Lecture - Louis Ptáček
8:00 am – 5:00 pm
King’s 1
ACNP Membership Committee Meeting
Panel Sessions
3:00 pm – 5:30 pm
Monarchy
The Noradrenergic System as a
Therapeutic Target for Drug Dependence
9:00 am – 5:00 pm
Kona 3
CINP Executive Committee Meeting
2:00 pm – 3:30 pm
King’s 2
Neuropsychopharmacology EIC & Deputy
Editors Meeting
3:00 pm – 5:30 pm
Kona 4
Striving for the Correct Diagnosis
of Mental Health Disorders
3:30 pm – 5:00 pm
King’s 2
ACNP Publications Committee Meeting
3:00 pm – 5:30 pm
Kona 1-3
Genetic and Molecular Mechanisms
of Normal Cognitive Aging
4:00 pm – 5:30 pm
ACNP Ethics Committee Meeting
Water’s Edge
Boardroom
3:00 pm – 5:30 pm
Kona 5
Memory Erasure: Mechanisms and
Potential Utility in Psychiatry
5:00 pm – 7:00 pm
Kona 2
ACNP Public Information Committee
3:00 pm – 5:30 pm
Kohala 3
Enteric Hormone Modulation of Cerebral
Neurotransmission and Eating Behaviors
in Obesity
Sunday, December 4, 2011
7:00 am – 8:30 am
Water’s Edge
Travel Awardee Breakfast Ballroom
(by invitation only)
8:30 am – 11:30 am
Monarchy
Neurpsychopharmacology Ballroom
Reviews Plenary:
Neurotherapeutics Teaching Day
11:30 am – 1:00 pm
Past Presidents’ Luncheon
Queen’s 4
11:30 am – 1:00 pm
Kona 1
ACNP Program Committee Meeting
11:30 am – 1:00 pm
Kona 3
ACNP Education & Training Committee
Meeting
3:00 pm – 5:30 pm
Queen’s 5-6
NMDA Receptor Complexes: A Point of
Convergence for Schizophrenia Candidate
Pathways
3:00 pm – 5:30 pm
Kohala 4
Adolescent Brains: The Constancy of
Change
5:30 pm – 7:30 pm Kohala 1-2, King’s &
Poster Session I
Grand Promenade
with Reception
Study Groups
7:30 pm – 9:00 pm
Kona 1-3
Assessing Brain Developmental
Trajectories from Infancy to Adulthood
1:00 pm – 2:30 pm
NIH Institutes Update
Monarchy
7:30 pm – 9:00 pm
Kona 4
Ethical, Legal, and Social challenges in
Research on Psychiatric Genetics
2:30 pm – 4:00 pm
History Lecture:
Neuropsychopharmacology,
The Past 50 Years
Monarchy
7:30 pm – 9:00 pm
PTSD Biomarkers
4:00 pm – 6:30 pm
Hot Topics - Basic
Kona 5
4:00 pm – 6:30 pm
Hot Topics - Clinical
Kona 4
7:00 pm – 9:00 pm Monarchy, Queen’s
50th Anniversary & Grand Promenade,
Celebration Gala
Grand Staircase &
Lagoon Lanai
Monday, December 5, 2011
Monarchy
7:30 pm – 9:00 pm
Queen’s 5-6
Can Vulnerability Markers Identify
Informative Neurodevelopmental
Abnormalities Across the Spectrum of
Early Psychosis?
7:30 pm – 9:00 pm
Kona 5
Crisis in Psychiatric Drug Discovery:
Solutions from Academia, Government
and the Advocacy Community
7:30 pm – 9:00 pm
Kohala 4
Utilizing the NIH’s CTSA Network to
Advance Neuropsychopharmacology
Research
6:30 am – 8:00 am
SOBP Editorial Board Meeting
Queen’s 4
7:30 pm – 9:00 pm
Kohala 3
The Alcohol Clinical Trials Initiative
(ACTIVE): Progress Report and Feedback
8:00 am – 11:30 am
President’s Plenary: Brave New World for
Brain Therapeutics
Monarchy
Ballroom
7:30 pm – 9:00 pm
Queen’s 4
Four Rodent Models of Psychosis:
(Not) Lost in Translation
12:00 pm – 1:30 pm
Women’s Luncheon
Water’s Edge
Ballroom
Tuesday, December 6, 2011
12:00 pm – 1:30 pm
Kohala 4
Teaching Neuropsychopharmacology
12:00 pm – 1:30 pm
Queen’s 4
ASCP Board of Directors Meeting
7:00 am – 8:30 am
Liaison Committee Meeting
Water’s Edge
Ballroom
7:00 am – 8:30 am
Imari
American Journal of Psychiatry Editorial
Board Meeting
iv
Tuesday, December 6, 2011
7:00 am – 8:30 am
Membership Advisory
Task Force Meeting
7:00 am – 8:30 am
NPPR Editors Meeting, Volumes 6 & 7
Queen’s 4
Water’s Edge
Boardroom
7:30 am – 8:30 am
Donatonio’s
ACNP Leadership & Institute Directors
Mini-Panel Sessions
8:30 am – 9:45 am
Monarchy
Medication Discovery for Addiction:
Translating the Dopamine D3 Receptor
Hypothesis
9:45 am – 11:00 am
Monarchy
Vaccines, Viral Vectors, and Cocaine
Addiction: Neutralizing Cocaine before
it gets to the Brain
Panel Sessions
8:30 am – 11:00 am
Kona 5
Cortical Dopamine in Schizophrenia:
Quantifying Levels, Understanding
Function
8:30 am – 11:00 am
Kohala 4
Synaptic Plasticity: From Adaptive
Molecular Mechanisms to Dysregulation
in Psychiatric Disorders
8:30 am – 11:00 am
Kona 1-3
Neuroimaging Genomics: Discovering
a Signal in the Complexity of Genes,
Brain and Behavior
8:30 am – 11:00 am
Kona 4
Feast or Famine: Is Disordered
Eating Related to Disordered Reward?
8:30 am – 11:00 am
Queen’s 5-6
Emerging Methods to Examine
Fear Regulation
8:30 am – 11:00 am
Kohala 3
Circadian Rhythms, Sleep
Deprivation and Mood Disorders
11:00 am – 12:30 pm
Donatonio’s
ACNP Corporate Liaison Luncheon
(by Invitation only)
11:30 am – 1:30 pm
Data Blitz Session
11:30 am – 1:30 pm
Faculty Research Fellowship
Presentations
11:30 am – 1:30 pm
Travel Awardee Research Presentations
Kona 4
Kohala 3
Kona 1-3,
Kohala 4,
Queen’s 5-6
1:30 pm – 3:00 pm
Kona 5
Issues in Ethics Plenary: The Perils
and Pitfalls of Biomedical Research:
Historical and Contemporary
Perspectives on the Ethics of Research
Panel Sessions
3:00 pm – 5:30 pm
Kona 4
Epigenetic Modifications in Development,
Aging and Mental Illness
3:00 pm – 5:30 pm
Kona 5
Molecular Mechanisms Informing
PTSD Risk, Treatment and Prophylaxis
ACNP 50th Annual Meeting • Final Program
Program at a Glance
Tuesday, December 6, 2011
Wednesday, December 7, 2011
Thursday, December 8, 2011
3:00 pm – 5:30 pm
Kona 1-3
New Directions in Understanding the
Neurocircuitry of Choice, Value, and
Decision-Making
8:30 am – 11:00 am
Kohala 4
Progress in Understanding the Role
of GABA and GABAA Receptor Biology
in Psychiatric Disease
7:00 am – 8:00 am
Queen’s 4
ACNP/AsCNP/CINP/ECNP/JSNP Meeting
3:00 pm – 5:30 pm
Kohala 3
A Convergence in Autism and
Schizophrenia Genetics: The Conundrum
of Shared Risks and Divergent Outcomes
11:15 am – 12:30 pm
Monarchy
ACNP Business Meeting (ACNP Fellows,
Members, and Associate Members Only)
Mini Panel Sessions
8:00 am – 9:15 am
Kona 5
The Use of Intraoperative Techniques to
Assess the Physiology of the Anterior
Cingulate Cortex
12:30 pm – 2:00 pm
Queen’s 4
SOBP Program Committee Meeting
9:15 am – 10:30 am
Kona 5
Genes, Fear and Anxiety: From Mice to Humans
12:30 pm – 2:00 pm
Water’s Edge
Travel Awardee Luncheon Ballroom
(by Invitation Only)
Panel Sessions
8:00 am – 10:30 am
Monarchy
Glutamate Targets for CNS Therapy: Insights
Obtained from a Potential Dynamic Duo
3:00 pm – 5:30 pm
Kohala 4
Neurodevelopmental Pathology of Cortical
Interneurons in Schizophrenia: Is it the
Journey or the Destination that Matters?
3:00 pm – 5:30 pm
Monarchy
Will We Have New Drugs or Not?
Addressing the Crisis in Neuropsychiatric
Drug Discovery
3:00 pm – 5:30 pm
Queen’s 5-6
Toward a Neuroimmune-Medicated
Subtype of Autism Spectrum Disorders
5:30 pm – 7:30 pm
Kohala 1-2,
Poster Session II with King’s & Grand
ReceptionPromenade
6:00 pm – 11:00 pm
ACNP Committee Chairs Waiting Room
Water’s Edge
Boardroom
6:00 pm – 11:00 pm
ACNP Council Meeting
Queen’s 4
7:30 pm – 9:00 pm
Water’s Edge
Neuropsychopharmacology Ballroom
Editorial Board
Wednesday, December 7, 2011
12:30 pm – 2:00 pm
Donatonio’s
SIRS Industry Task Force Meeting
12:30 pm – 2:00 pm
Queen’s 5-6
ACNP Special Session: “Ask the Experts”
Career Development Program
Mini Panel Sessions
3:00 pm – 4:15 pm
Kona 5
Downstream Effects of Visual and
Auditory Perceptual Impairment in
Schizophrenia
Panel Sessions
3:00 pm – 5:30 pm
Kohala 4
From Genome to Marco-Connectome:
Integrating High-Dimensional Genetic,
Imaging and Behavioral Data, with
Application to Large-Scale Studies of
Alzheimer’s
8:00 am – 10:30 am
Kohala 4
Serotonin Signaling During Development:
Unexpected sources, Large Neuron
Heterogeneity, Limited System Plasticity
and Big Impact on Physiology and Behavior
Panel Sessions
8:30 am – 11:00 am
Role of Phagocytes in Synaptic
Plasticity and Remodeling of
Tissues in the Nervous System
3:00 pm – 5:30 pm
Kona 1-3
The Autism Sequencing Consortium (ASC):
Unraveling the Genetic and Functional
Architecture of Autism Spectrum
Disorders
8:30 am – 11:00 am
Monarchy
Novel Approaches to Therapeutic
Development in Alzheimer’s Disease
3:00 pm – 5:30 pm
Kona 4
Neural Mechanisms of Environmental
Risk for Psychiatric Disorders
3:00 pm – 5:30 pm
Queen’s 5-6
Gimme Another Hit of Chocolate.
Is Food Addictive?
8:30 am – 11:00 am
Kona 4
Translational Approaches to
Understanding Negative Symptoms
3:00 pm – 5:30 pm
Kohala 3
Drug of Abuse during Adolescence:
A Development Period of Vulnerability
or Resilience?
8:30 am – 11:00 am
Queen’s 5-6
The Development of Novel Pain
Therapeutics: New Strategies to
Overcome Drug Discovery Barriers
5:30 pm – 7:30 pm
Poster Session III with Reception
8:30 am – 11:00 am
Kona 5
Novel Functions of Prefrontal Cortex
Regions in Motivated Behavior:
Implication for Psychiatric Disorders
8:00 am – 10:30 am
Queen’s 5-6
The Putative Role of ER Stress in
Neuropsychiatric Illnesses
8:00 am – 10:30 am
Kohala 3
Sex Difference in Brain and Behavior:
Emerging Genetic and Cellular Mechanisms
3:00 pm – 5:30 pm
Monarchy
Novel Synaptic Targets in Depression
Emerging from Clinical, Biochemical,
and Circuit Based Approaches
8:30 am – 11:00 am
Kona 1-3
Neuroactive Cytokines: Critical
Therapeutic Targets for Depression
and Treatment Resistant Depression?
8:00 am – 10:30 am
Kona 4
Rapid Acting Antidepressants Increase
Synaptogenesis
Kona 5
4:15 pm – 5:30 pm
GABA, Glutamate and Neural
Synchrony in Schizophrenia
7:00 am – 8:30 am
Water’s Edge
ACNP Women’s Task Force
Boardroom
Meeting
Kohala 3
8:00 am – 10:30 am
Kona 1-3
Beyond Genome - Wide Association Studies:
New Approaches to Risk of Psychiatric Illness
Kohala 1-2,
King’s & Grand
Promenade
7:30 pm – 9:00 pm
Monarchy
ACNP Special Session: An Oral
History of Neuropsychopharmacology
9:00 am – 12:00 pm
ACNP Council Meeting
Queen’s 4
Panel Sessions
12:00 pm – 2:30 pm
Monarchy
APOE and Alzheimer’s Disease:
Neurosusceptibility, Neuroprotection and
New Treatments
12:00 pm – 2:30 pm
Kona 1-3
From Transcription to Oscillations:
How Sick Interneurons create a
Schizophrenia-Like Phenotype
12:00 pm – 2:30 pm
Kona 5
Is Love Epigenetic? Transformative Effects
of Social Experiences and of Oxytocin
12:00 pm – 2:30 pm
Kona 4
Contribution of Genetic Epidemiology to
Identifying Genetic and Environmental
Risk Factors for Neurologic and
Psychiatric Disorders
12:00 pm – 2:30 pm
Queen’s 5-6
Enhancing Cognitive Performance: Molecular,
Pharmacological, and Experiential Strategies
12:00 pm – 2:30 pm
Kohala 2
Functional Connectivity in Neural Systems
as a Developmental Abnormality in
Creating Risk for Bipolar Disorder
12:00 pm – 2:30 pm
Kohala 3
Optogenetic Dissection of Cortico-Limbic
Circuit Function and Dysfunction
12:00 pm – 2:30 pm
Kohala 4
Translating Pharmacogenetics into Clinical
Utility: Optimizing the Phenotype
v
Disclosures for 2011 speakers (panel, mini-panel, study group, and plenary) and poster
presenters may be found online at: www.acnp.org (click the Annual Meeting tab)
Hotel Maps. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ii
Acknowledgments. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xii
Council. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Program Committee . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Dates and Location . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Program Book. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Itinerary Planner. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Executive Office. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Continuing Medical Education (CME) and Continuing Education (CE). . . . . . . 3
Meeting Evaluation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Scientific Program. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Videotaping Sessions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Neuropsychopharmacology Reviews Plenary. . . . . . . . . . . . . . . . . . . . . . . . . 6
NIH Institutes Update. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
History Lecture. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Hot Topics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
President’s Plenary. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Distinguished Lecture. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Data Blitz Session. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Travel Awardee Research Presentations. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Faculty Research Fellowship Presentations. . . . . . . . . . . . . . . . . . . . . . . . . . 7
Issues in Ethics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Panels (times and dates). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Mini-Panels (times and dates) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Study Groups (times and dates). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Poster Sessions (times and dates). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Registration. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Hotel Facilities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Services. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Refreshment Breaks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Luncheons. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
ACNP Anniversary Celebration. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Special Events. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
College Meetings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Council Meetings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Committee/Task Force Meetings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
ACNP Sanctioned Meetings. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Future ACNP Annual Meetings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
In Memoriam . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
General
Information
Table Of Contents
General
Information
Program Listings
Sunday, December 4th
Neuropsychopharmacology Reviews Plenary. . . . . . . . . . . . . . . . . . . . . . . . . .
Institute Directors Briefing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
History Lecture. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Hot Topics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
19
30
31
33
Monday, December 5th
President’s Plenary. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
Teaching Neuropsychopharmacology. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90
Distinguished Lecture. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
Afternoon Panel Sessions
• The Noradrenergic System as a Therapeutic Target
for Drug Dependence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
• Genetic and Molecular Mechanisms of Normal Cognitive Aging. . . . . 95
• Striving for the Correct Diagnosis of Mental Health Disorders. . . . . . . 96
• Memory Erasure: Mechanisms and Potential Utility in Psychiatry. . . . 97
• NMDA Receptor Complexes:
A Point of Convergence for Schizophrenia Candidate Pathways. . . . . . 98
• Enteric Hormone Modulation of Cerebral Neurotransmission
and Eating Behaviors in Obesity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99
• Adolescent Brains: The Constancy of Change. . . . . . . . . . . . . . . . . . . 100
Study Groups
• PTSD Biomarkers Study Group. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
• Assessing Brain Developmental Trajectories
from Infancy to Adulthood. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
• Ethical, Legal, and Social Challenges in Research
on Psychiatric Genetics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
• Crisis in Psychiatric Drug Discovery: Solutions from
Academia, Government and the Advocacy Community. . . . . . . . . . . . 102
• Can Vulnerability Markers Identify Informative Neurodevelopmental
Abnormalities across the Spectrum of Early Psychosis?. . . . . . . . . . . 103
• The Alcohol Clinical Trials Initiative (ACTIVE):
Progress Report and Feedback. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104
• Utilizing the NIH’s CTSA Network to Advance
Neuropsychopharmacology Research. . . . . . . . . . . . . . . . . . . . . . . . . . 104
• Four Rodent Models of Psychosis: (Not) Lost in Translation . . . . . . . 105
ACNP 50th Annual Meeting • Final Program
Tuesday, December 6th
Mini Morning Panel Sessions
• Medication Discovery for Addiction: Translating the Dopamine D3 Receptor Hypothesis . . . . . . . . . . . . . . 107
• Vaccines, Viral Vectors, and Cocaine Addiction: Neutralizing Cocaine before it gets to the Brain. . . . . . . . . . . . . . . . . . 108
Morning Panel Sessions
• Cortical Dopamine in Schizophrenia: Quantifying Levels, Understanding Function. . . . . . . . . . . . . . . . . . . .
• Synaptic Plasticity: From Adaptive Molecular Mechanisms to Dysregulation in Psychiatric Disorders. . . . . . . . . . . .
• Neuroimaging Genomics: Discovering a Signal in the Complexity of Genes, Brain and Behavior. . . . . . . . . . . . . . . . .
• Feast or Famine: Is Disordered Eating Related to Disordered Reward? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
• Emerging Methods to Examine Fear Regulation. . . . . . . . . . . . . . . . .
• Circadian Rhythms, Sleep Deprivation and Mood Disorders . . . . . . .
Data Blitz Session
• DREADDed Decision-Making: Revealing a Role for the ‘Direct’ Pathway in Reward Preference. . . . . . . . . . . . . . . . . . . . . . . .
• A Functional Role for Interleukin 6 in Susceptibility to Depression. .
• The Impact of Placebo on IL-18 and its Relation to Analgesic
Expectation and Central µ-Opioid Receptor Activation. . . . . . . . . . . .
• Motivational Saliency Signal in Ventral Striatum is Modulated by Genetic Variation in the ARC Gene Region . . . . . . . . .
• Dopamine Transporter Knockdown Mice Exhibit Poorer Within-Session Risk Learning in a Mouse Iowa Gambling Task Consistent With Bipolar Mania Patients. . . . . . .
• Evidence that Mutation in Neuregulin 1, a Schizophrenia Susceptibility Gene, Alters Glucose Tolerance in Animals . . . . . . . . .
• A Multi-Center Investigation of Folate plus B12 Supplementation in Schizophrenia. . . . . . . . . . . . . . . . . . . . . . . . . . . .
• A Zebrafish Model for the Functional Analysis of Genes in Autism. .
• Sensory and Motor Contributions to Visuomotor Impairments in Individuals with Autism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
• The Role of Orexin in Adverse Menopause-Associated “Hot Flash” and Anxiety Symptoms . . . . . . . . . . . . . . . . . . . . . . . . . .
vii
109
110
111
112
113
114
117
119
121
123
125
127
129
131
134
136
ACNP 50th Annual Meeting • Final Program
Tuesday, December 6th (continued)
• Progression of Drug Cue-Induced Phasic Dopamine Release from Limbic to Sensorimotor Striatum Mediates Action Selection of
Drug-Taking Behavior in a Rodent Model of Drug Addiction. . . . . . . 138
• Mechanisms underlying Hippocampal Dysfunction in Schizophrenia and Related Psychotic Disorders. . . . . . . . . . . . . . . . . . 140
Travel Awardee Research Presentations
• Basic Neuroscience of Addiction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142
• Basic Neuroscience of Depression, Anxiety & Stress . . . . . . . . . . . . . 148
• Clinical and Translational Research. . . . . . . . . . . . . . . . . . . . . . . . . . . 155
Faculty Research Fellowship Presentations. . . . . . . . . . . . . . . . . . . . . . . . . 163
Issues in Ethics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170
Afternoon Panel Sessions
• Epigenetic Modifications in Development, Aging and Mental Illness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177
• Molecular Mechanisms informing PTSD Risk, Treatment and Prophylaxis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178
• New Directions in Understanding the Neurocircuitry of Choice, Value, and Decision-Making. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179
• A Convergence in Autism and Schizophrenia Genetics: The Conundrum of Shared Risks and Divergent Outcomes. . . . . . . . . 180
• Neurodevelopmental Pathology of Cortical Interneurons in Schizophrenia:
Is it the Journey or the Destination that Matters?. . . . . . . . . . . . . . . . . 181
• Will We Have New Drugs or Not? Addressing the Crisis in Neuropsychiatric Drug Discovery. . . . . . . . . 182
• Toward a Neuroimmune-Mediated Subtype of Autism Spectrum Disorders. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183
viii
ACNP 50th Annual Meeting • Final Program
Wednesday, December 7th
Morning Panel Sessions
• Role of Phagocytes in Synaptic Plasticity and Remodeling of Tissues in the Nervous System. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
• Neuroactive Cytokines: Critical Therapeutic Targets for Depression and Treatment Resistant Depression?. . . . . . . . . . . . . . . . . . . . . . . . . . 186
• Novel Approaches to Therapeutic Development in Alzheimer’s Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 187
• Translational Approaches to Understanding Negative Symptoms . . . 188
• The Development of Novel Pain Therapeutics: New Strategies to Overcome Drug Discovery Barriers. . . . . . . . . . . . 189
• Novel Functions of Prefrontal Cortex Regions in Motivated Behavior: Implication for Psychiatric Disorders . . . . . . . . . . . . . . . . . 190
• Progress in Understanding the Role of GABA and GABAA Receptor Biology in Psychiatric Disease . . . . . . . . . . . . . . . . . . . . . . . 191
ACNP Special Session:
“Ask the Experts” Career Development Program. . . . . . . . . . . . . . . . . . . . 192
Mini Afternoon Panel Sessions
• Downstream Effects of Visual and Auditory Perceptual Impairment in Schizophrenia. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
• GABA, Glutamate and Neural Synchrony in Schizophrenia. . . . . . . . 194
Afternoon Panel Sessions
• From Genome to Macro-Connectome: Integrating High-Dimensional Genetic, Imaging and Behavioral Data, with Application to Large-Scale Studies of Alzheimer’s Disease, Schizophrenia, and Substance Abuse. . . . . . . . . . . . . . . . . . . . . . . . . . 195
• Novel Synaptic Targets in Depression Emerging from Clinical, Biochemical and Circuit Based Approaches . . . . . . . . . . . . . . . . . . . . 196
• The Autism Sequencing Consortium (ASC): Unraveling the Genetic and Functional Architecture of Autism Spectrum Disorders . . . . . . . . 197
• Neural Mechanisms of Environmental Risk for Psychiatric Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198
• Gimme Another Hit of Chocolate. Is Food Addictive?. . . . . . . . . . . . . 199 • Drug of Abuse during Adolescence: A Development Period of Vulnerability or Resilience?. . . . . . . . . . . . 200
Special Session: An Oral History of Neuropsychoparmacology. . . . . . . . . 201
ix
ACNP 50th Annual Meeting • Final Program
Thursday, December 8th
Morning Mini Panel Sessions
• The Use of Intraoperative Techniques to Assess the Physiology of the Anterior Cingulate Cortex . . . . . . . . . . . . . . . . . 203
• Genes, Fear and Anxiety: From Mice to Humans . . . . . . . . . . . . . . . . 204
Morning Panel Sessions
• Glutamate Targets for CNS Therapy: Insights Obtained from a Potential Dynamic Duo. . . . . . . . . . . . . . . .
• Beyond Genome-Wide Association Studies: New Approaches to Risk of Psychiatric Illness . . . . . . . . . . . . . . . . . .
• Rapid Acting Antidepressants increase Synaptogenesis. . . . . . . . . . . .
• The Putative Role of ER Stress in Neuropsychiatric Illnesses. . . . . . .
• Sex Differences in Brain and Behavior: Emerging Genetic and Cellular Mechanisms. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
• Serotonin Signaling during Development: Unexpected Sources, Large Neuron Heterogeneity, Limited System Plasticity and Big Impact on Physiology and Behavior. . . . . . . . . . . . . . . . . . . .
Afternoon Panel Sessions
• APOE and Alzheimer’s Disease: Neurosusceptibility, Neuroprotection and New Treatments . . . . . . . . . . . . . . . . . . . . . . . . .
• From Transcription to Oscillations: How Sick Interneurons create a Schizophrenia-like Phenotype. . . . . . . . . . . . . . . . . . . . . . . . .
• Contribution of Genetic Epidemiology to Identifying Genetic and Environmental Risk Factors for Neurologic and Psychiatric Disorders . . . . . . . . . . . . . . . .
• Is Love Epigenetic? Transformative Effects of Social Experiences and of Oxytocin. . . . . . . . . . . . . . . . . . . . . . . . .
• Enhancing Cognitive Performance: Molecular, Pharmacological, and Experiential Strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
• Functional Connectivity in Neural Systems as a Developmental
Abnormality in Creating Risk for Bipolar Disorder. . . . . . . . . . . . . . .
• Optogenetic Dissection of Cortico-Limbic Circuit Function and Dysfunction. . . . . . . . . . . . . . . . . . . . . . . . . . . . .
• Translating Pharmacogenetics into Clinical Utility: Optimizing the Phenotype. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
x
205
206
207
208
209
210
211
212
213
214
215
216
217
218
ACNP 50th Annual Meeting • Final Program
Thursday, December 8th (continued)
Poster Sessions
Poster Session I. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
Poster Session II. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
Poster Session III . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301
Nonmember Participant List. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341
Disclosures. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
Author Index. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369
xi
ACNP 50th Annual Meeting • Final Program
Acknowledgments
The American College of Neuropsychopharmacology appreciates the support of
our supporting corporations:
Abbott Laboratories
Actelion Pharmaceuticals Ltd.
AstraZeneca Pharmaceuticals LP
Bristol-Myers Squibb Company
Eisai Medical Research, Inc.
Eli Lilly and Company
Forest Laboratories, Inc.
Genentech
H. Lundbeck A/S, Denmark
Hoffmann-LaRoche, Inc.
Janssen Pharmaceutica Products, L.P.
Merck
Novartis Pharmaceuticals Corporation
Otsuka Pharmaceutical Development, Inc.
Pfizer, Inc.
Shire
Sunovion Pharmaceuticals, Inc.
Takeda Pharmaceuticals
Targacept, Inc.
Vanderbilt University School of Medicine Department of Psychiatry and the
American College of Neuropsychopharmacology express appreciation to the
following companies for their support of this educational activity by providing an
unrestricted educational grant:
Eli Lilly & Company
Otsuka America Pharmaceuticals, Inc.
Pfizer
xii
ACNP 50th Annual Meeting • Final Program
Council
Officers and Council
President
President-Elect
Secretary
Treasurer
Eric J. Nestler
John H. Krystal
Alan Frazer
David J. Kupfer
Council
Karen F. Berman
David L. Braff
John G. Csernansky
Cindy Ehlers
Mark A. Geyer
Anthony A. Grace
Herbert D. Kleber
David R. Rubinow
Program Committee
2011 Program and Scientific Communications Committee
Chair
William Carlezon
Co-Chair
Anissa Abi-Dargham
Council Liaison
Eric J. Nestler
Members
Ted Abel
Elizabeth Abercrombie
Victoria Arango
David Baker
Aysenil Belger
Karen Berman
Randy Blakely
Joseph Buxbaum
Kristin Cadenhead
Marie Francoise Chesselet
Michael Davidson
Karl Deisseroth
Michael Egan
Igor Elman
Jay Gingrich
Stephan Heckers
Walter Kaye
Richard Keefe
Joel Kleinman
1
Thomas Lehner
Arnold Mandell
Lisa Monteggia
Maria Oquendo
Kerry Ressler
Peter Schmidt
David Schoepp
David Self
Pamela Sklar
Arielle Stanford
Trisha Suppes
Audrey Tyrka
Daniel Weinberger
Dean Wong
Rachel Yehuda
Carlos Zarate
Ad Hoc:
David Goldman
Paul Kenny
ACNP 50th Annual Meeting • Final Program
General Information
Dates and Location
Dates Location
Sunday, December 4, 2011 - Thursday, December 8, 2011
Hilton Waikoloa Village, Waikoloa, Hawaii
Program Book
All scientific registrants will receive a Program Book as part of their registration material. The Program Book will also be available on the ACNP
website, www.acnp.org.
Itinerary Planner
All scientific registrants will be able to access the itinerary planner for the 50th ACNP Annual Meeting at http://www.acnp-itinerary-planner.org
ACNP Executive Office
ACNP Executive Office
5034A Thoroughbred Lane
Brentwood, Tennessee 37027 USA
Phone: 615-324-2360
Fax:
615-523-1715
E-mail: [email protected]
2
ACNP 50th Annual Meeting • Final Program
Continuing Medical Education
The 2011 ACNP Annual Meeting is jointly sponsored by the Vanderbilt
School of Medicine and the ACNP. This activity has been planned and
implemented in accordance with the Essentials Areas and Policies of the
Accreditation Council for CME (ACCME) through the joint sponsorship
of Vanderbilt School of Medicine and the ACNP. Vanderbilt School of Medicine is accredited by the Accreditation Council for
Continuing Medical Education to provide continuing medical education for
physicians. Vanderbilt School of Medicine designates this live activity for a maximum of
37.5 AMA PRA Category 1 Credit(s)TM. Physicians should claim only the credit
commensurate with the extent of their participation in the activity. There will be a $40.00 charge for scientific registrants to obtain CME credits. CME instructions will be available at the meeting registration desk and on the
ACNP website (www.acnp.org).
It is the policy of Vanderbilt School of Medicine to require disclosure of financial
relationships from individuals in a position to control the content of a CME
activity; to identify and resolve conflicts of interest related to those relationships;
and to make disclosure information available to the audience prior to the CME
activity. Presenters are required to disclose discussions of unlabeled/unapproved
uses of drugs or devices during their presentations.
Program Overview/Statement of Need
The annual meeting of the American College of Neuropsychopharmacology
is designed to meet the educational needs of ACNP members and invited nonmember colleagues who are engaged at the leading edge of clinical and basic
science research on psychiatric disorders and their management. ACNP members
have been among the leaders in identifying underlying mechanisms for these
disorders and developing new treatment strategies. To continue and expand the
current level of research success, member researchers should develop research
projects that incorporate the latest developments that are appropriate in their areas
of research and of research in other associated areas into their research projects.
3
ACNP 50th Annual Meeting • Final Program
Continuing Medical Education (continued)
Target Audience
The target audience includes members of the American College of Neuropsychopharmacology and invited experts. The audience includes physicians,
psychologists, and basic neuroscientists from across the United States as well
as Europe and Asia. The physicians include a number of specialties, with
psychiatrists representing the majority of attendees, and neurologists next most
common. Psychologists include clinical psychologists and neuropsychologists. Learning Objectives:
After participating in this CME activity, participants should be able to describe
and discuss:
• How the results of recent or ongoing basic science and/or clinical studies
of psychiatric disorders in your area of interest or a related area impact
your current or potential future research projects.
• How you will change or modify a current approach or strategy in your
current or potential future research projects based on what you learned
from the results of recent or ongoing basic science and/or clinical studies
of psychiatric disorders in your area of interest or a related area. • How recent progress in identifying genetic variations that are risk factors
for the development of psychiatric disorders affect your current or
potential future research projects.
Information about CE Credit
Vanderbilt School of Medicine is approved by the American Psychological
Association to sponsor continuing education for psychologists. Vanderbilt School
of Medicine maintains responsibility for this program and its content.
Learning Objectives:
After participating in this CE activity, participants should be able to:
• Describe and discuss recent advances in treatment strategies for psychiatric
disorders, including psychoses and addictive disorders, and consider
application of these advances in their clinical practices and research
activities.
4
ACNP 50th Annual Meeting • Final Program
Continuing Medical Education (continued)
• Describe and discuss recent advances in basic and clinical neuroscience
that affect the development of new treatments and may modify current
treatment practices.
• Describe and discuss recent progress in identifying genetic variations that
are risk (or benefit) factors for the development of psychiatric disorders. Vanderbilt School of Medicine designates this educational activity for 37.5 CE
credits toward the continuing education of psychologists. No partial credit may
be awarded.
Americans with Disabilities Act
It is the policy of Vanderbilt School of Medicine not to discriminate against any
person on the basis of disabilities. If you feel you need services or auxiliary aids
mentioned in this act in order to fully participate in this continuing education
activity, please call the Executive Office at 615-324-2360 or send an email to
[email protected].
Meeting Evaluation
All meeting attendees are urged to complete an evaluation of the meeting. Attendees who are requesting CME or CE (APA) credit for the meeting are
required to complete the evaluation. This form is available online only. You
may complete the evaluation in the ACNP Computer Center located in meeting
room Waikoloa 2 or on-line at www.acnp.org (click the Annual Meeting tab). All
evaluations must be completed by January 16, 2012.
Scientific Program
VIDEOTAPING SESSIONS
Attendees may not videotape, audiotape, or photograph (camera or camera
phone) presentations at the Annual Meeting without prior permission from the
panel chair.
5
ACNP 50th Annual Meeting • Final Program
PLENARY SESSIONS
NEUROPSYCHOPHARMACOLOGY REVIEWS PLENARY. The year’s title
is “Neurotherapeutics Teaching Day” and will be chaired by Gwenn Smith,
Xiaohua Li and Jeffrey Conn. The speakers this year include Jeremy VeenstraVanderWeele, Bita Moghaddam, Yoland Smith, Sarah Lisanby and Barbara
Sahakian. The discussant will be William Potter. Sunday, December 4th
8:30 a.m. - 11:30 a.m.
This session is located in the Monarchy Ballroom.
NIH INSTITUTES UPDATE Join the Institute Directors from NIMH, NIAAA,
NIDA, NIA, and NINDS for an update on funding priorities in the coming year
and the strategic direction for the institutes. Sunday, December 4th
1:00 p.m. - 2:30 p.m.
This session is located in the Monarchy Ballroom.
HISTORY LECTURE. The lecture will be chaired by Joel Elkes. This year’s
lecture is presented in six parts by Joel Elkes, Don Klein, Judith Rapoport, Myrna
Weissman, Alan Schatzberg and William Bunney. The title of this presentation is
“Neuropsychopharmacology, the past 50 years.”
Sunday, December 4th
2:30 p.m. - 4:00 p.m.
This session is located in the Monarchy Ballroom.
HOT TOPICS. Each presentation should last about ten minutes with a fiveminute period for questions and open discussion. There are two categories: Basic/
Translational topics & Clinical topics. The two sessions will be concurrent.
Sunday, December 4th
Basic Clinical Kona 5
Kona 4
6
4:00 p.m. - 6:30 p.m.
ACNP 50th Annual Meeting • Final Program
PRESIDENT’S PLENARY. This year’s President’s Plenary, chaired by ACNP
President, Eric Nestler, is titled “Brave New World for Brain Therapeutics.”
Monday, December 5th
8:00 a.m. - 11:30 a.m.
This session is located in the Monarchy Ballroom.
DISTINGUISHED LECTURE. This plenary is chaired by Eric Nestler. The
lecture will be presented by Louis Ptáček. The title of his talk is, “Insights into
Circadian Clock and Sleep from Human Genetics.”
Monday, December 5th
1:30 p.m. - 3:00 p.m.
This session is located in the Monarchy Ballroom.
DATA BLITZ SESSION. This new session is comprised of rigorously timed
5-min presentations by 12 young investigators that are linked to posters scheduled
for that same evening.
Tuesday, December 6th 11:30 a.m. - 1:30 p.m.
The session is located in Kona 4.
TRAVEL AWARDEE RESEARCH PRESENTATIONS. Our current travel
awardees had the option of submitting oral presentations for review and possible
selection by the Education and Training Committee for this session. There will
be three concurrent sessions containing three presentations each.
Tuesday, December 6th
11:30 a.m. - 1:30 p.m.
The sessions are located in Kona 1-3, Kohala 4, Queen’s 5-6.
FACULTY RESEARCH FELLOWSHIP PRESENTATIONS. These
awardees are engaged in a three-year research project with an ACNP member
mentor. The Fellows must present their plan for their fellowship and research
project for their three-year study.
Tuesday, December 6th
This session is located in Kohala 3.
7
11:30 a.m. - 1:30 p.m.
ACNP 50th Annual Meeting • Final Program
ISSUES IN ETHICS. This year’s topic is “The Perils and Pitfalls of
Biomedical Research: Historical and Contemporary Perspectives on the
Ethics of Research” and is co-chaired by Ellen Frank and Jeffrey Lieberman. The speakers this year include Arthur Caplan with discussion from David Jentsch
and Nina Schooler.
Tuesday, December 6th
1:30 p.m. - 3:00 p.m.
This session is located in Kona 5.
CONCURRENT SESSIONS
PANELS. The title and location of each Panel is indicated in the Program. The
presentations in each session are scheduled at approximately 30-minute intervals,
allowing for 20-minute presentations and 10-minute discussion periods. A thirtyminute general discussion period is scheduled after the last presenter in each
session. Please note that the panels on Thursday are scheduled at a different
time. Panels are scheduled:
Monday, December 5th
3:00 p.m. - 5:30 p.m.
Tuesday, December 6th
8:30 a.m. - 11:00 a.m.
3:00 p.m. - 5:30 p.m.
Wednesday, December 7th
8:30 a.m. - 11:00 a.m.
3:00 p.m. - 5:30 p.m.
Thursday, December 8th
8:00 a.m. - 10:30 a.m.
12:00 p.m. - 2:30 p.m.
MINI PANELS. The title and location of each Panel is indicated in the Program. A mini-panel is a moderately formal 75-minute session that includes 1 chair and
3 presenters. Each of the 3 presentations lasts 25 minutes which allows for a
20-minute presentation and 5-minute discussion period. Tuesday, December 6th
8:30 a.m. - 9:45 a.m.
9:45 a.m. - 11:00 a.m.
Wednesday, December 7th
3:00 p.m. - 4:15 p.m.
4:15 p.m. - 5:30 p.m.
Thursday, December 8th
8:00 a.m. - 9:15 a.m.
9:15 a.m. - 10:30 a.m.
8
ACNP 50th Annual Meeting • Final Program
STUDY GROUPS. Study groups will be issue oriented again this year. They
are scheduled for Monday evening. The title and location of each Study Group is
indicated in the Program. They are:
Monday, December 5th
7:30 p.m. - 9:00 p.m.
POSTER SESSIONS
POSTERS. Poster presentations are grouped by general topic area. Topic areas
are often repeated. Monday, December 5th
5:30 p.m. - 7:30 p.m.
Poster Session I
This session is located in Kohala 1-2, King’s & Grand Promenade.
Tuesday, December 6th
5:30 p.m. - 7:30 p.m.
Poster Session II
This session is located in Kohala 1-2, King’s & Grand Promenade.
Wednesday, December 7th
5:30 p.m. - 7:30 p.m.
Poster Session III
This session is located in Kohala 1-2, King’s & Grand Promenade.
9
ACNP 50th Annual Meeting • Final Program
Registration
ON-SITE REGISTRATION PERIODS
Date Saturday, December 3th
Sunday, December 4th Monday, December 5th
Tuesday, December 6th
Wednesday, December 7th
Thursday, December 8th
TimeLocation
9:00 a.m. - 7:00 p.m. Grand Promenade
7:00 a.m. - 7:00 p.m. Grand Promenade
7:00 a.m. - 6:00 p.m. Grand Promenade
7:00 a.m. - 6:00 p.m. Grand Promenade
7:30 a.m. - 5:30 p.m. Grand Promenade
8:00 a.m. - 3:00 p.m. Grand Promenade
ON-SITE REGISTRATION FEES
Categories ACNP Associate Members ACNP Members & Fellows ACNP Emeritus
Members of AsCNP, CCNP, CINP, ECNP, JSNP (on list)
Corporate Representatives Non-Member Program Participant 2010 Travel Awardees
Trainee*
Invited Guest Past Travel Awardee (2007-2010) Optional Journal Subscription for non-ACNP members**
Accompanying Person*** On-Site Fee
$250
$600
$250
$600
$850
Waived
Waived
$250
$850
$250
$150
$200
*A Trainee is a person in a training position and not a full-time, permanent position. A Trainee can be a M.D., Ph.D., Postdoctoral Fellow, Resident, or Research Fellow. Trainees have all the rights and privileges of Invited Guests.
**Non-ACNP members may pay $150 extra and receive a 1-year subscription (calendar
year 2012) to Neuropsychopharmacology. This is a substantial discount from the normal
non-member rate of $413.
***An accompanying person is a spouse, relative, or significant other who accompanies
an attendee. An accompanying person may only attend the social functions of the meeting,
which includes the morning breaks, lunches, and the ACNP Anniversary Celebration. Accompanying persons are also allowed to attend the poster sessions.
10
ACNP 50th Annual Meeting • Final Program
Hotel Facilities
The Hilton Waikoloa Village guest rooms, common areas, and transportation
services are in compliance with the public accommodation requirements of the
Americans with Disabilities Act. These facilities will be accessible to and usable
by individuals with disabilities who attend and participate in the Annual Meeting.
Meeting and activity rooms are smoke-free.
Services
ACNP Computer Center
The ACNP Computer Center is provided this year for the convenience of meeting
attendees. The computers will be available in meeting room Waikoloa 2 for the
following: e-mail, internet access, and meeting evaluation.
Sunday through Wednesday Thursday
7:30 a.m. - 9:00 p.m.
7:30 a.m. - 2:00 p.m.
Speaker Ready Room
A speaker ready room will be available in Waikoloa 1. The speaker ready room
will be open Sunday through Wednesday, 7:30 a.m. - 6:30 p.m.; Thursday, 7:00
a.m. - 2:30 p.m.
Travel Information Desk
The Travel Information Desk is located next to the ACNP registration desk and
is staffed by ACNP staff. Participants can obtain lodging, travel, or car rental
information at this desk
Refreshment Breaks
All registered individuals are invited.
Coffee and light pastries for registered individuals will be available Sunday at
7:30 a.m. and Monday through Thursday at 7:00 a.m. All breaks are in the Grand
Promenade.
Afternoon coffee breaks will be available Sunday through Wednesday from 2:30
p.m. to 4:30 p.m. in the Grand Promenade.
11
ACNP 50th Annual Meeting • Final Program
Luncheons
All registered individuals are invited.
Date: Monday, December 5th
Time: 11:30 a.m. - 1:30 p.m.
Place: Grand Promenade/Lagoon Lanai
Date: Tuesday, December 6th
Time: 11:00 a.m. - 12:30 p.m.
Place: Grand Promenade/Lagoon Lanai
Date: Wednesday, December 7th
Time: 12:30 p.m. - 2:00 p.m.
Place: Grand Promenade/Lagoon Lanai
Date: Thursday, December 8th
Time: 10:30 a.m. - 12:00 p.m.
Place: Grand Promenade/Lagoon Lanai
ACNP Anniversary Celebration
All registered individuals are invited.
Date: Sunday, December 4th
Time: 7:00 p.m. - 9:00 p.m.
Place: Monarchy and Queen’s Ballroom, Grand Promenade, Lagoon Lanai and Grand Staircase
Special Events
Sunday, December 4, 2011
Travel Award Breakfast
(by invitation only)
Time: 7:00 a.m. - 8:30 a.m.
Place: Water’s Edge Ballroom
12
ACNP 50th Annual Meeting • Final Program
Sunday, December 4, 2011
Past Presidents’ Luncheon
(by invitation only)
Time: 11:30 a.m. - 1:00 p.m.
Place: Queen’s 4
Monday, December 5, 2011
Women’s Luncheon
Time: 12:00 p.m. - 1:30 p.m.
Place: Water’s Edge Ballroom
Teaching Neuropsychopharmacology
Time: 12:00 p.m. - 1:30 p.m.
Place: Kohala 4
Wednesday, December 7, 2011
Travel Award Luncheon
(by invitation only)
Time: 12:30 p.m. - 2:00 p.m.
Place: Water’s Edge Ballroom
Career Development Session, “Ask the Expert”
Time: 12:30 p.m. - 2:00 p.m. Place: Queen’s 5-6
An Oral History of Neuropsychopharmacology
Time: 7:30 p.m. - 9:30 p.m. Place: Monarchy
13
ACNP 50th Annual Meeting • Final Program
College Meetings
Saturday, December 3, 2011
Neuropsychopharmacology Editor-in-Chief & Deputy Editors
Time: 2:00 p.m. - 3:30 p.m.
Place: King’s 2
Tuesday, December 6, 2011
Neuropsychopharmacology Reviews Editors
Time: 7:00 a.m. - 8:30 a.m.
Place: Water’s Edge Boardroom
ACNP Leadership & NIH Directors
Time: 7:30 a.m. - 8:30 a.m.
Place: Donatonio’s
Corporate Liaison Luncheon
(by invitation only)
Time: 11:00 a.m. - 12:30 p.m.
Place: Donatonio’s
Neuropsychopharmacology Editorial Board
Time: 7:30 p.m. - 9:00 p.m.
Place: Water’s Edge Ballroom
Wednesday, December 7, 2011
Annual Business Meeting
(ACNP Fellows, Members and Associate Members only)
Chair: Eric Nestler
Time: 11:15 a.m. - 12:30 p.m.
Place: Monarchy
Thursday, December 8, 2011
ACNP/AsCNP/CINP/ECNP/ JSNP Officers
Time: 7:00 a.m. - 8:00 a.m.
Place: Queen’s 4
14
ACNP 50th Annual Meeting • Final Program
Council Meetings
Council is involved in many activities during the Annual Meeting. The official
Council meetings are listed below:
Saturday, December 3, 2011
ACNP Council
Time: 8:00 a.m. - 3:00 p.m.
Place: Queen’s 4
Tuesday, December 6, 2011
ACNP Council
Time: 6:00 p.m. - 11:00 p.m.
Place: Queen’s 4
Thursday, December 8, 2011
ACNP Council
Time: 9:00 a.m. - 12:00 p.m.
Place: Queen’s 4
Committee/Task Force Information
Meetings listed below are as scheduled by the Committee Chairs. Saturday, December 3, 2011
Membership Committee
Time: 8:00 a.m. - 5:00 p.m.
Place: King’s 1
Publications Committee
Time: 3:30 p.m. - 5:00 p.m.
Place: King’s 2
Ethics Committee
Time: 4:00 p.m. - 5:30 p.m.
Place: Water’s Edge Boardroom
15
ACNP 50th Annual Meeting • Final Program
Saturday, December 3, 2011 (continued)
Public Information Committee
Time: 5:00 p.m. - 7:00 p.m.
Place: Kona 2
Sunday, December 4, 2011
Education & Training Committee
Time: 11:30 a.m. - 1:00 p.m.
Place: Kona 3
Program Committee
Time: 11:30 a.m. - 1:00 p.m.
Place: Kona 1
Tuesday, December 6, 2011
Liaison Committee
Time: 7:00 a.m. - 8:30 a.m.
Place: Water’s Edge Ballroom
Membership Advisory Task Force
Time: 7:00 a.m. - 8:30 a.m.
Place: Queen’s 4
Wednesday, December 7, 2011
Women’s Task Force
Time: 7:00 a.m. - 8:30 a.m.
Place: Water’s Edge Boardroom
16
ACNP 50th Annual Meeting • Final Program
ACNP Sanctioned Meetings
CINP Executive Committee
Date: Saturday, December 3, 2011
Time: 9:00 a.m. - 5:00 p.m.
Place: Kona 3
ASCP Board of Directors Meeting
Date: Monday, December 5, 2011
Time: 12:00 p.m. - 1:30 p.m.
Place: Queen’s 4
American Journal of Psychiatry
Date: Tuesday, December 6, 2011
Time: 7:00 a.m. - 8:30 a.m.
Place: Imari
Schizophrenia International Research Society
Industry Task Force Meeting
Date: Wednesday, December 7, 2011
Time: 12:30 p.m. - 2:00 p.m.
Place: Donatonio’s
SOBP (Society of Biological Psychiatry)
Council
Date: Friday, December 2, 2011
Time: 11:00 a.m. - 6:00 p.m. Place: Kona 3
Program Committee
Date: Wednesday, December 7, 2011
Time: 12:30 p.m. - 2:00 p.m.
Place: Queen’s 4
17
Editorial Board Meeting
Date: Monday, December 5, 2011
Time: 6:30 a.m. - 8:00 a.m.
Place: Queen’s 4
ACNP 50th Annual Meeting • Final Program
Future ACNP Annual Meetings
DatesHotel
Location
December 2 - 6, 2012
The Westin Diplomat
Hollywood, Florida
December 8 - 12, 2013
The Westin Diplomat
Hollywood, Florida
December 7 - 11, 2014
Marriott Desert Ridge Resort
Phoenix, Arizona
December 6 - 10, 2015
The Westin Diplomat
Hollywood, Florida
December 4 - 8, 2016
The Westin Diplomat
Hollywood, Florida
In Memoriam
Maressa Hecht Orzack
November 11, 2010
Alan A. Boulton
November 13, 2010
Charles Roberts Schuster
February 21, 2011
Alfred M. Freedman
April 17, 2011
Seymour M. Antelman
June 10, 2011
John A. Harvey
June 25, 2011
Alexander H. Glassman
July 19, 2011
Joseph V. Brady
July 29, 2011
Jose M. Delgado
September 5, 2011
18
Sunday At A Glance
7:00 am – 8:30 am
Travel Award Breakfast
(by invitation only)
8:30 am – 11:30 am Neuropsychopharmacology Reviews Plenary
11:30 am – 1:00 pm
Education and Training Committee Meeting
11:30 am – 1:00 pm
Past President’s Luncheon
(by invitation only)
11:30 am – 1:00 pm
Program Committee Meeting
1:00 pm – 2:30 pm
NIH Institutes Update
Monarchy
2:30 pm – 4:00 pm
History Lecture
Monarchy
4:00 pm – 6:30 pm
Hot Topics – Basic
Kona 5
4:00 pm – 6:30 pm Hot Topics - Clinical
Kona 4
Monarchy Ballroom
Kona 3
Queen’s 4
Kona 1
Monarchy, Queen’s &
Grand Promenade,
Grand Staircase &
Lagoon Lanai
PA-Panel MP-Mini Panel PL-Plenary SG-Study Group
Sunday
7:00 pm – 9:00 pm 50th Anniversary Celebration Gala
Water’s Edge Ballroom
Sunday
Notes
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
Neuropsychopharmacology Reviews Plenary
Neurotherapeutics Teaching Day
Chair: Gwenn Smith, Xiaohua Li and Jeffrey Conn
8:30 a.m.
Autism Therapeutics
Jeremy Veenstra-VanderWeele
8:55 a.m.
From Revolution to Evolution: The Glutamate Hypothesis of
Schizophrenia and its Implication for Treatment
Bita Moghaddam
9:20 a.m.
Parkinson’s Disease Therapeutics
Yoland Smith
9:45 a.m.
Translational Development of Transcranial Approaches to
Depression Treatment
Sarah Lisanby
10:10 a.m.
Lifespan Studies of Depression: Implications for Treatment
Barbara Sahakian
10:35 a.m.
Discussant: William Potter
19
PL
ACNP 50th Annual Meeting • Final Program
PL
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
Autism Therapeutics
Jeremy Veenstra-VanderWeele
Vanderbilt University
Autism Spectrum Disorder (ASD) is a common neurodevelopmental disorder
affecting approximately 1% of children. ASD is defined by core symptoms in two
domains: negative symptoms of impairment in social and communication function,
and positive symptoms of restricted and repetitive behaviors. Available treatments
are inadequate for treating both core symptoms and associated conditions. Twin
studies indicate that ASD susceptibility has a large heritable component. Genetic
studies have identified promising leads, with converging insights emerging from
single gene disorders that bear ASD features, with particular interest in mTORlinked synaptic plasticity mechanisms. Mouse models of these disorders are
revealing not only opportunities to model behavioral perturbations across species
but also evidence of postnatal rescue of brain and behavioral phenotypes. An
intense search for ASD biomarkers has consistently pointed to elevated platelet
serotonin (5-HT) levels and a surge in brain growth in the first two years of life. Following a review of the diversity of ASD phenotypes and its genetic origins
and biomarkers, we will discuss opportunities for translation of these findings
into novel ASD treatments, focusing on mTor and 5-HT signaling pathways and
their possible intersection. Paralleling the progress made in understanding the
root causes of rare genetic syndromes that affect cognitive development, we
anticipate progress in models systems using bona fide ASD-associated molecular
changes that have the potential to accelerate development of ASD diagnostics and
therapeutics.
Jeremy Veenstra-VanderWeele completed his undergraduate degree in Psychology
at Harvard University. He then attended medical school at the University of
Chicago, where he completed a pre-doctoral year and molecular genetic research
training in the lab of Dr. Edwin H. Cook, Jr., M.D. After completing general and
child psychiatry residency, he moved to Vanderbilt University for a postdoctoral
fellowship with Randy Blakely, Ph.D., to learn how to study mouse models of
autism and OCD-related susceptibility genes. Jeremy has now established his own
lab as an Assistant Professor at Vanderbilt, where his group works to understand
the contributions of genes in the serotonin and glutamate systems to social and
20
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
Autism Therapeutics (continued)
Jeremy Veenstra-VanderWeele
repetitive behavior in mice. This work has been funded by the National Institute
of Mental Health, Autism Speaks, NARSAD, and the American Academy of Child
and Adolescent Psychiatry. In parallel, he has developed a translational clinical
trials program in Fragile X Syndrome and autism spectrum disorder. Ultimately,
he hopes to contribute to new treatments that will relieve symptoms in children
with autism spectrum disorders.
21
PL
ACNP 50th Annual Meeting • Final Program
PL
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
From Revolution to Evolution:
The Glutamate Hypothesis of Schizophrenia
and its Implication for Treatment
Bita Moghaddam
University of Pittsburgh
Glutamate is the primary excitatory neurotransmitter in mammalian brain. Disturbances in glutamate-mediated neurotransmission have been increasingly
documented in a range of neuropsychiatric disorders including schizophrenia,
substance abuse, mood disorders, Alzheimer’s disease and autism-spectrum
disorders. Glutamatergic theories of schizophrenia are based on the ability
N-methyl-D-aspartate receptor (NMDAR) antagonists to induce schizophrenialike symptoms, as well as emergent literature documenting disturbances of
NMDAR-related gene expression and metabolic pathways in schizophrenia. Research over the past two decades has highlighted promising new targets for
drug development based on potential pre- and postsynaptic, and glial mechanisms
leading to NMDAR dysfunction. Reduced NMDAR activity on inhibitory
neurons leads to disinhibition of glutamate neurons increasing synaptic activity
of glutamate, especially in the prefrontal cortex. Based on this mechanism,
normalizing excess glutamate levels by metabotropic glutamate group 2/3
receptor agonists has led to the identification of the first non-monoaminergic
target with comparable efficacy as conventional antipsychotic drugs for treating
positive and negative symptoms of schizophrenia. In addition, NMDAR has
intrinsic modulatory sites that are active targets for drug development, several of
which show promise in preclinical/early clinical trials targeting both symptoms
and cognition. To date, most studies have been done with orthosteric agonists
and/or antagonists at specific sites. However, allosteric modulators, both positive
and negative, may offer superior efficacy with less danger of downregulation. Bita Moghaddam, PhD is Professor of Neuroscience, Psychiatry, and
Pharmaceutical Sciences at the University of Pittsburgh. She received a PhD in
Biochemistry from the University of Kansas in the laboratory of Ralph N. Adams
and postdoctoral training in pharmacology at Yale University in the laboratory
of Steve Bunney. Her research focuses on the use of animal models to study the
22
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
From Revolution to Evolution:
The Glutamate Hypothesis of Schizophrenia
and its Implication for Treatment (continued)
Bita Moghaddam
cellular basis of cognitive constructs that are critical to psychiatric disorders,
including schizophrenia. Dr. Moghaddam is the author of over 100 scientific
papers in leading journals, including Science and Proceedings of the National
Academy of Sciences. Her work has led to the discovery of the first non-monoamine
targeting compound (targeting metabotropic glutamate receptors) for potential
treatment of schizophrenia. Her research has been funded continuously since
1991, including a MERIT award from the National Institute of Mental Health. Her education experience involves extensive didactic teaching of neuroscience
to undergraduates, graduate students, medical students and residents. She is the
recipient of many research awards, including ACNP’s Efron award for excellence
in research related to neuropsychopharmacology and CINP’s Paul Jansen Award
for excellence in schizophrenia research. 23
PL
ACNP 50th Annual Meeting • Final Program
PL
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
Parkinson’s Disease Therapeutics
Yoland Smith
Emory University
The demonstrations that dopamine loss is the key pathological feature of Parkinson’s
disease, and the subsequent introduction of Levodopa, have revolutionalized
the field of Parkinson’s disease (PD) therapeutics. In this presentation, I will
discuss the significant progress that has been made in the development of new
pharmacological and surgical tools to treat PD motor symptoms since these
major breakthroughs in the 1960’s. However, I will also highlight some of
the challenges the field of PD therapeutics has been struggling with during the
past decades. The lack of neuroprotective therapies and the limited treatment
strategies for the nonmotor symptoms of the disease (i.e. cognitive impairments,
autonomic dysfunctions, psychiatric disorders, etc.) are among the most pressing
issues to be addressed in years to come. It appears that the combination of early
PD nonmotor symptoms with imaging of the nigrostriatal dopaminergic system
offers a promising path towards the identification of PD biomarkers which, once
characterized, will set the stage for efficient use of neuroprotective agents that
could slow down and alter the course of the disease. Dr. Smith got his PhD degree in Neurobiology from Laval University, Quebec,
Canada in 1988, under the supervision of Professor Andre Parent. He then spent
two years of postdoctoral training in the laboratory of Professor Paul Bolam in
the Medical Research Council Unit in Oxford UK. After his postdoc training
in Oxford, he spent a year in the laboratory of Professor Mahlon DeLong in
Johns Hopkins University. After this stay in USA, he got a faculty position in the
Department of Anatomy of Laval University in Quebec where he spent five years
developing a research program that focused on the synaptic organization of the
primate basal ganglia. In 1996, he moved to the Yerkes National Primate Research
Center of Emory University in Atlanta where he is now a full Professor in the
Department of Neurology and the Yerkes Center. His research is supported by
grants from the National Institute of Health, the National Parkinson Foundation,
the Tourette Syndrome Association, the Michael J Fox Foundation and the RJG
Foundation. He is a member of the Emory University`s UDALL Center for
Parkinson`s disease. He has published over 200 peer-reviewed manuscripts and
24
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
Parkinson’s Disease Therapeutics (continued)
Yoland Smith
book chapters. He has been section editor for the International IBRO journal
“Neuroscience” and he sits on many reviewing committees and editorial boards
at national and international levels. He has received various awards and he is
currently the Director of the PhD Graduate Neuroscience Program of Emory
University.
25
PL
ACNP 50th Annual Meeting • Final Program
PL
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
Translational Development of Transcranial
Approaches to Depression Treatment
Sarah Lisanby
Duke University School of Medicine
Targeted modulation of brain function transcranially, without the need for surgery,
offers great potential in the treatment of depression. From the powerful efficacy
of electroconvulsive therapy (ECT), to the relative safety of transcranial magnetic
stimulation (TMS), transcranial approaches have broadened the therapeutic
spectrum for depression, and new developments in the field promise a growing
array of safe and effective alternatives to come. Transcranial approaches to the
treatment of depression include a range of magnetic and electrical interventions
including the seizure therapies {ECT, magnetic seizure therapy (MST), focal
electrically administered seizure therapy (FEAST)} and subconvulsive therapies
{TMS, transcranial direct current stimulation (tDCS), cranial electrical stimulation
(CES)}. These interventions are similar with respect to their lack of invasiveness
and use of electrical currents to stimulate the brain, either directly or indirectly
via electromagnetic induction. Key to the successful clinical implementation
of these interventions is the determination of the optimal dosage to induce safe
and effective antidepressant response. Indeed, dosage has long been known to
determine the safety and efficacy of the gold standard transcranial approach to
depression – ECT. However, even in the case of ECT, understanding of how
to best quantify and control the dose of transcranial treatments is evolving. Recent research has questioned the wisdom of summary metrics that collapse
information across stimulus parameters, and highlights the critical importance of
individual parameters in determining impact on brain function. Preclinical studies
offer the opportunity to systematically evaluate the individual contributions of
parameters to determining specific physiological outcomes. However, there
are barriers to translating findings from the preclinical to the clinical context. These barriers include limits to homology across species, inappropriate scaling
of stimulation paradigms across head sizes and shapes, and limits to the validity
of animal models of clinical syndromes. These barriers can be addressed through
realistic head modeling to scale fields induced in preclinical models to match the
clinical context and the selection of appropriate intermediate outcome measures
26
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
Translational Development of Transcranial
Approaches to Depression Treatment (continued)
Sarah Lisanby
associated with efficacy. These concepts will be illustrated in the translational
development of magnetic seizure therapy, and results having implications for the
dosing of novel as well as traditional seizure therapies will be highlighted. The
potential of translation to improve the efficacy of TMS will also be discussed,
with implications for coil design and parameter selection.
Sarah Hollingsworth Lisanby, MD, an internationally recognized leader in the
field of brain stimulation, is Professor and Chair of the Department of Psychiatry
and Behavioral Sciences at Duke University School of Medicine. She is also
Professor of Psychology and Neuroscience at Duke University School of Arts
and Sciences. She is the co-author on more than 150 publications in prestigious
scientific journals, including The New England Journal of Medicine. Dr. Lisanby
has a distinguished academic record as a member of the FDA Neurological
Devices Advisory Panel, NIH Study Sections, co-Principal Investigator of
an 8-center NIH funded U01 on Prolonging Remission in Depressed Elders
(RPIDE), former President of the leading international professional organizations
on brain stimulation (Association for Convulsive Therapy / International Society
of Neurostimulation, and the International Society for Transcranial Stimulation),
and as Chair of both American Psychiatric Association committees related to
brain stimulation (APA committee on ECT and the APA Task Force to Revise the
Guidelines on ECT). She also co-Chairs the National Network of Depression
Centers (NNDC) Task Group on transcranial magnetic stimulation (TMS). She has
received many prestigious awards. Dr. Lisanby earned her BS in mathematics and
psychology magna cum laude at Duke University and her MD at Duke University
School of Medicine in Durham, North Carolina. She completed her residency in
psychiatry at Duke University Medical Center, where she served as Executive
Chief Resident. Dr. Lisanby joined Columbia in 1995 to pursue a postdoctoral
research fellowship in Affective Disorders and Geriatric Psychiatry. She joined
Columbia’s psychiatry faculty in 1998, and was the founding director of the
Brain Stimulation and Therapeutic Modulation Division there from 2005-2010.
27
PL
ACNP 50th Annual Meeting • Final Program
PL
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
Lifespan Studies of Depression: Implications for Treatment
Barbara Sahakian
University of Cambridge Department of Psychiatry
Many neuropsychiatric disorders are of neurodevelopmental origin, with an onset
or prodromal stage in childhood or adolescence. The identification of cognitive
and other biomarkers, including from neuroimaging and genetics, for detecting
vulnerability and onset is important for the prevention of neuropsychiatric
disorders, such as depression, becoming chronic and relapsing. Neuropsychiatric
disorders typically manifest as problems with attentional biases, aberrant learning,
dysfunctional reward systems and lack of top down cognitive control by prefrontal
cortex. The key to prevention, early detection and early effective treatment is the
use of cognitive biomarkers such as negative attentional biases (Roiser, Elliott &
Sahakian, in press). Otherwise, disorders may lead to changed states including
entrenched habits which are difficult, if not impossible, to treat such as is the
case of drug addiction. Similarly, in depression when the disorder has become
chronic and relapsing, it is often resistant to treatment, although fortunately, new
approaches, such as fast acting ketamine or deep brain stimulation are likely to
become more widely used (Holtzheimer & Mayberg, 2010; Kennedy et al, 2011;
Zarate et al, 2006; See also Drevets & Furey, 2010). This lifespan approach may
prevent some individuals from developing depression and lead to early effective
treatment in others. Given the cost of common mental health disorders in terms of
distress to the individual and their family as well as the cost financially to society
and governments, new developments for treatments across the lifespan should
be a priority so that all members of society can flourish (Beddington et al, 2008;
Collins et al, 2011; Insel, 2009; Sahakian et al, 2010).
Dr. Sahakian has an international reputation in the fields of cognitive psychopharmacology, neuroethics, neuropsychology, neuropsychiatry and neuroimaging. She is co-inventor of the CANTAB computerised neuropsychological tests, which
are in use world-wide. Indeed, she has over 300 publications covering these
topics in scientific journals, including Science, Nature, Nature Neuroscience,
The Lancet, British Medical Journal, Archives of General Psychiatry, American
Journal of Psychiatry, Biological Psychiatry, the Journal of Neuroscience, Brain,
Psychopharmacology and Psychological Medicine. From 2006 to 2010 she was
28
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Neuropsychopharmacology Reviews Plenary
Monarchy Ballroom
Lifespan Studies of Depression: Implications for Treatment
(continued)
Barbara Sahakian
on the Medical Research Council Neurosciences and Mental Health Board, and
in 2008 she was appointed to the MRC Expert Group on Strategy on Mental
Health. In 2009 to 2010 she was awarded the Distinguished International Scholars Award from the University of Pennsylvania. In 2010 she was appointed to the
Scientific Advisory Board of the Grand Challenges in Global Mental Health. She
is a founder member and on the executive board of the Neuroethics Society and
co-editor of The Oxford Handbook of Neuroethics (2011). 29
PL
ACNP 50th Annual Meeting • Final Program
PL
1:00 p.m. – 2:30 p.m.
NIH Institute Directors Briefing
Monarchy
NIH Institutes Update
Chair: Eric Nestler
1:00 p.m.
Neil Buckholtz
NIA
1:15 p.m.
Thomas Insel
NIMH
1:30 p.m.
Story Landis
NINDS
1:45 p.m.
Kenneth Warren
NIAAA
2:00 p.m.
Nora Volkow
NIDA
2:15 p.m. Open Discussion
30
ACNP 50th Annual Meeting • Final Program
2:30 p.m. – 4:00 p.m.
History Lecture
Monarchy
History Lecture
Neuropsychopharmacology, The Past 50 Years
History Committee Chair: James Anthony
Honorary Chair: Joel Elkes
2:30 p.m. Introductions
Alan Frazer
2:40 p.m. Reflections on ACNP’s Pre-History and the Developments
Leading up to ACNP’s Emergence in the Early 1960s
Joel Elkes
3:00 p.m. Highlights, Events, Discoveries, and Progress made during the
Early 1960s through 1970
Donald Klein
3:10 p.m. 3:20 p.m. Highlights, Events, Discoveries, and Progress made during the
Early 1970s through 1980
Judith Rapoport
Highlights, Events, Discoveries, and Progress made during the
Early 1980s through 1990
Myrna Weissman
3:30 p.m. What can we learn from the Early 21st Century?
Alan Schatzberg
3:40 p.m. Beyond 2011
William Bunney
3:50 p.m. Closing Remarks
Joel Elkes
31
PL
ACNP 50th Annual Meeting • Final Program
PL
2:30 p.m. – 4:00 p.m.
History Lecture
Monarchy
History Lecture
Neuropsychopharmacology, The Past 50 Years
History Committee Chair: James Anthony
Honorary Chair: Joel Elkes
The panel’s Honorary Chairman, Professor Joel Elkes, ACNP’s first president,
will reflect on what he calls ACNP’s ‘pre-history’ and the developments leading
up to ACNP’s emergence in the early 1960s. Donald Klein will draw attention
to highlights, events, discoveries, and progress made during the years from the
early 1960s through 1970, based upon his own selection of these highlights. Judy
Rapoport, Myrna Weissman, and Alan Schatzberg will do the same for subsequent
decades. William Bunney will offer a forecast of what may be important in our
future work. Professor Elkes will offer a synthesis and closing remarks, after
which Alan Frazer, panel moderator, will invite discussion and questions from
the floor.
32
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Hot Topics
Basic
Chair: William Carlezon
4:00 p.m.
4:15 p.m.
Systems Biological Analyses implicate Glutamate Signaling
Abnormalities in Autism and in Intellectual Disability:
Implications for Psychopharmacology
Joseph Buxbaum
Small and Whole Transcriptome RNA Sequencing identifies
Key Regulation Patterns in the Medial Prefrontal Cortex of the
Alcohol Dependent Rat
Jenica Tapocik
4:30 p.m.
Selective Optogenetic Stimulation of Parvalbumin-positive
Basal Forebrain Neurons Reliably entrains Cortical Gamma
Oscillations and Promotes Wakefulness
Robert W. McCarley
4:45 p.m.
Epigenetic Differences in the Developing Hippocampus and
Amygdala in a Novel Rat Model of Anxiety and Depression
Sarah M. Clinton
5:00 p.m.
Essential Role of Ventral Tegmental Area Dopamine Neurons in
Mediating the Induction and Rapid Reversal of Depression-Like
Behaviors
Ming-Hu Han
5:15 p.m.
Stress Exposure produces a Switch from Appetitive to Aversive
Signaling by Corticotropin-releasing Factor in the Nucleus
Accumbens
Paul Phillips
33
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Hot Topics
Basic (continued)
Chair: William Carlezon
5:30 p.m.
Altered Anxiety-Like Behavior in BDNF Val66Met Mice is
rescued with Early-Life Fluoxetine
Iva Dincheva
5:45 p.m.
The Involvement of Alpha3-Containing Nicotinic Acetylcholine
Receptors in the Habenulo-Interpeduncular Pathway in Nicotine
Self-Administration
Christie Fowler
6:00 p.m.
The Novel Opioid Receptor Modulator RDC-0313 (ALKS 33)
reduces Olanzapine-induced Weight Gain and Adipose Accretion
in a Novel Nonhuman Primate Model of Antipsychotic-related
Weight Changes
Mark Todtenkopf
6:15 p.m.
Discovery of the First & β-Arrestin-Biased Dopamine
D2 Ligands for Probing Signaling Pathways Essential for
Antipsychotic Efficacy
Jian Jin
34
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Systems Biological Analyses implicate Glutamate Signaling
Abnormalities in Autism and in Intellectual Disability:
Implications for Psychopharmacology
Tuesday, Poster #111
Joseph Buxbaum, Avi Ma’ayan, Yan Kuo, Ruth Dannenfelser, Catalina Betancu
Mount Sinai School of Medicine
BACKGROUND: Autism spectrum disorders (ASDs) are characterized by
deficits in social communication and by repetitive behaviors and/or restricted
interests. Numerous rare genetic variants of major effect have been identified in
ASDs. To understand how such etiological heterogeneity translates into common
neurobiological pathways, we used gene enrichment and pathway analyses. METHODS: We made use of a manually collated list of genes where mutations
have been shown to be associated with high risk for ASD. These genes include
those that are well known (e.g., NRXN3, NRXN4, SHANK2, SHANK3, FMR1,
UBE3A, MECP2) and many other less common genes. To extend the findings to
an additional neurodevelopmental disorder related genetically to ASD, we also
curated a list of almost 200 genes where mutations lead to intellectual disability
(ID). We then used unbiased enrichment analyses, making use of data from largescale proteomic studies, to determine whether there was evidence for enrichment
of ASD genes in synaptic and subsynaptic compartments. As a further step we
took advantage of emerging whole exome sequencing data in ASD to determine
whether there was an enrichment of de novo variation in synaptic and subsynaptic
compartments. RESULTS: We observed strong enrichment of ASD genes in the murine or
human synaptic proteome (P < 1.8x10-4, hypergeometric test). Remarkably,
much of this enrichment could be traced to just two subsynaptic proteomes, that
of the NMDA-receptor complex (NRC) and that of the AMPA-receptor complex
(ARC). In contrast, the metabotropic glutamate receptor pathway, previously
hypothesized to be broadly implicated in ASD based on Fragile X syndrome,
did not show such enrichment. In validation experiments we found that these
pathways were also enriched for genes mutated in intellectual disability (ID). As we had two independent datasets (one for ASD and one for ID) we were able
35
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Systems Biological Analyses implicate Glutamate Signaling
Abnormalities in Autism and in Intellectual Disability:
Implications for Psychopharmacology
Tuesday, Poster #111 (continued)
Joseph Buxbaum
to assess whether using existing protein-protein interaction (PPI) databases we
were able to identify known neurodevelopmental genes. In these analyses, genes
that were associated with ASD were significantly enriched for ID genes and vice
versa. This provided a strong rationale for using PPI databases to assess novel
ASD findings. We therefore made use of emerging data from a large-scale NIMH
and NHGRI-funded whole exome sequencing study (MH089025, Mark Daly,
communicating PI, Joseph Buxbaum, Bernie Devlin, Richard Gibbs, Gerard
Schellenberg, James Sutcliffe, collaborating PI’s), examining de novo mutations
in ASD trios to determine whether there was an enrichment of genes that were
associated with the NRC and ARC genes found in ASD and ID. We developed a
distance metric (Di) that assessed the average distance between a novel gene and
the prior list of ASD and ID genes, using a background PPI network, comparing
case to control variants. Even with data from only a modest number of trios
available to date, we found a significant (P<0.02, t-test) reduction in average
distance for case variants, indicating that amongst the de novo variants there is
an enrichment of genes that closely associated with prior ASD genes, particularly
those in the NRC and ARC complexes.
DISCUSSION: These studies highlight several important points. First,
ionotropic glutamate receptor signaling appears to be broadly disrupted in both
ASD and ID. Second, in spite of diverse molecular origins in specific cases of
ASD and/or ID certain compartments and pathways are recurrently impacted. Identifying pathways impacted by multiple independent mutations indicates that
novel therapeutics that target such pathways can have broader benefit. Third,
the findings underscore the role of synaptic function in these developmental
disorders and provide a strong rationale for using neurobiological approaches
in model systems to understand pathophysiology. Finally, these studies indicate
that systems biological approaches using unbiased datasets can be useful for
identifying additional genes in ASD and ID.
36
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Small and Whole Transcriptome RNA Sequencing identifies
Key Regulation Patterns in the Medial Prefrontal Cortex of the
Alcohol Dependent Rat
Wednesday, Poster #90
Jenica Tapocik, Estelle Barbier, Jesse Schank, Kornel Schuebel, Zhifeng Zhou,
Qiaoping Yuan, David Goldman, Markus Heilig
National Institutes of Health, National Institutes of Alcohol Abuse and
Alcoholism
BACKGROUND: MicroRNAs (miRNAs) are small interfering RNAs that
regulate gene expression by binding and inhibiting target mRNAs. miRNAs
expressed in the brain are critical for synaptic development and plasticity. Chronic ethanol exposure can cause lasting changes in the medial prefrontal
cortex (mPFC), in part due to changes in expression of genes involved in synaptic
plasticity. Using next-generation sequencing, we cataloged whole transcriptome
(WT) and miRNA expression alterations in the mPFC to further our understanding
of the global regulation patterns associated with alcohol dependence. METHODS: Rats were exposed to alcohol vapor for 7 consecutive weeks. Blood
alcohol concentrations (BACs) were taken weekly. Three weeks after exposure,
the mPFC was harvested using the atlas of Paxinos and Watson as reference. Total
RNA was isolated from the mPFC and run through the WT RNA sequencing
and small RNA sequencing protocol (n = 4/group). Log2 transformation and
normalization of the raw data using the Limma package from Bioconductor was
used. WT and Small RNA sequence reads were mapped to rat genomic sequences
(UCSC rn4) using Bowtie. Bioinformatics analysis (Ingenuity Pathway Analysis;
GO analysis) and the miRNA Sanger Database were used to determine miRNAgene interactions, pathways and functions, and networks involved in alcohol
dependence. RESULTS: For the mPFC, we generated a total of 3.4 million and 10.3 million
reads for WT and small RNA sequencing, respectively. Quantile probability plots
identified that the sequencing data are normally distributed. 566 genes and 20
miRNA families were found to be differentially regulated within our data set. Gene
Ontology analysis identified 150 of the 566 genes to be involved in neurological
37
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Small and Whole Transcriptome RNA Sequencing identifies
Key Regulation Patterns in the Medial Prefrontal Cortex of the
Alcohol Dependent Rat
Wednesday, Poster #90 (continued)
Jenica Tapocik
disorders. 16 genes are specifically involved in regulating the quantity of synaptic
vesicles and activation of synaptic transmission. The miRNA Sanger Data base
identified that the 8 top differentially expressed genes are potentially regulated by
miR-200. miR-200 is known to be upregulated by oxidative stress and members
of the miR-200 family regulate genes involved in neurogenesis. A subset of these
mRNAs and miRNAs were further confirmed by Taqman qRT-PCR.
DISCUSSION: Our results demonstrate a significant and lasting shift in miRNA
and mRNA expression patterns in the mPFC following a history of alcohol
dependence. Alcohol dependent-regulated miRNAs may contribute to longlasting drug-induced neuroplasticity by fine-tuning regulatory pathways that
modulate oxidative stress, neurotransmitter quantity and release, and synaptic
plasticity. Ongoing in vitro and in vivo experiments are currently determining the
functional link between our dysregulated miRNAs and their target genes.
38
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Selective Optogenetic Stimulation of Parvalbumin-positive
Basal Forebrain Neurons Reliably entrains Cortical Gamma
Oscillations and Promotes Wakefulness
Monday, Poster #40
Robert McCarley, James McKenna, James McNally, Karl Deisseroth, Robert
Strecker, Ritchie Brown, Radhika Basheer, Tae Kim
VA Boston Healthcare System/Harvard Medical School
BACKGROUND: The basal forebrain (BF) plays a crucial role in the
modulation of cortical activity across sleep-wake cycles via cortically projecting
cholinergic and non-cholinergic neurons. One of the more important facets of
cortical activation is the presence of gamma band (40Hz) activation, known to
be important in feature binding and to be impaired in schizophrenia. However,
little is known about the role of the BF in this important feature. Among noncholinergic neurons in BF, parvalbumin (PARV)-containing, gamma-aminobutyric
acid (GABA)ergic neurons are one of the important components. They project
to GABAergic PARV+ cortical neurons and their firing rates increase during
the electroencephalographic (EEG) low-voltage fast activity characteristic of
waking. However, their precise contribution to cortical activation and sleep-wake
regulation is not well understood. Therefore we sought to selectively express a
light-activated opsin (i.e. channelrhodopsin2, ChR2) in PARV-positive neurons
within BF to enable their selective activation using optogenetic stimulation, and
to determine the effect on the EEG and sleep-wake behavior. METHODS: To target channelrhodopsins selectively to BF PARV neurons,
adeno-associated viral vectors with double-floxed Channelrhodopsin2 (ChR2)eYFP were injected stereotactically into the BF of transgenic mice expressing Cre
recombinase under the control of the PARV promoter (PARV-Cre mice). Posthoc
immunohistochemistry was done for histological confirmation of selective
expression. To evaluate the physiological effect of activation of BF PARV neurons,
optical stimulation (laser light, 473 nm, 10 ms pulse width, various frequencies
from 2 to 60 Hz) was delivered through an optical fiber inserted into a guide
cannula targeting the BF. The effect on the sleep-wake cycle was investigated by
comparing one hour of baseline EEG with that of same time of day of one hour of
optical stimulation (40 Hz frequency, 5 s train duration, every 60 s). 39
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Selective Optogenetic Stimulation of Parvalbumin-positive
Basal Forebrain Neurons Reliably entrains Cortical Gamma
Oscillations and Promotes Wakefulness
Monday, Poster #40 (continued)
Robert McCarley
RESULTS: Immunohistochemistry, performed at least two weeks after injection
of the virus, confirmed high levels of double labeling of ChR2-eYFP (green)
and PARV (red) indicating selective expression of ChR2-eYFP in BF PARV
neurons. BF entrainment of the cortical EEG was particularly dramatic when
the BF stimulation was at the gamma oscillation frequency (40 Hz). Notably,
this entrainment could be reproducibly elicited over the course of an hour of
stimulation; each and every train of 40 Hz BF stimulation was reliably followed
by cortical 40 Hz activity. 20 Hz stimulation elicited a clear 40 Hz harmonic. The
sleep-wake behavior was altered by optical stimulation, increasing wakefulness
from 9.2 % to 45.2 % and decreasing NREM sleep from 75.3 % to 43.5 %,
excluding the 5 s of stimulation. DISCUSSION: We believe this BF PARV-specific elicitation of cortical gamma
oscillation has not been previously reported, and may represent an important
but unsuspected feature of BF activation and of generation of cortical gamma
oscillations. We conclude that optogenetic stimulation of PARV-positive BF
neurons entrains cortical rhythms, particularly those in the gamma range, and
enhances wakefulness. A role of abnormalities of BF in the abnormalities of
cortical gamma oscillations in schizophrenia appears to be an intriguing, but as
yet unexplored, possibility raised by these findings.
40
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Epigenetic Differences in the Developing Hippocampus and
Amygdala in a Novel Rat Model of Anxiety and Depression
Monday, Poster #7
Sarah Clinton, Rebecca Simmons, Danielle Simpson, Stanley Watson, Huda Akil
University of Alabama, Birmingham
BACKGROUND: Innate differences in human temperament strongly
influence how individuals cope with stress and predispose for specific types
of psychopathology. The present study examines developing brain circuits
in an animal model of temperamental differences to understand how altered
neurodevelopment may engender differences in emotional reactivity that are stable
throughout the animal’s life, and may put an individual at risk for a depressive/
anxious phenotype. METHODS: We utilize selectively-bred High Responder (bHR) and Low
Responder (bLR) rats that exhibit dramatic emotional behavior differences,
with bHRs exhibiting exaggerated novelty-exploration, aggression, impulsivity
and drug self-administration, and bLRs showing marked behavioral inhibition,
exaggerated anxiety- and depressive-like behavior. Using Affymetrix microarrays,
we assessed bLR/bHR gene expression in the developing brain on postnatal
days (P)7, 14, and 21, focusing on the hippocampus and nucleus accumbens,
two regions related to emotionality and known to differ in adult bLR/bHR rats. Stereological studies were used to examine potential differences in brain structure. Subsequent studies began to examine potential epigenetic mechanisms (e.g. DNA
methylation) that may contribute to putative gene expression differences in the
developing bHR versus bLR brain. To this end, we used in situ hybridization to
assess the mRNA expression of DNA methyltransferase 1 (DNMT1) – one of the
chief enzymes involved in DNA methylation in the brain. RESULTS: The microarray study revealed dramatic bLR/bHR gene expression
differences in the P7 and P14 hippocampus, with minimal differences in the
nucleus accumbens. Some of the most profound differences involved genes
critical for neurodevelopment and synaptogenesis. Stereological studies evaluated
hippocampal structure in developing bHR/bLR pups, revealing enhanced
hippocampal volume and cell proliferation in bLR animals. Results of the DNMT1
41
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Epigenetic Differences in the Developing Hippocampus and
Amygdala in a Novel Rat Model of Anxiety and Depression
Monday, Poster #7 (continued)
Sarah Clinton
in situ hybridization study point to potential bHR/bLR differences in DNA
methylation within specific subregions of the hippocampus (upper blade of
the dentate gyrus and the CA3 region). Interestingly, additional analysis also
uncovered similar bHR/bLR DNMT1 differences within select nuclei of the
amygdala – another key brain region that controls emotional behavior, in part via
reciprocal connections with the hippocampus. DISCUSSION: Our work-to-date with the bHR/bLR rat lines demonstrates the
heritability of the bHR and bLR behavioral phenotypes. Their underlying genetic
differences appear to drive distinct formation of the hippocampus, leading to
marked differences in hippocampal morphology and gene expression during the
first two weeks of life. The DNMT1 findings in the hippocampus and amygdala
suggest that the observed bHR/bLR differences in gene expression, brain
morphology and behavior may be linked to epigenetic changes occurring in early
life. Ongoing efforts will continue to examine DNA methylation status in the
developing bHR/bLR hippocampus and select amygdalar nuclei, and determine
whether early-life interventions (such as environmental enrichment) known to
improve LRs’ anxious/depressive phenotype act by influencing hippocampal
DNA methylation levels. Taken together, this body of work may provide important
insight into the possible genesis of individual differences in emotionalty and
related risks for the emergence of emotional disorders (e.g. the anxiety-prone
nature of bLRs or drug addiction proclivity of bHRs).
42
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Essential Role of Ventral Tegmental Area Dopamine Neurons in
Mediating the Induction and Rapid Reversal of Depression-Like
Behaviors
Wednesday, Poster #216
Ming-Hu Han, Dipesh Chaudhury, Jessica Walsh, Barbara Juarez, Mary Kay
Lobo, Herbert Covington III, Vincent Vialou, Hsing-Chen Tsai, Karl Deisseroth,
Eric Nestler, Allyson Friedman
Mount Sinai School of Medicine
BACKGROUND: The surprising rapid clinical effect of deep brain stimulation
in depressed patients supports the notion that depression is a neural circuit
disorder. In contrast, less is known if depression-like behaviors can be induced
or reversed rapidly in animal models. Our previous ex vivo and in vivostudies
showed that chronic social defeat consistently increased the firing rate and phasic
firing events of ventral tegmental area (VTA) dopamine (DA) neurons in the
brain reward circuitry of susceptible mice, but not of the resilient subgroup (Cell
2007; J Neurosci 2010). However, the physiological function of this increased
firing and its ionic mechanisms, as well as potential therapeutic targets, remain to
be elucidated. METHODS: Susceptible and resilient mice were segregated following a chronic
(10-day) social defeat paradigm, a well established model of depression. Utilizing
optogenetic techniques – Cre-dependent expression of channelrhodopsin-2
(ChR2) in tyrosine hydroxylase (TH)-Cre mice, phasic firing was induced by
high frequency optoactivation of ChR2 specifically in VTA DA neurons, and
behavioral tests (social interaction and sucrose preference) were performed in
behaving mice. The ionic mechanisms that underlie the hyperactivity of these
neurons were also examined in TH-GFP mice by use of electrophysiological
techniques. RESULTS: Consistent with our earlier findings, we found that optoactivation of
ChR2 (mimicking phasic firing) during sub-threshold defeat, a paradigm that does
not induce depressive-like behaviors in normal mice, increased social avoidance
behavior and reduced sucrose intake. Importantly, the same optoactivation of
these neurons during social interaction tests instantly induced similar susceptible
43
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Essential Role of Ventral Tegmental Area Dopamine Neurons in
Mediating the Induction and Rapid Reversal of Depression-Like
Behaviors
Wednesday, Poster #216 (continued)
Ming-Hu Han
behaviors in both resilient mice and the mice treated with sub-threshold defeat. These findings indicate that the phasic firing of VTA DA neurons encodes, and
is tightly linked to, the susceptible phenotype. Next, toward understanding the
ionic mechanisms of the higher VTA DA neuron firing, we previously found that
the current of Ih, an important channel in the transition between tonic and phasic
firing patterns, was upregulated in susceptible mice and that local infusion of
Ih inhibitors ZD 7288 and DK-AH 269 into the VTA normalized depressionlike social avoidance in susceptible mice within one hour after the infusion. This rapid effect is very different from classic antidepressants that take weeks to
show treatment efficacy. Surprisingly, in our ongoing studies, we also observed
that the antidepressant effect induced by a single-dose infusion of Ih inhibitor
DK-AH 269 lasted at least two weeks. More importantly, this long-lasting effect
was also seen with a single intraperitoneal dose of DK-AH 269. Following these
studies, we also found that chronic defeat induced a larger increase in Ih current
in resilient mice than in the susceptible subgroup. This finding suggests that the
force driving the pathological hyperactivity persists in resilient mice, and that
additional compensatory ionic mechanisms such as K+ channels are necessary to
drive the higher firing back to normal levels in resilient mice. Consistently, our
data showed that K+ currents were significantly increased selectively in resilient
mice, which is consistent with our earlier microarray data. In addition, we found
that local infusion of the K+ (KCNQ) channel activator flupirtine into the VTA
had similar rapid antidepressant effects as seen with Ih inhibitors. We are now
investigating the effect of systemic administration of this activator and others. DISCUSSION: Here we demonstrate that optogenetics is a powerful tool to
precisely imitate stress-induced physiological adaptations and reliably define
VTA DA neurons in the brain reward circuit as a therapeutic target. Interestingly,
optogenetically-induced phasic firing rapidly induces avoidance behavior during
interaction tests, even in resilient mice. Our data also strongly support that Ih
44
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Essential Role of Ventral Tegmental Area Dopamine Neurons in
Mediating the Induction and Rapid Reversal of Depression-Like
Behaviors
Wednesday, Poster #216 (continued)
Ming-Hu Han
channels are one of the passive pathological ion mechanisms that underlie
the susceptible firing increase, while K+ channels are an important active ion
mechanism that drives the pathological hyperactivity back to normal levels in
resilient mice. Importantly, passive ion channel inhibitors and active ion channel
activators that inhibit the higher pathological firing of VTA DA neurons are both
antidepressant and pro-resilient. Our studies provide fundamentally novel drug
targets forrapid or long-lasting antidepressants, which are mechanistically distinct
from predominant monoamine-based medications.
45
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Stress Exposure produces a Switch from Appetitive to Aversive
Signaling by Corticotropin-releasing Factor in the Nucleus
Accumbens Tuesday, Poster #12
Paul Phillips, Julia Lemos, Matthew Wanat, Jeffrey Smith, Beverly Reyes,
Elisabeth Van Bockstaele, Charles Chavkin
University of Washington
BACKGROUND: Stressors motivate an array of adaptive responses ranging
from “fight or flight” to an internal urgency signal facilitating long-term goals. However, traumatic or chronic uncontrollable stress promotes the onset of Major
Depressive Disorder where acute stressors lose their motivational properties. An emerging neurobiological substrate of clinical depression is the nucleus
accumbens which has the capacity to mediate a diverse range of stressor responses
by interfacing limbic, cognitive and motor circuitry. Here, we studied the actions
of the stress-related neuropeptide, corticotropin releasing factor (CRF) in the
nucleus accumbens on dopamine transmission and behavior. METHODS: Inmmunohistochemistry and transmission electron microscopy
were used to study anatomical interactions between CRF and dopamine terminals,
and fast-scan cyclic voltammetry was used to assess the effect of CRF on
dopamine transmission in the nucleus accumbens of mice. Subjective effects of
intrathecal administration of CRF into the nucleus accumbens was tested using a
place-conditioning paradigm. These experiments were carried out in stress-naïve
mice, or those exposed to a forced-swim-stress paradigm to elicit a depressive
phenotype. RESULTS: Within the nucleus accumbens, CRF-containing fibers were
interdigitated with dopamine-containing fibers. Both CRF R1 and R2 receptors
were co-localized with tyrosine hydroxylase expression, indicating that they are
expressed on dopamine-containing terminals. In the nucleus accumbens of stressnaïve mice, CRF increased dopamine release through co-activation of CRF R1
and R2 receptors. However, severe-stress exposure completely abolished this
effect without recovery for at least 90 days. This loss of CRF’s capacity to regulate
dopamine release in the nucleus accumbens was accompanied by a switch in the
46
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Stress Exposure produces a Switch from Appetitive to Aversive
Signaling by Corticotropin-releasing Factor in the Nucleus
Accumbens Tuesday, Poster #12 (continued)
Paul Phillips
perception of CRF from an appetitive to an aversive stimulus, indicating a
diametric change in the emotional response to acute stressors.
DISCUSSION: Here we demonstrate a specific defect in the regulation of
dopamine transmission in the nucleus accumbens in the genesis of depression. We show that severe stress disables the capacity of CRF to positively regulate
dopamine, removing CRF’s appetitive qualities, leaving a negative perceptual
bias. Indeed, depressive disorders produce a profound change in the perception
and behavioral response to acute stressors and other arousing environmental
stimuli that elicit CRF signaling. Taken together, our findings provide a
neurobiological mechanism for the affective shift from engagement of the
environment to withdrawal following severe stress, central to the manifestation
of Major Depressive Disorder.
47
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Altered Anxiety-Like Behavior in BDNF Val66Met Mice is
Rescued with Early-Life Fluoxetine
Tuesday, Poster #100
Iva Dincheva, Jianmin Yang, Tina Marinic, Kevin Bath, Helena Frielingsdorf,
Theresa Teslovic, James Kocsis, Barbara Hempstead, Francis Lee
Weill Cornell Medical College, Cornell University
BACKGROUND: The Val66Met single nucleotide polymorphism (SNP) in
the human brain-derived neurotrophic factor (BDNF) gene is associated with
alterations in brain anatomy and memory in humans, however its role in affective
disorders still remains unclear. Previously, we generated a variant BDNF mouse
(BDNFMet/Met) that reproduces the phenotypic hallmarks of human Met allele
carriers such as alterations in hippocampal anatomy and hippocampal dependent
memory. Variant BDNFMet is expressed in mouse brains at normal levels,
however its secretion from neurons is decreased. In addition, BDNFMet/Met
mice displayed an increased anxiety-like phenotype that emerges in adulthood,
which was not previously established in human carriers. This behavior is not
normalized by the selective serotonin reuptake inhibitor, fluoxetine, administered
in adulthood. We hypothesized that this behavioral consequence of the genetic
variant BDNFMet observed in the adult central nervous system was due to
reduced BDNF availability during postnatal development. We tested whether
transiently increasing BDNF levels via fluoxetine administration during an earlylife “sensitive period” when BDNF levels are peaking may prevent the emergence
of the increased anxiety-like behavior in BDNFMet/Met mice in adulthood. METHODS: BDNFVal/Val and BDNFMet/Met mice received fluoxetine
(160mg/L in drinking water) in three age groups: P21-P42, P40-P61, and
P60-P81. Anxiety-like behaviors were tested after a 3-4 week wash-out period. Hippocampal BDNF levels and neuronal morphology were also assessed. RESULTS: Early-life (P21-P42) fluoxetine treatment significantly reduced
anxiety-like behaviors in BDNFMet/Met mice in adulthood. Fluoxetine given
at other time periods to either BDNFVal/Val or BDNFMet/Met mice had no
significant effects on anxiety-like behaviors. BDNF levels increased at the end of
each treatment period, but were normalized with wash out. The highest levels of
48
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Altered Anxiety-Like Behavior in BDNF Val66Met Mice is
Rescued with Early-Life Fluoxetine
Tuesday, Poster #100 (continued)
Iva Dincheva
total BDNF levels were achieved when fluoxetine was administered during the
P21-P42 period.
DISCUSSION: Fluoxetine administration during a peri-adolescent period
(P21-P42) with subsequent wash–out leads to a rescue of an adult-onset anxietylike phenotype in BDNFMet/Met mice. This transient drug administration led
to peak BDNF levels, which may have increased BDNF availability during
this developmental window in BDNFMet/Metmice. The findings suggest that
correctly timed interventions which raise BDNF levels during a defined postnatal
period can rescue subsequent anxiety-like phenotypes in a mouse model of the
BDNF Val66Met SNP.
49
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
The Involvement of Alpha3-Containing Nicotinic Acetylcholine
Receptors in the Habenulo-Interpeduncular Pathway in Nicotine
Self-Administration
Wednesday, Poster #87
Christie Fowler, Qun Lu, Paul Kenny
The Scripps Research Institute
BACKGROUND: Allelic variation in the CHRNA3-CHRNA5-CHRNAB4
gene cluster, which encode the alpha3, alpha5 and beta4 nicotinic acetylcholine
receptor (nAChR) subunits, has been repeatedly associated with a significantly
increased risk of tobacco addiction in humans. Until recently, very little research
has focused on the role of these nAChR subunits in the motivational properties of
nicotine, the major addictive agent in tobacco smoke. We recently demonstrated
that alpha5-containing nAChRs in the medial habenulo-interpeduncular (MHbIPN) pathway exert an inhibitory influence on nicotine intake, particularly when
high doses of the drug are available for consumption. Similar to the alpha5
nAChR subunit, the alpha3 nAChR subunit demonstrates a restricted distribution
pattern in the brain, with particularly dense expression in MHb-IPN pathway. Here, we investigated the role of alpha3 nAChR subunits in regulating nicotine
consumption in rats. METHODS: A lentiviral vector was developed to selectively knockdown
expression of the alpha3 nAChR subunit gene (Lenti-CHRNA3). Rats were
injected with the Lenti-CHRNA3 or empty vector (Lenti-control) stereotaxically
into either the MHb or IPN. Thereafter, they were trained to respond for food
reward under a fixed ratio 5, time-out 20 sec (FR5TO20 sec) schedule of
reinforcement during 1-h daily testing sessions. Next, the rats were permitted
access to a training dose of nicotine (0.03 mg kg-1 per infusion) for intravenous
self-administration. After stable responding on the training dose, a full nicotine
dose-response function was characterized. RESULTS: Rats injected with the Lenti-CHRNA3 and Lenti-Control vectors
into the MHb or IPN similarly responded for food reward and acquired nicotine
self-administration behavior at the lower training nicotine dose. However, when
provided access to a range of doses, rats with selective knockdown of alpha3
50
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
The Involvement of Alpha3-Containing Nicotinic Acetylcholine
Receptors in the Habenulo-Interpeduncular Pathway in Nicotine
Self-Administration
Wednesday, Poster #87 (continued)
Christie Fowler
nAChR subunits in either the MHb or IPN persisted in responding for nicotine
infusions at higher levels than control rats at higher unit doses of the drug. DISCUSSION: These data demonstrate that alpha3-containng nAChRs in the
MHb and IPN exert an inhibitory influence on nicotine intake similar to that
previously reported for alpha5-containing nAChRs and suggest that alpha3 and
alpha5 nAChR subunits may combine to form a functional receptor subtype in the
MHb-IPN tract that negatively regulates nicotine intake. Together, these findings
reveal fundamental insights into the mechanisms of nicotine reinforcement that
are likely to have clinical and therapeutic implications. Supported by the National
Institute on Drug Abuse (DA026693 to CDF; DA020686 to PJK).
51
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
The Novel Opioid Receptor Modulator RDC-0313(ALKS 33)
reduces Olanzapine-induced Weight Gain and Adipose Accretion
in a Novel Nonhuman Primate Model of Antipsychotic-related
Weight Changes
Wednesday, Poster #206
Mark Todtenkopf, Reginald Dean, Michael Brunner, Michael Knopp, Daniel
Deaver
Alkermes, Inc
BACKGROUND: Antipsychotics (ATAP) can cause metabolic dysfunction and
weight gain. Female patients have a 3.6-fold increased risk of weight gain than
male patients (Hakko et al., 2006) and increases in BMI can be observed in as
little as 1 week post initiation of treatment (Kluge et. al., 2009). Weight gain
has been reported with risperidone, clozapine, sertraline and olanzapine (OLZ). ALKS 33 is a new chemical entity, currently under development for central
nervous system-related disorders that acts as an antagonist at μ opioid receptors,
with mixed agonist/antagonist activity at κ and δ receptors. We have previously
demonstrated in rats that co-administration with ALKS 33 mitigates OLZ-induced
weight gain whereas naltrexone does not (SFN, 2010). We are unaware of studies
using nonhuman primates (NHP) to investigate OLZ-induced changes in weight
gain or metabolic effects. Consequently, we investigated if: 1) OLZ would cause
similar metabolic changes in female cynomolgus monkeys as seen in humans;
and 2) would ALKS 33 attenuate OLZ-induced changes. METHODS: Three groups of late-adolescent female cynomolgus monkeys (n =
5/group; 4.04 ± 0.05 yrs) were used for this study: 1) vehicle control; 2) OLZ only;
and 3) OLZ with ALKS 33. Beginning two weeks prior to the study, monkeys
were given ad libitum access to a highly palatable, high caloric diet. On the day
prior to the start of treatment, monkeys were weighed and assigned to groups
using a randomized block design based on body weight (BW); average BW across
the 3 groups was 3.1 ± 0.08 kg on the day of randomization. Also, baseline whole
body CT scans were taken on the day prior to initiation of treatments. Monkeys
receiving OLZ were dosed twice daily (6 hours between doses) for 28 days. The
initial daily dose of OLZ was 1 mg/kg (PO, in 1% methylcellulose) and increased
52
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
The Novel Opioid Receptor Modulator RDC-0313(ALKS 33)
reduces Olanzapine-induced Weight Gain and Adipose Accretion
in a Novel Nonhuman Primate Model of Antipsychotic-related
Weight Changes
Wednesday, Poster #206 (continued)
Mark Todtenkopf
every 3 days to a daily dose of 6 mg/kg by Day 10. For group 3, ALKS 33 (0.4 mg/
kg, IM) was administered in the morning immediately after administration of OLZ. Weights were taken every 3 days and on Day 28 blood samples were collected for
serum chemistry analysis. A second CT scan was conducted on day 29.
RESULTS: BWs were relatively constant for two months prior to the initiation
of ad libitum feeding. Vehicle treated monkeys gained an average of 0.28 kg (9%
of Day 0 BW) over the 28-day study. This gain was attributed to the ad libitum
feeding of the highly palatable diet. Over the same 28-day period, OLZ-treated
monkeys gained an average of 0.46 kg (15% of Day 0 BW). This marked increase
in average weight gain was driven by 3 of the 5 monkeys who gained between
19.8 and 37.8 % of their initial
body weight. An accretion of adipose tissue was observed in all monkeys compared
to baseline control values. However, monkeys in the OLZ group gained relatively
more adipose tissue compared to the vehicle group. Also, there was a difference
in the location of adipose tissue deposition with the OLZ group showing more
abdominal fat accretion. Concentrations of triglycerides (TGs) and LDL were
higher in OLZ-treated monkeys (86.6 and 105.8 mg/dL, respectively) compared
to the vehicle group (62 and 87.8 mg/dL, respectively). In monkeys treated with
OLZ and ALKS 33 the average BW gain over the 28-days was only 0.08 kg
(2.6% of Day 0 BW). While these monkeys also gained adipose tissue, the extent
and distribution of fat was similar to that observed in the vehicle group and lower
than the OLZ-only group. Finally, co-administration of ALKS 33 prevented OLZinduced elevations in TGs and LDL concentrations. DISCUSSION: Based on these data, treatment of nonhuman primates with OLZ
for 28-days resulted in qualitative changes in weight, adipose tissue accretion,
TGs and LDLs similar to those reported in patients. Furthermore, OLZ-induced
changes were mitigated by co-administration of ALKS 33.
53
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Discovery of the First β-Arrestin-Biased Dopamine D2 Ligands for
Probing Signaling Pathways Essential for Antipsychotic Efficacy
Monday, Poster #204
Jian Jin, John Allen, Julianne Yost, Vincent Setola, Xin Chen, Maria Sassano,
Meng Chen, Niels Jensen, Sean Peterson, Prem Yadav, Xi-ping Huang, Stephen
Frye, William Wetsel, Marc Caron, Bryan Roth
University of North Carolina at Chapel Hill
BACKGROUND: G protein-coupled receptors (GPCRs) signal not only via
canonical pathways involving heterotrimeric large G proteins, but also via
non-canonical G protein-independent interactions with other signaling proteins
including, most prominently, β-arrestins. The process by which GPCR ligands
differentially modulate canonical and non-canonical signal transduction pathways
is a phenomenon known as “functional selectivity”. Such functionally selective
ligands preferentially engage either canonical or non-canonical GPCR pathways. Clearly, the discovery of ligands with discrete functional selectivity profiles will
be extremely useful for elucidating the key signal transduction pathways essential
for both the therapeutic actions and side-effects of drugs. However, only a small
number of functionally selective GPCR ligands have been reported to date. In
particular, although β-arrestin-biased ligands of Gq and Gs-coupled GPCRs
are known, β-arrestin-biased GPCR ligands that selectively activate β-arrestin
signaling pathways over Gi-coupled pathways have not been reported. In this
presentation, we report the discovery of the first β-arrestin-biased dopamine D2
ligands (UNC9975, UNC0006 and UNC9994) as chemical tools for probing
signal transduction pathways essential for antipsychotic efficacy. METHODS: Novel analogs of aripiprazole (OPC-14597), an FDA-approved
atypical antipsychotic drug, were designed and synthesized. These newly
synthesized compounds were evaluated in dopamine D2 receptor binding,
D2-mediated cAMP accumulation, and 3 orthogonal and complementary D2mediated β-arrestin-2 translocation assays (using the Tango, DiscoveRx, and
bioluminescence resonance energy transfer (BRET) assay technologies). In
addition, β-arrestin-biased compounds were evaluated in an extracellular
signal-regulated kinase phosphorylation (p-ERK) reporter assay to assess their
54
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Discovery of the First β-Arrestin-Biased Dopamine D2 Ligands for
Probing Signaling Pathways Essential for Antipsychotic Efficacy
Monday, Poster #204 (continued)
Jian Jin
effects on β-arrestin-mediated signaling. UNC9975 and UNC9994 were tested in
C57BL/6 wild-type (WT) and β-arrestin-2 knockout (βARR2 KO) mice for their
ability to inhibit amphetamine- or phencyclidine-stimulated hyperlocomotion. A
drug-induced catalepsy model was used to assess potential extrapyramidal sideeffects of these compounds in WT and β-ARR2 KO mice.
RESULTS: We intensely explored 4 regions of the aripiprazole template and
prepared more than 150 novel compounds. Through this robust diversity-oriented
modification of the aripiprazole scaffold, we discovered UNC9975, UNC0006
and UNC9994 as the first functionally selective, β-arrestin-biased dopamine D2
ligands, which were simultaneously neutral antagonists of Gi-regulated cAMP
production and partial agonists for D2R/β-arrestin-2 interactions. In the D2mediated cAMP accumulation assay, UNC9975, UNC0006 and UNC9994 did
not activate this Gi-coupled signaling pathway, in stark contrast to aripiprazole,
which was a potent partial agonist with an Emax of 80%. In the 3 orthogonal and
complementary D2-mediated β-arrestin-2 translocation assays, these compounds
were potent partial agonists for β-arrestin-2 recruitment to D2 receptors. In
addition, UNC9975 and UNC0006 potently activated pERK as partial agonists
and co-expression of β-arrestin-2 and GRK2 significantly enhanced the efficacy
of these compounds. Importantly, UNC9975 displayed potent antipsychotic-like
activity without inducing motoric side-effects in inbred C57BL/6 mice in vivo. Genetic deletion of β-arrestin-2 simultaneously attenuated the antipsychotic
actions of UNC9975 and transformed it into a typical antipsychotic drug with
a high propensity to induce catalepsy. Taken together, our results suggest
that β-arrestin signaling and recruitment can be simultaneously a significant
contributor to antipsychotic efficacy and protective against motoric side-effects. DISCUSSION: We discovered the first functionally selective, β-arrestin-biased
dopamine D2 ligands through a combined medicinal chemistry and comprehensive
pharmacological profiling approach. Significantly, evaluation of these novel
55
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Basic
Kona 5
Discovery of the First β-Arrestin-Biased Dopamine D2 Ligands for
Probing Signaling Pathways Essential for Antipsychotic Efficacy
Monday, Poster #204 (continued)
Jian Jin
chemical probes in WT and β-ARR2 KO mice show that β-arrestin can emerge as
an important contributor to both antipsychotic drug efficacy and antipsychotic sideeffects. This study represents a successful proof-of-concept for how functionally
selective GPCR ligands can be discovered and validated. The β-arrestin-biased
D2 ligands discovered here are valuable tools for the biomedical community to
further investigate D2R signaling in health and disease.
56
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Hot Topics
Clinical
Chair: Anissa Abi-Dargham
4:00 p.m.
4:15 p.m.
Fetal Programming of Sex Differences in Stress Response
Circuitry, Endocrine and ANS Deficits in Adulthood:
Implications for Understanding Sex Differences in Comorbidity
of Depression and CVD Risk
Jill Goldstein
Maternal Brain Responses to Baby-Stimuli are modulated by
Psychopathological Risk
James Swain
4:30 p.m.
Social Rejection activates Endogenous Opioid Systems David Hsu
4:45 p.m.
Deep Brain Stimulation in Treatment Resistant Alcohol
Addiction - Longterm Results of the Magdeburg Pilot Study
Ulf Mueller
5:00 p.m.
Baclofen as a Novel Pharmacotherapy for Alcohol Dependence:
Preliminary Findings from a Human Laboratory Double-Blind
Placebo-Controlled Randomized Study
Lorenzo Leggio
5:15 p.m.
Ketamine for Treatment-Refractory Obsessive-Compulsive
Disorder
Michael Bloch
5:30 p.m.
The Alpha7 Neuronal Nicotinic Receptor (NNR) Modulator TC5619 had Beneficial Effects and was Generally Well Tolerated in
a Phase 2 Trial in Cognitive Dysfunction in Schizophrenia (CDS) David Hosford
57
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Hot Topics
Clinical (continued)
Chair: Anissa Abi-Dargham
5:45 p.m.
EVP-6124, an Alpha-7 Nicotinic Partial Agonist, produces
Positive Effects on Cognition, Clinical Function, and Negative
Symptoms in Patients with Chronic Schizophrenia on Stable
Antipsychotic Therapy Dana Hilt
6:00 p.m.
Gradual and Overlapping Antipsychotic Switch Strategies
are associated with Less All-Cause Discontinuation in
Schizophrenia: Results from a Meta-analysis of Different Switch
Strategies
Christoph Correll
6:15 p.m.
The Hypo-Functional 7-Repeat Allele of DRD4 predicts both
Objective and Reported Fat Intake in 4- to 6- Year Old Girls
Robert Levitan
58
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Fetal Programming of Sex Differences in Stress Response
Circuitry, Endocrine and ANS Deficits in Adulthood:
Implications for Understanding Sex Differences in Comorbidity
of Depression and CVD Risk
Tuesday, Poster #214
Jill Goldstein, Laura Holsen, Brandon Abbs, Sara Cherkerzian, Anne Klibanski,
Mady Hornig, Richard Sloan, Stephen Buka
Brigham and Women’s Hospital/Harvard Medical School/Massachusetts
General Hospital
BACKGROUND: Comorbidity of major depressive disorder (MDD) and
cardiovascular disease risk (CVD), with a prevalence of ~20%, will be the number
one cause of disability worldwide by 2020, and is significantly higher in women
than men. We are investigating shared pathophysiology beginning in fetal brain
development that implicates a prenatal stress model. Preeclampsia and growth
restriction, implicated in MDD and CVD, are associated with maternal immune
activation and fetal disruption of the development of the HPA axis. Here we
hypothesized that these fetal risk factors significantly predict higher comorbidity
in women than men. Further, their impact will be expressed in adulthood as sexspecific stress response circuitry, and endocrine and autonomic nervous system
(ANS) deficits that mediate the association between fetal risk and MDD and CVD
risk in women. METHODS: In a 40-year cohort study with mothers followed through pregnancy
and sera stored at NIH, 295 fetally-exposed and unexposed adult siblings were
recruited. Pro- and anti-inflammatory prenatal assays included IL-1β, IL-6,
TNF-α, IL-10, IL-8. 60 of these adult offspring were scanned using fMRI tasks
of the stress response circuitry (~half exposed/half unexposed; 26 with recurrent
DSM-IV MDD in remission). Subjects viewed images with negative valence/high
arousal vs. neutral valence/low arousal stimuli. 3T Siemens MR scanner was used
and SPM8 for analyses of blood oxygen-level dependent (BOLD) signal changes
in hippocampus (HIPP), amygdala, anterior hypothalamus (aHYP), orbitofrontal
and medial prefrontal cortices (OFC; mPFC), anterior cingulate cortex (ACC),
and periacqueductal gray (PAG). Hormones, heart rate, and high frequency R-R
interval variability (HF-RRV)) were collected during scanning timed to stress
59
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Fetal Programming of Sex Differences in Stress Response
Circuitry, Endocrine and ANS Deficits in Adulthood:
Implications for Understanding Sex Differences in Comorbidity
of Depression and CVD Risk
Tuesday, Poster #214 (continued)
Jill Goldstein
response (e.g., DHEAS, cortisol, E2, progesterone, and testosterone). General
linear models were used to relate adult hormone and HF-RRV and prenatal
cytokines to BOLD signal changes in fetal-exposed vs. unexposed subjects,
MDD and sex. RESULTS: Fetal exposure was significantly associated with low HF-RRV in
response to stress 40 years later (F = 6.06, P = 0.01), which was 3 times greater
among MDD vs. non-MDD. Fetal-exposed females had a significantly higher
risk for comorbidity of MDD and low HF-RRV than males (RR= 1.38, p <
0.01), which was significantly related to TNF-α in maternal sera of exposed vs. unexposed comorbid women (t=2.53, p<.01). In response to stress, there were
significantly greater BOLD changes in aHYP (p<.01; ES= 1.0), HIPP (p<.04;
ES=.67), and ACC (p<.05; ES= .74) and hypoactivity in OFC (ES= .48) in fetalexposed women (the latter of which was the only significant finding in exposed
men). Fetal-exposed female MDD cases showed the greatest hyperactivity in
aHYP (ES=1.5) and hypoactivity in ACC, OFC, and HIPP (ESs = .80; 1.15; .62),
suggesting hyperarousal and lack of cortical and HIPP control in fetal-exposed
MDD women. Key stress response brain regions were significantly associated
with loss of parasympathetic cardiac regulation. Higher TNF-α/IL-10 and IL-6
were significantly related to lower BOLD changes in HIPP and ACC, which cooccurred with disruptions in HPA and HPG hormones (i.e, lower DHEAS, E2,
and testosterone, and higher progesterone and cortisol in response to stress). DISCUSSION: Findings suggest shared fetal risk factors for the comorbidity of
MDD and CVD risk in women. They may, in part, act through a disruption of the
maternal immune response resulting in sex-specific effects on offspring’s HPA
circuitry, expressed in adulthood as brain activity, endocrine and ANS deficits. Thus, the fetal programming of the stress response circuitry may be important for
understanding vulnerability to MDD and CVD risk, particularly in women.
60
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Maternal Brain Responses to Baby-Stimuli are modulated by
Psychopathological Risk
Monday, Poster #81
James Swain, S. Shaun Ho, Katherine Rosenblum, Eric Finegood, Patricia
Richardson, Leyla Akce, Sheila Marcus, K. Luan Phan, Maria Muzik
University of Michigan
BACKGROUND: Parenting constitutes an evolutionarily conserved set of
attachment behaviors and associated thoughts that contribute to responses to a
distressed infant. These include immediate attention to needs and safety as well
as soothing and caring that shape child development. Parenting behaviors and
thoughts are influenced by a mother’s own life experiences and repertoire of
parenting-related thoughts. Upon hearing the distress signal of baby-cry, maternal
neurophysiological systems respond to ensure adaptive responses, including
increases in motivation to approach and decreases avoidance drives as a function
of parenting-related thoughts. However, the capacity of mothers to generate
caring behaviors and thoughts may be compromised by the accumulation of
risk factors, including a history of child abuse, post-traumatic stress disorder,
and/or major depression disorder. Thus, in this functional magnetic resonance
imaging (fMRI) study, we hypothesize that: 1. Mothers brain responses to babycry, as a function of psychopathological risk, will be associated with decreased in
positive motivation, rewarding and caring regions and increased avoidance and
fear regions 2. Mothers at risk for depression will have inhibited self-reflection/
empathy brain responses in response to personalized messages about parenting
METHODS: Eighteen mothers of 2-7 year old children were assessed for current
and cumulative psychopathological risk, including histories of child abuse, posttraumatic stress disorder, major depression. In addition, the working model of
the child interview (WMCI) was administered. In a Phillips 3T scanner, the
participants underwent two types of task in a pseudo-randomized block-design:
1. An auditory baby-cry task, in which mothers listened to 30 second-blocks of 3
conditions and a pattern-matched white noise preceded by one of three primers:
“a baby crying”, “your baby crying”, or “you yourself are crying as a baby”. 2. A personally tailored message task, in which mothers were shown excerpts from
their own responses to the WMCI, administered within a few days of the brain
61
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Maternal Brain Responses to Baby-Stimuli are modulated by
Psychopathological Risk
Monday, Poster #81 (continued)
James Swain
scanning over two conditions: directed feedback (e.g., “You find it most difficult
to handle when [child’s name] screams.”) and open-ended feedback (e.g., “Think
about when [child’s name] screams.”). In addition there was a non-parentingrelated control message condition (e.g., “Think about the speed of the internet”). All data were analyzed with SPM 8. RESULTS: For the baby-cry task, we found the following: Listening to “a
baby-crying” vs. white noise differentially activated salience-related regions of
extended amygdala and insula, and that these neural responses increased with
cumulative psychopathological risk. Listening to “your baby crying” vs. “a baby
crying” differentially activated reward-related and salience regions of nucleus
accumbens, and hippocampus. Listening to “you yourself as a baby crying”
vs. “a baby crying” differentially activated anxiety- or stress-related regions
of middle frontal gyrus, caudate, posterior insula, and habenula proportionally
with cumulative psychopathological risk. To benchmark the neural regulation
between positive and negative motivations, we contrasted “you yourself as a
baby crying” with “your baby crying” and, as predicted, found that mothers with
higher cumulative psychopathological risks showed reduced neural responses in
the nucleus accumbens and hippocampus associated with positive motivation, but
enhanced neural responses in the hypothalamus, midbrain, amygdala, caudate,
anterior cingulate cortex (ACC), insula, and habenula associated with negative
emotions of fear and avoidance. For the maternal interview task, we contrasted
directed + open-ended feedback vs. control to find differential activation in the
self-reflection regions of the dorsomedial prefrontal cortex, precuneus, posterior
cingulate (PCC) cortex, and ACC. However, grouping subjects according
psychopathological risk showed reductions in neural response in PCC and
precuneus in to high vs. low risk. These results suggest a link between maternal
psychopathology and diminished neural response in brain regions that are integral
to self-reflection.
62
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Maternal Brain Responses to Baby-Stimuli are modulated by
Psychopathological Risk
Monday, Poster #81 (continued)
James Swain
DISCUSSION: Human parenting involves behaviors driven by key stimuli like
baby-cries, as well as thoughts about being a parent. This work makes use of
novel neuroimaging tasks to explore the brain activity underlying behaviors and
thoughts across psychopathology risk. Previous mood and anxiety appear to alter
brain responses important for parenting, suggesting opportunities for intervention
and improved child mental health.
63
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Social Rejection activates Endogenous Opioid Systems
Wednesday, Poster #124
David Hsu, Tiffany Love, Heng Wang, Kortni Meyers, Kathleen Hazlett, Lisong
Ni, Sara Walker, Steven Korycinski, Robert Koeppe, Jennifer Crocker, Scott
Langenecker, Jon-Kar Zubieta
University of Michigan
BACKGROUND: Endogenous opioids, which alleviate physical pain, are
also thought to have broader effects on the regulation of responses to stressors,
including social distress in several species including chicks, rats, guinea pigs,
dogs, and monkeys. It is not known if endogenous opioids perform a similar
function in humans. In this study, it was hypothesized that the endogenous opioid
system would respond to social rejection, an explicit declaration that one is not
liked, by measuring μ-opioid receptor binding in vivo at baseline and during
social rejection. Reductions in receptor availability (i.e., binding potential, BP)
are indicative of endogenous opioid release and μ-opioid receptor activation. METHODS: 10 healthy volunteers (7 females; mean age, 33 ± 11 years) were
clear of active medical illness, current or past psychiatric disorders, and had
no history of medication at the time of study. [11C]carfentanil, a selective and
specific μ-opioid receptor radioligand, was intravenously administered during
positron emission tomography (PET). BP was calculated using Logan plots with
occipital cortex as a reference region. Participants completed online ratings for
profiles of the opposite sex, and only the most-liked profiles were presented to
increase feedback saliency. During the scan, participants viewed these profiles
along with feedback that they were not liked (12 profiles/block). Baseline blocks
contained a similar visual presentation with no feedback. Blocks were presented
in a randomized, counterbalanced design across participants, and measures of
emotional state were obtained after each feedback. A prioriregions of interest
included the ventral striatum, amygdala, thalamus, insula, anterior cingulate
cortex, and periaqueductal gray (PAG). These regions have high levels of μ-opioid
receptors and have been previously shown to respond to physical pain. RESULTS: Measures of emotional state indicated that participants felt
significantly more “rejected” during rejection compared to baseline blocks (P
= 0.006). Reductions in μ-opioid binding potential was found during rejection
64
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Social Rejection activates Endogenous Opioid Systems
Wednesday, Poster #124 (continued)
David Hsu
compared to baseline blocks in the bilateral ventral striatum (left, P = 0.002; right,
P = 0.003), left amygdala (P = 0.006), midline thalamus (P = 0.006), bilateral
insula (left, P = 0.02; right, P = 0.01), right anterior cingulate cortex (P = 0.03),
and PAG (P = 0.03). In contrast, no significant decreases in binding were found
during baseline compared to rejection blocks.
DISCUSSION: This is the first demonstration that the endogenous opioid system
responds to social rejection. The PAG, midline thalamus, amygdala, ventral
striatum, insula, and anterior cingulate comprise a pathway by which stressrelated information may influence emotion, mood, and motivation. Decreased
μ-opioid BP reflects the release of endogenous opioids interacting with μ-opioid
receptors, which may function to regulate activity along this pathway. Negative
repercussions of social rejection include lowered self-esteem, major depressive
disorder, social anxiety disorder, substance abuse disorders, risk-taking behavior,
and aggression. The results suggest that endogenous opioids are involved in
the link between social rejection and the potential to develop these psychiatric
illnesses and maladaptive behaviors.
65
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Deep Brain Stimulation in Treatment Resistant Alcohol
Addiction - Longterm Results of the Magdeburg Pilot Study
Monday, Poster #169
Ulf Mueller, Bernhard Bogerts
University Hospitals Magdeburg
BACKGROUND: Worldwide, over 76 million people suffer from alcohol use
disorders. They are at increased risk for early death by somatic complications
with a reduced life expectancy by over 20 years and show an increased suicide
risk compared to the nonpsychiatric ill. Unfortunately, following current
treatment options, over 50% of alcoholics do not achieve long-term abstinence
and subsequently develop severe complications. Aside from these individual
medical consequences, addiction causes an immense financial damage to society. Thus, finding new treatment options remains a leading challenge in clinical
psychiatry. In the last years, there has been increased evidence that chronic
alcohol consumption leads to a dysfunction of the Nucleus accumbens (NAc),
thereby causing cue-induced craving, which is considered one of the main reasons
why patients cannot stay abstinent and relapse. The observation of remission of a
secondary alcohol addiction in a patient who received deep brain stimulation of
the NAc for treatment resistant anxiety disorder led to the initiation of this trial. METHODS: On the basis of an OFF-label single patient use, 5 patients with very
severe treatment resistant alcohol addiction for many years received bilateral deep
brain stimulation to the Nucleus accumbens at our centre. They were operated in
general anaesthesia using a modified Riechert-Mundinger stereotactic system with
a deep fronto-lateral approach. Quadrupolar electrodes (Medtronic Quad 3389)
for DBS were placed bilaterally in the shell region of the nucleus after presurgical
planning using high resolution T1-weighted MRI-scans. Subsequently, electrodecables were connected to an impulse generator located beneath the left pectoral
muscle (Kinetra, Medtronic®) similar to a cardiac pacemaker. RESULTS: All patients reported an immediate and ongoing absence of craving
for alcohol. All patients show a severe reduction of drinking days and amount
of drinks/day. 4/5 patients are continuously employed again after years of
unemployment. 2 patients remain completely abstinent for 4 years. Aside from
4 very short relapses for 2-3 days each a third patient remains abstinent as well. 66
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Deep Brain Stimulation in Treatment Resistant Alcohol
Addiction - Longterm Results of the Magdeburg Pilot Study
Monday, Poster #169 (continued)
Ulf Mueller
DISCUSSION: DBS of the NAc showed to be safe and effective in this small
sample of 5 patients with a very severe treatment resistant alcohol addiction. As
relapses to drinking are considered to be a part of disease in this subsample of
severely ill patients, hard-reduction strategies such as reducing the days drinking
and drinks per day are increasingly being considered as a primary treatment
option. Therefore, our results even exceed this goal clearly in 3 out of 5 patients,
whereas the other patients still fulfil these criteria. Based on this favourable pilot
data, we believe that a randomized and sham-controlled clinical trial is justifiable
and required to further study the possibility of DBS as a novel treatment option
in alcohol addiction. The prospective, randomized, double-blinded and shamcontrolled multicenter study DeBraSTRA (Deep Brain Stimulation in Treatment
Resistant Alcoholism) has been authorized by federal authorities in Germany
and will be funded entirely independent from industries by the German Research
Foundation.
67
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Baclofen as a Novel Pharmacotherapy for Alcohol Dependence:
Preliminary Findings from a Human Laboratory Double-Blind
Placebo-Controlled Randomized Study
Wednesday, Poster #187
Lorenzo Leggio, George Kenna, William Zywiak, Steven Edwards, Samuel
Fricchione, Tonya Tavares, Jessica Shoaff, John McGeary, Robert Swift
Brown University Center for Alcohol and Addiction Studies
Catholic University of Rome, Institute of Internal Medicine
BACKGROUND: Prior studies have identified the selective GABAB receptor
agonist baclofen as a possible novel pharmacotherapy for alcohol dependence
(AD). We conducted a human laboratory study to investigate putative biobehavioral
mechanisms by which baclofen reduces drinking [attenuating urges to drink,
decreasing attention to alcohol cues, attenuating pleasurable affects of alcohol
and/or increasing the aversive effects of alcohol, attenuating pleasurable effects
of slip]; by ways of a cue-reactivity (CR) experiment, followed by an alcohol
self-administration (ASA) experiment. METHODS: This was a between-subject double-blind randomized controlled
pilot trial. We enrolled 14 non-treatment seeking alcohol dependent (AD)
participants who received baclofen 10mg t.i.d. or an ‘active’ placebo (i.e. cyproheptadine 2mg t.i.d.) for 8 days. Then, participants came to our lab to
perform a session consisting of an alcohol cue-reactivity (CR) experiment,
followed by an alcohol self-administration (ASA) experiment. Measurements of
craving [Alcohol Use Questionnaire (AUQ)], attention [Alcohol Attention Scale
(AAS)], salivation, sedation and stimulation [Biphasic Alcohol Effects Scale
(BAES)] were performed. Moreover, stress-related hormones were determined,
i.e. cortisol, prolactin (PRL) and growth hormone (GH). RESULTS: 13 out of the 14 participants completed the laboratory session. No
severe or serious side effects were reported during the study. There were no
significant differences in terms of craving or attention during the CR. As for
stimulation and sedation, we found an increase in the baclofen group compared
to placebo, stimulation: F(1,92)= 32.4, p<.001 and for sedation, F(1,88)=28.53,
p<.001. During the ASA, we found an effect size d of .76, t(6.4)=1.43 with means
68
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Baclofen as a Novel Pharmacotherapy for Alcohol Dependence:
Preliminary Findings from a Human Laboratory Double-Blind
Placebo-Controlled Randomized Study
Wednesday, Poster #187 (continued)
Lorenzo Leggio
of 1.43 vs. 0.17 drinks with p=.20. Furthermore, we found a similar robust effect
size t(12)=1.20, p=.25 of reported drinking during the two days before the ASA. We also found that after controlling for baseline GH, there was a trend toward
significance for the baclofen group and lower GH [r(5)=-.70, p=.08; repeated
measures ANCOVAs for GH determined between CR and ASA]. At the end of
the ASA, GH levels also correlated positively with the AUQ craving score r(7) =
0.76, p=.03. DISCUSSION: Although the small sample size, there was a non-significant
trend toward statistical significance with a robust effect (d=0.76) in the amount
of alcohol consumed during the ASA as well as in reducing standard drinking
units (SDUs) consumed on the two days prior to the lab session (after baclofen
dose was titrated up). Also, this study design was unique in that this is one of the
first studies to conduct the ASA after the CR experiment, resulting in additional
alcohol cues other than the priming drink in the ASA. Of particular interest is
baclofen’s effects on stimulation and sedation during the ASA. It’s been suggested
that baclofen reduces alcohol drinking by reducing the individual’s craving for
alcohol. This study shows that the reduction in craving might be secondary to
the stimulation and sedation effects from baclofen. Interestingly, sedation was
‘controlled’ for by using an ‘active’ placebo that induces sedation. Additionally,
the trend toward significance in lower GH levels for baclofen group and the
positive correlation between the AUQ scores and GH levels at the end of the ASA
suggest a possible link between lower GH and a reduction in alcohol drinking. These several mechanisms may account for the ability of baclofen to reduce
alcohol drinking even after individuals have been exposed to alcohol cues and
have received an alcohol ‘priming’ (a model of ‘lapse’). Future research is needed
to investigate these preliminary findings.
69
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Ketamine for Treatment-Refractory Obsessive-Compulsive
Disorder
Tuesday, Poster #178
Michael Bloch, James Leckman, John Krystal, Zubin Bhagwagar, Gerard
Sanacora, Christopher Pittenger
Yale University
BACKGROUND: Roughly one-third of patients with obsessive-compulsive
disorder (OCD) experience little clinical benefit from first-line interventions
such as pharmacotherapy with selective serotonin reuptake inhibitors (SSRIs)
or cognitive behavioral therapy (CBT). Furthermore, the full treatment benefits
of first-line interventions are only realized after two to three months. Limited
symptom relief and delay of symptom relief from first-line treatments are sources
of substantial morbidity and decreased quality of life in OCD patients. Convergent
evidence from neuroimaging, genetic and pharmacological studies suggests that
glutamate abnormalities may contribute to the pathogenesis/or pathophysiology
of OCD. Ketamine is a potent noncompetitive antagonist of the N-methyl-Daspartate (NMDA) receptor, a major type of glutamate receptor in the brain. Several trials have reported that a single dose of ketamine (0.5 mg/kg, delivered
intravenously over 40 minutes) had rapid antidepressant effects in depressed
patients.
METHODS: We conducted a pilot, open-label trial of ketamine (0.5mg/kg IV
over 40 minutes) in patients with treatment-refractory OCD. Patients had severe
OCD symptoms (Y-BOCS>24) despite at least two SSRI trials of adequate dose
and duration. Clinical ratings of OCD (Y-BOCS) and depression (HAM-D) were
performed at baseline, 1, 2 and 3 hours and 1, 2, 3, 5 and 7 days after ketamine
infusion. Response for OCD was defined as a greater than 35% improvement
in symptoms from baseline at any time between 1-7 days following infusion. Response in depressive symptoms was defined as a greater than 50% reduction
in HAM-D ratings from baseline anytime between 1-7 days following infusion. RESULTS: We enrolled 8 patients with treatment-refractory OCD (6 with
comorbid depression). None experienced a clinical response in OCD symptoms
during the week following ketamine infusion. On average, the reduction of OCD
70
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Ketamine for Treatment-Refractory Obsessive-Compulsive
Disorder
Tuesday, Poster #178 (continued)
Michael Bloch
symptoms peaked 1 day following infusion (improving by 10±12%; not
significant). By contrast, 3 of 6 patients with OCD and comorbid depression
experienced a clinical response in their depressive symptoms. Improvement in
depressive symptoms peaked 2 days following ketamine infusion with a 45±40%
average reduction in symptoms (p=0.04). DISCUSSION: We observed a clear dissociation between OCD and depressive
symptoms in response to ketamine infusion. This pilot trial demonstrates no
effect of ketamine, an NMDA antagonist, in the treatment of OCD. By contrast,
we demonstrate a potent, though short-lived, antidepressant effect of ketamine
infusion, consistent with previous studies.
71
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
The Alpha7 Neuronal Nicotinic Receptor (NNR) Modulator TC5619 had Beneficial Effects and was Generally Well tolerated in
a Phase 2 Trial in Cognitive Dysfunction in Schizophrenia (CDS)
Monday, Poster #48
David Hosford, Geoffrey Dunbar, Jeffrey Lieberman, Anthony Segreti
Targacept, Inc.
BACKGROUND: TC-5619 is a selective alpha7 NNR modulator. TC-5619 had
efficacy in preclinical models of memory and schizophrenia1 and was generally
well tolerated in phase 1 trials in healthy volunteers, demonstrating a robust
improvement in attention at 6.8mg in a multiple rising dose study. This trial
tested the effect of TC-5619 on cognitive and negative symptoms in subjects with
schizophrenia. METHODS: In the US and India 185 outpatients (18 – 65 years; male 69%; 46%
tobacco users) with stable schizophrenia and taking quetiapine or risperidone
were randomized to 12 weeks of placebo (n = 91) or TC-5619 (n = 94: 1mg po qd
Day 1 to Week 4; 5mg po qd Week 4 to Week 8; 25 mg po qd Week 8 to Week 12). The primary outcome measure tested executive function at Weeks 4, 8 and 12:
Groton Maze Learning Task (GMLT) of the computerized CogState Schizophrenia
Battery (CSB). Secondary outcome measures included: CSB composite score;
Scale for Assessment of Negative Symptoms (SANS); CGI-Global Impression
(CGI-I); CGI-Severity (CGI-S); Subject Global Impression–Cognition (subjectrated scale assessing Speed of Thinking, Memory & Attention). Safety measures
included: AEs; physical exam; vital signs; serum and urine labs; ECG; Abnormal
Involuntary Movement Scale (AIMS); Columbia Suicide Severity Rating Scale
(CSSRS); and Calgary Depression Scale for Schizophrenia (CDSS). RESULTS: Blinded GMLT data showed a positive skew; all GMLT data were
log(10) transformed. GMLT results favored TC-5619 (Hochberg adjusted p =
0.054) and met predefined success criteria (1-sided p < 0.10). SANS, CGI-I and
SGI-Cog results favored TC-5619 (unadjusted p < 0.05) on a measurement date. The effect was driven primarily by tobacco users. TC-5619 was generally well
tolerated and there were no noteworthy safety findings. 72
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
The Alpha7 Neuronal Nicotinic Receptor (NNR) Modulator TC5619 had Beneficial Effects and was Generally Well tolerated in
a Phase 2 Trial in Cognitive Dysfunction in Schizophrenia (CDS)
Monday, Poster #48 (continued)
David Hosford
DISCUSSION: The concordance between objective, clinician-rated and subjectrated scores supports the promise of TC-5619 for treating the cognitive dysfunction
and negative symptoms in patients with schizophrenia. 1Hauser TA, Kucinski
A, Jordan KG, Gatto GJ, Lippiello PM, Bencherif M: TC-5619: An α7 NNR
selective agonist that demonstrates efficacy in animal models of schizophrenia. Biochem Pharmacol 1009; 78: 803-812.
73
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
EVP-6124, an Alpha-7 Nicotinic Partial Agonist, produces
Positive Effects on Cognition, Clinical Function, and Negative
Symptoms in Patients with Chronic Schizophrenia on Stable
Antipsychotic Therapy
Monday, Poster #178
Dana Hilt , Herbert Meltzer, Maria Gawry, Susan Ward, Nancy Dgetluck, Chaya
Bhuvaneswaran, Gerhard Koenig, Michael Palfreyman
EnVivo Pharmaceuticals, Inc.
BACKGROUND: Patients with schizophrenia have residual cognitive deficits,
even after treatment with available antipsychotic therapies. Currently, several
procognitive therapies are under development, including agonists of the alpha-7
nicotinic acetylcholine (ACh) receptor (N-A7A). These receptors are located in
several brain areas involved in various domains of cognition, including attention
and long term and working memory. EVP-6124 is a novel, potent, and selective
N-A7A agonist. Nine clinical studies with EVP-6124 have been completed in 561
unique subjects. Of these, 403 received EVP-6124 and 158 received placebo. In
these studies, EVP-6124 was safe and well-tolerated and exhibited linear kinetics
with a long half-life (>60 hours) suitable for once daily dosing. METHODS: A Phase 2b study in participants with schizophrenia (n=319)
receiving stable chronic atypical antipsychotic therapy has recently been
completed. The study assessed the safety and efficacy of two doses of EVP-6124
(0.3 and 1 mg once daily) versus placebo. Efficacy was evaluated by quantitative
cognitive measurements using the Overall Cognition Index (OCI) from the
CogState testing battery and Trails 2 and 4 of the Neuropsychological Test Battery
(NTB) (all subjects), the MATRICS Consensus Cognitive Battery (MCCB)
(only in a subset of subjects enrolled in the US), the Schizophrenia Cognition
Rating Scale (SCoRS) and the Positive and Negative Syndrome Scale (PANSS). Statistical results, as defined in the protocol, were considered significant at
P < 0.10 (one-sided tests). RESULTS: Patients with chronic stable schizophrenia, both smokers and nonsmokers, treated with stable doses of an antipsychotic drug other than clozapine
for at least 4 weeks before screening were treated with either placebo (n=106),
74
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
EVP-6124, an Alpha-7 Nicotinic Partial Agonist, produces
Positive Effects on Cognition, Clinical Function, and Negative
Symptoms in Patients with Chronic Schizophrenia on Stable
Antipsychotic Therapy
Monday, Poster #178 (continued)
Dana Hilt
0.3 mg/d (n=107) or 1 mg/d (n=106) of EVP-6124 for a total of 84 days. Participants
with a score of >4 on the Brief Psychiatric Rating Scale (PBRS) were excluded
from enrollment. Approximately 54 % of the subjects were enrolled in the US. Participants were 18 to 55 years of age (both inclusive) at screening. Most of the
subjects were male (68%) and white (66%). The drug was well tolerated; there
were no clinically significant findings with respect to 12 lead ECGs, vital signs,
hematology, and serum chemistry evaluations or suicidal ideation and behavior. A total of 192 treatment-emergent adverse events (TEAEs) were reported in 101
(31.9%) subjects, including 25 (23.4%) subjects in the 0.3 mg dose group, 35
(33.3%) subjects in the 1 mg dose group, and 41 (39%) subjects in the placebo
group. The most commonly reported TEAEs were headache (3.8%), nausea
(3.2%) and nosopharyngitis (2.5%). The incidence of serious adverse events
was similar among the three dosing groups; none were judged related to drug. The OCI plus Trails 2 and 4 suggested that 0.3 mg of EVP-6124, compared to
placebo, was associated with improvement in general cognitive function (P =
0.034) and that this improvement was due mainly to the beneficial effects of the
drug on visual learning, visual attention, and social cognition. The effect on the
OCI (minus Trails 2 and 4) was also significantly different among the treatment
groups (P = 0.05); further analysis indicated this was due to greater improvement
in the 0.3 mg dose group compared to placebo (P = 0.009). This positive effect
on the OCI was supported by a strong positive trend (NS) for improved cognition
on the MCCB Battery which was performed only in the US in a subset of patients
(n=166). For the 1 mg dose group, the mean change from baseline at day 84 in
the overall Composite T-score and the associated percentile change, respectively
(3.6;5.7), was higher than for the 0.3 mg dose group (3.0;2.6) and placebo group
(1.8;2.3). Significant effects in clinical function were also seen with EVP-6124
75
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
EVP-6124, an Alpha-7 Nicotinic Partial Agonist, produces
Positive Effects on Cognition, Clinical Function, and Negative
Symptoms in Patients with Chronic Schizophrenia on Stable
Antipsychotic Therapy
Monday, Poster #178 (continued)
Dana Hilt
treatment as measured by the SCoRS Interviewer Rating of clinical function. The mean change from baseline in the SCoRS Interviewer Rating, over all visits,
between the 1 mg dose group and the placebo group was significant (P = 0.065). Improvement was also seen in the negative symptoms of schizophrenia (derived
from the PANSS). For the negative subscale of the PANSS, mean decreases were
greater in the 1 mg EVP-6124 group compared to the placebo group (P = 0.028 at
end of study versus baseline; P = 0.117 over all visits). DISCUSSION: In this study, EVP-6124 treatment of subjects with stable
schizophrenia resulted in improved cognition and clinical function, and
decreased negative symptoms. In addition, EVP-6124 was well tolerated in this
population. The beneficial effects of EVP-6124 will be further investigated in
larger confirmatory studies.
76
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Gradual and Overlapping Antipsychotic Switch Strategies are
associated with less All-Cause Discontinuation in Schizophrenia:
Results from a Meta-analysis of Different Switch Strategies
Monday, Poster #173
Christoph Correll, Vishesh Agarwal, John Kane
The Zucker Hillside Hospital
BACKGROUND: Antipsychotic switching is common in schizophrenia. However, randomized, controlled trials (RCTs) of different switch strategies have
been scarce and inconsistent. METHODS: Meta-analysis of RCTs comparing different switch strategies in
schizophrenia, defined by abrupt vs. slow initiation or discontinuation of the
pre-switch or post-switch antipsychotic and by type of overlap (absent, partial
or full=“plateau”). Primary outcome was all-cause discontinuation; secondary
outcomes included specific-cause discontinuation, psychopathology and adverse
effects. Pooled relative risk (RR) [+/-95%CIs] was calculated using randomeffects model, with numbers-needed-to-treat/harm (NNT/NNH) calculations
where appropriate. RESULTS: In eight trials, 1162 patients were randomized to 2 or 3 of 5 different
switch strategies. Significantly more patients discontinued treatment with a) abrupt
vs. gradual initiation of the post-switch antipsychotic (N=2, n=467, RR:2.49
(CI:1.43,4.35), p=0.001; NNH=14 (CI:9-33); b) abrupt vs. gradual discontinuation
of the pre-switch antipsychotic (N=7, n=1109, RR:1.28 (CI:1.08,1.51), p=0.004;
NNH=16 (CI:9-100), and c) non-plateau vs. plateau-switching (N=5, n=587,
RR:1.42 (CI:1.12,1.79], p=0.003; NNH=11 (CI:7-25). Non-overlapping vs. partially/fully overlapping switching lead to significantly greater dropout when
switching from atypical to atypical antipsychotics (N=4, n=683, p=0.03), but not
when switching from typical to atypical antipsychotic (N=4, n=426, p=0.39). Global psychopathology changes did not differ across different switch strategies
(p=0.49-0.98), but few data were meta-analyzable (N=1-3, n=52-257). Abrupt vs. gradual discontinuation was associated with more insomnia (RR:2.61 (CI:1.07,
6.41; NNH 10 (CI:6-100), while abrupt vs. gradual initiation was associated with
less akathisia (RR:0.61 (CI:0.39, 0.96); NNH=-13 (CI:-7 to -100). 77
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
Gradual and Overlapping Antipsychotic Switch Strategies are
associated with less All-Cause Discontinuation in Schizophrenia:
Results from a Meta-analysis of Different Switch Strategies
Monday, Poster #173 (continued)
Christoph Correll
DISCUSSION: Non-abrupt and plateau antipsychotic switching leads to
significantly lower all-cause discontinuation, especially during atypical
antipsychotic switching.
78
ACNP 50th Annual Meeting • Final Program
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
The Hypo-Functional 7-Repeat Allele of DRD4 predicts both
Objective and Reported Fat Intake in 4- to 6- Year Old Girls
Tuesday, Poster #115
Robert Levitan, Patrícia Silveira, André Portella, James Kennedy, Hélène
Gaudreau, Caroline Davis, Meir Steiner, Claudio Soares, Stephen Matthews,
Laurette Dubé, Michael Meaney
Centre for Addiction and Mental Health, University of Toronto
BACKGROUND: The prevention and treatment of overeating and obesity
continues to be a major challenge. While the ease of availability of highly palatable
foods is a major factor in this regard, not all individuals overeat or become obese in
this environment. The current project examines individual differences in palatable
food intake at a critical time in human development. The current analysis studies
the relationship between dopamine genes and eating ehavior in children from 4
to 6 years of age. A particular focus is the hypo-functional 7-repeat (7R) allele
of the dopamine-4 receptor gene (DRD4), which associates with overeating and
obesity in several female overeater populations (e.g. Levitan et al, 2004, 2006,
2010). Recent imaging work suggests that weaker activation of the brain’s reward
circuitry may play a role in these associations (Stice et al, 2010). METHODS: The current sample consists of children taking part in a longitudinal
cohort study (Maternal Adversity, Vulnerability and Neurodevelopment) based in
Canada. The mothers of the children, oversampled based on maternal depression
and low SES, were recruited at 13-120 weeks of pregnancy, and the children
have been followed intensively since birth. Key outcome measures include: 1. A laboratory-based snack test at 48 months of age which provides an objective
measure of food preferences and overall caloric intake. 2. A food frequency
questionnaire at 48 and 72 months, based on maternal report, which assesses
naturalistic food intake and feeding behavior. 3. The childhood eating behavior
questionnaire (CEBQ; Wardle et al, 2001) which measures eating styles likely to
promote over- or under-weight. RESULTS: To date, 118 children have completed the 48 month snack test in the
lab. Of these, 47 (39.8%) carry the 7R allele, while 71 (60.2%) do not. Controlling
for total caloric intake, there is a significant gender X genotype interaction in
79
PL
ACNP 50th Annual Meeting • Final Program
PL
4:00 p.m. – 6:30 p.m.
Hot Topics – Clinical
Kona 4
The Hypo-Functional 7-Repeat Allele of DRD4 predicts both
Objective and Reported Fat Intake in 4- to 6- Year Old Girls
Tuesday, Poster #115 (continued)
Robert Levitan
predicting fat intake during the snack test (F=7.16, df = 4, 113, p=.009). Girls
who carry the 7R variant are consuming 31.2 % more fat during this snack
than are non-carriers (mean=12.2 vs. 9.3 grams respectively) while in boys, 7R
carriers are eating 19.2% less fat than are non-carriers (mean=11.3 vs. 13.5 grams
respectively). This suggests that the 7R allele increases objective fat intake in girls
but not boys by four years of age. Strikingly, when intake is measured using the
48 and 72 month food diaries, which were completed two years apart, the gender
X 7R interaction is even more robust, accounting for 21 % of the variance in fat
intake (F=11.3, df=3, 42; p=.002; N=46). Based on these diaries, girls who carry
the 7R allele are consuming 39.8% more fat in their natural environments at 48
months than are non-7R carrier girls (820.5 vs. 587.0 calories/day respectively). Importantly, this difference remains robust at 72 months of age (881.5 calories as
fat in 7R carriers vs. 609.2 calories as fat in non-carriers, a 44.7 % difference). In boys, 7R carriers are consuming 40.1 % less calories from fat than are noncarriers at 48 months, and 25.4 less calories from fat at 72 months based on these
diaries. The consistency of these data across both laboratory based measures and
food diaries, and the consistency of the food diary data over a two year time
span, adds greatly to these findings. These findings are also highly consistent with
earlier work in female adult overeater populations (see above). DISCUSSION: These converging results show a highly robust association
between the hypo-functional 7R allele of DRD4 and fat intake in girls, but not
boys. The consistency of these findings across different outcome measures and
across a two year age span adds greatly to their validity and potential impact. Pending replication and detailed anthropomorphic outcomes later in childhood,
these results may identify a significant subgroup of young girls at higher risk
for childhood obesity and/or eating disorders. If so, this could inform novel
prevention strategies implemented as early as birth in this subgroup.
80
ACNP 50th Annual Meeting • Final Program
Notes
81
ACNP 50th Annual Meeting • Final Program
Notes
82
Monday At A Glance
6:30 am – 8:00 am
SOBP Editorial Board Meeting
Queen’s 4
7:00 am – 9:30 am
Continental Breakfast
8:00 am – 11:30 am
President’s Plenary: Brave New World
for Brain Therapeutics
11:30 am – 1:30 pm
Buffet Lunch
12:00 pm – 1:30 pm
Women’s Luncheon
12:00 pm – 1:30 pm
Teaching Neuropsychopharmacology
12:00 pm – 1:30 pm
ASCP Board of Directors Meeting
1:30 pm – 3:00 pm
Distinguished Lecture
Grand Promenade/Lagoon Lanai
Monarchy Ballroom
Grand Promenade
Water’s Edge Ballroom
Queen’s 4
Monarchy Ballroom
Panel Sessions
3:00 pm – 5:30 pm
The Noradrenergic System as a Therapeutic
Target for Drug Dependence
Monarchy
3:00 pm – 5:30 pm
Striving for the Correct Diagnosis for Mental
Health Disorders
Kona 4
3:00 pm – 5:30 pm
Genetic and Molecular Mechanisms of
Normal Cognitive Aging
3:00 pm – 5:30 pm
Memory Erasure: Mechanisms and Potential
Utility in Psychiatry
Kona 1-3
PA-Panel MP-Mini Panel PL-Plenary SG-Study Group
Kona 5
Monday
Kohala 4
Monday At A Glance (continued)
Panel Sessions
3:00 pm – 5:30 pm
Enteric Hormone Modulation of Cerebral
Neurotransmission and Eating Behaviors
In Obesity
3:00 pm – 5:30 pm
NMDA Receptor Complexes: A Point of
Convergence for Schizophrenia Candidate
Pathways
3:00 pm – 5:30 pm
Kohala 3
Queen’s 5-6
Kohala 4
Adolescent Brains: The Constancy of Change
5:30 pm – 7:30 pm
Poster Session I with Reception
Kohala 1-2,
King’s & Grand Promenade
Monday
Study Groups
7:30 pm – 9:00 pm
Assessing Brain Development Trajectories
from infancy to Adulthood
7:30 pm – 9:00 pm
Ethical, Legal, and Social Challenges
In Research on Psychiatric Genetics
7:30 pm – 9:00 pm
PTSD Bookmarkers
7:30 pm – 9:00 pm
Can Vulnerability Markers identify Informative
Neurodevelopmental Abnormalities across the
Spectrum of Early Psychosis?
7:30 pm – 9:00 pm
Crisis in Psychiatric Drug Discovery:
Solutions from Academia, Government
and the Advocacy Community
7:30 pm – 9:00 pm
Utilizing the NIH’s CTSA Network to
Advance Neuropsychopharmacology
Kohala 4
7:30 pm – 9:00 pm
The Alcohol Clinical Trials Initiative
(ACTIVE): Progress Report and Feedback
Kohala 3
7:30 pm – 9:00 pm
Four Rodent Models of Psychosis:
(Not) Lost in Translation
Kona 1-3
Kona 4
Monarchy
Queen’s 5-6
Kona 5
Queen’s 4
PA-Panel MP-Mini Panel PL-Plenary SG-Study Group
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 11:30 a.m.
President’s Plenary
Monarchy Ballroom
President’s Plenary
Welcoming Remarks and Moment of Silence
Eric J. Nestler
President
Presentation of Honorific Awards
David Rubinow
Chair, Honorific Awards Committee
Brave New World for Brain Therapeutics
8:30 a.m.
Rapidly Acting Chemical Antidepressants
Dennis Charney
9:15 a.m.
Rethinking Depression and its Treatment: Insights from Studies
of Deep Brain Stimulation
Helen Mayberg
10:00 a.m.
Viral-Mediated Gene Therapy for Neuropsychiatric Disorders
Michael Kaplitt
10:45 a.m.
Using Human Stem Cells to Understand and Treat Diseases of the Brain
Lawrence Goldstein
83
PL
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 11:30 a.m.
President’s Plenary
Monarchy Ballroom
Rapidly Acting Chemical Antidepressants
PL Dennis Charney
Mount Sinai School of Medicine
This presentation will review the clinical data indicating that ketamine and
scopolamine have rapid (within hours) antidepressant effects in patients with
treatment resistant depression. Putative mechanisms underlying these clinical
effects will be discussed. A theoretical synthesis will be developed as a guide for
the discovery of a new generation of antidepressant drugs.
Dr. Charney’s distinguished career as a researcher and educator began in 1981
at Yale University School of Medicine. While at Yale, Dr. Charney chaired the
National Institute of Mental Health (NIMH) Board of Scientific Counselors,
which advises the institute’s director on intramural research programs. In 2000
NIMH recruited Dr. Charney to lead the Mood and Anxiety Disorder Research
Program — one of the largest programs of its kind in the world — and the
Experimental Therapeutics and Pathophysiology Branch. That same year Dr. Charney was elected to the Institute of Medicine of the National Academy of
Sciences. 84
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 11:30 a.m.
President’s Plenary
Monarchy Ballroom
Rethinking Depression and its Treatment: Insights from Studies
of Deep Brain Stimulation
Helen Mayberg
Emory University School of Medicine
Critical to the development of deep brain stimulation (DBS) as a novel therapy for
patients with treatment resistant depression has been the further characterization
of brain systems mediating normal and abnormal mood states as well as those
mediating successful and unsuccessful response to various antidepressant
interventions using functional neuroimaging. Building on converging evidence
implicating the subcallosal cingulate as a critical node within an imaging-based,
putative depression network model, the subcallosal cingulate white matter was
targeted for initial proof-of-principle testing of DBS in patients with treatment
resistant major depression, adopting neuromodulation techniques routinely used
to treat Parkinson’s disease and other movement disorders. The theoretical and
data-driven foundation for this new treatment strategy as well as results from
ongoing experimental studies will be presented. Dr. Mayberg is Professor of Neurology and Psychiatry and the Dorothy C. Fuqua
Chair in Psychiatric Neuroimaging and Therapeutics at the Emory University
School of Medicine where she has an active research program in the neuroimaging
of depression. Dr. Mayberg is a neurologist, trained at Columbia’s Neurological
Institute in New York, with fellowship training in nuclear medicine at Johns
Hopkins. She received a BA in Psychobiology from UCLA and an M.D. from
University of Southern California. She is active in the Society for Neuroscience,
Society of Biological Psychiatry, the American Neurological Association, and the
Organization for Human Brain Mapping, and is a member of the Dana Alliance,
the NARSAD Scientific Advisory Board and the Institute of Medicine. 85
PL
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 11:30 a.m.
President’s Plenary
Monarchy Ballroom
Viral-Mediated Gene Therapy for Neuropsychiatric Disorders
PL Michael Kaplitt
Weill Cornell Medical College
Gene therapy has held increasing promise in recent years as a novel approach
to neurological disorders. An important factor in the evolution of clinical gene
therapy for neurological diseases has been identification of important focal targets
within the brain which may widely influence dysfunctional circuitry essential to
the pathophysiology of specific maladies. Animal models and sophisticated human
functional brain imaging have in recent years led to a greater understanding of
circuits and focal brain regions which are dysfunctional in a variety of psychiatric
disorders. With increasing application of gene therapy to the human brain, and
our recent demonstration of the first successful randomized, double-blind, sham
surgery controlled clinical trial of gene therapy for a neurological disorder,
the possibility of clinical translation of this approach to psychiatric diseases is
now more realistic than previously contemplated. A variety of genes have been
altered with gene therapy agents over the years in animal models of psychiatric
disorders. Here we will focus on one gene, p11, as an example of how gene
therapy may evolve for psychiatric diseases. This gene was originally identified
by Paul Greengard and colleagues as a potential binding partner modulating the
activity of specific serotonin receptor subtypes. Mice lacking p11 (p11 KO mice)
manifest depression-like behaviors. Utilizing adeno associated virus (AAV)mediated gene therapy, we demonstrated that focal blockade of p11 expression
exclusively in the nucleus accumbens (NAc) of otherwise normal adult mice
resulted in depression-like behaviors similar to p11 KO mice. Focal restoration
of p11 expression only in the NAc of p11 KO mice using gene therapy then
reversed the depression-like behaviors, suggesting that this may be a potential
therapy for human depression if p11 is relevant to this disorder. To this end, in
collaboration with Carol Tamminga and Per Svenningson, we observed that NAc
p11 expression was in fact significantly lower in post-mortem brain specimens
from depressed human patients compared with matched controls, suggesting
that p11 deficiency in the NAc is a characteristic of human depression. We are
currently collaborating with investigators at the National Institutes of Mental
Health to examine NAc p11 gene therapy in non-human primates, to generate
initial data which may eventually support a human clinical trial. The other key
86
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 11:30 a.m.
President’s Plenary
Monarchy Ballroom
Viral-Mediated Gene Therapy for Neuropsychiatric Disorders
Michael Kaplitt (continued)
PL
region currently of interest in depression, particularly for surgical interventions,
is the subgenual cingulate cortex (BA 25). Human functional imaging suggests
that increased activity of BA25 is associated with major depression, as opposed
to reduced activity of the NAc. Interestingly, we have recently demonstrated
that focal knockdown of p11 using gene transfer in the rodent equivalent of
BA25 (infralimbic cortex) also has a different effect than in the NAc, with no
effect on baseline depression but a potentiation of the action of anti-depressant
medications.
Finally, we have collaborated with Scott Russo and Eric Nestler to demonstrate
that p11 KO mice have increased susceptibility to cocaine using a placepreference model (CPP). Again p11 gene therapy in the NAc reduced CPP in
both p11 KO mice and in normal mice, suggesting that this may be an unusual
treatment which could block addictive behaviors while improve depressionlike behaviors as well. Ongoing development of psychosurgery and functional
imaging, along with greater experience with gene therapy in the human brain,
should help facilitate translation of this technique into clinical application for
refractory psychiatric diseases in the near future.
Michael G. Kaplitt, M.D. Ph.D. is a tenured Associate Professor, Vice-Chair
for Research, Residency Program Director and Director of the Laboratory of
Molecular Neurosurgery I the Dept. of Neurological Surgery, Weill Cornell
Medical College and New York-Presbyterian Hospital. Dr. Kaplitt graduated
Magna Cum Laude from Princeton University in 1987 with a degree in Molecular
Biology and Russian Studies. He received his Ph.D. in Molecular Neurobiology
in 1993 from The Rockefeller University and his M.D. in 1995 from Cornell
Medical College. Following a Neurosurgery residency and chief residency at
Cornell, he completed a fellowship in Stereotactic and Functional Neurosurgery
with Dr. Andres Lozano at the University of Toronto. His clinical activities focus
upon using minimally invasive, stereotactic techniques for the treatment of
functional neurological disorders such as Parkinson’s disease, essential tremor,
epilepsy and pain.
87
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 11:30 a.m.
President’s Plenary
Monarchy Ballroom
PL
Using Human Stem Cells to Understand and Treat Diseases of
the Brain
Lawrence Goldstein
University of California, San Diego, School of Medicine
Stem cell technologies bring new and powerful opportunities to understand
and treat previously untreatable brain disorders. In my talk, I will discuss how
we are trying to use human embryonic and other types of stem cells to develop
new methods to understand, and eventually treat Lou Gehrig’s Disease (ALS),
Alzheimer’s Disease, and other related diseases of the brain by generating new
approaches for drug discovery or therapy. For example, Alzheimer’s Disease is
a severe, progressive, and incurable disease characterized by memory loss and
dementia. Although common pathological features of the disorder are known,
their relationship to the development of the disease remains problematic. Rare
hereditary forms of Alzheimer’s Disease identify genetic changes that can
cause disease. In one approach, we are working to generate human neurons
from human stem cells that carry the genetic changes that can cause hereditary
Alzheimer’s Disease to try and test several ideas about what causes this terrible
disease. However, the relationship of hereditary forms of Alzheimer’s Disease
to the common “sporadic” form of the disease remains unclear. Human stem
cell technology letting us begin to probe the relationship of the two forms of the
disease. A second example of an approach we are taking is from ALS, which is
characterized by progressive loss of the cells that control movement, resulting in
paralysis and ultimately death. We have been working to identify which cells are
at fault in this disease, and have begun to develop methods that might allow stem
cells to someday be used for therapeutic intervention.
Dr. Goldstein is Distinguished Professor of Cellular and Molecular Medicine at
the University of California, San Diego, School of Medicine and Director of the
UC San Diego Stem Cell Program. He is also an Investigator with the Howard
Hughes Medical Institute. He has been with the UCSD School of Medicine since
1993. A UCSD alumnus, he received his B.A. degree in biology and genetics
from UCSD in 1976 and his Ph.D. degree in genetics from the University of
Washington, Seattle in 1980. Dr. Goldstein did postdoctoral research at the
88
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 11:30 a.m.
President’s Plenary
Monarchy Ballroom
Using Human Stem Cells to Understand and Treat Diseases of
the Brain
Lawrence Goldstein (continued)
University of Colorado at Boulder from 1980 to 1983 and the Massachusetts
Institute of Technology in 1983-1984. He was assistant, associate, and full
professor at Harvard University in the Department of Cellular and Developmental
Biology from 1984 to 1993, when he moved to UCSD and HHMI. His awards
include a Senior Scholar Award from the Ellison Medical Foundation, an
American Cancer Society Faculty Research Award, the Loeb Chair in Natural
Sciences when he was at Harvard University, election to the American Academy
of Arts and Sciences, and the 2009 Public Service Award from the American
Society for Cell Biology.
89
PL
ACNP 50th Annual Meeting • Final Program
12:00 p.m. – 1:30 p.m.
Teaching Neuropsychopharmacology
Kohala 4
Teaching Neuropsychopharmacology
PL
A Method for Innovative Thinking as Applied to
Neuropsychopharmacology
Chair: Mark H. Rapaport
Presented by: Roberta Ness
Innovation, defined as creativity with a purpose, is widely considered to be the
engine of scientific progress. Although technical inventions in the last generation
have transformed everyday life, novel solutions to some of the greatest concerns
in neuropsychopharmacology (addiction, schizophrenia, intractable depression,
to name a few) remain less forthcoming. Indeed, American science in general was
deemed to be lagging in the “Principal ingredients of scientific innovation and
competitiveness – knowledge capital, human capital, and a creative ecosystem,”
when recently assessed by a blue ribbon Committee of the National Academies
of Science. That Committee, entitling its report, Rising Above the Gathering
Storm, opinioned that America is losing its international hegemony. Our normal mental tendency is to think in ways that are habitual and formulaic. Daring new solutions emerge only when we explode our ordinary patterns of
thought. You are familiar with the “war on cancer”. How would you react to the
alternative suggestion of “cancer as neighbor”? Crazy? Maybe not. In fact, this
alternative idea might lead to more rational policies toward cancer screening and
more humane approaches toward cancer treatment. Dr. Roberta Ness, Dean of
the University of Texas School of Public Health, describes Innovation Generation
a method she has developed for enhancing innovative thinking. Consisting of
a readily applicable toolbox, this method provides a means for enlarging the
innovative space within neuropsychopharmacology.
90
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Distinguished Lecture
Monarchy Ballroom
Distinguished Lecture
Insights into Circadian Clock and Sleep from Human Genetics
Chair: Eric J. Nestler
Presented by:
Louis J. Ptáček
91
PL
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Distinguished Lecture
Monarchy Ballroom
Insights into Circadian Clock and Sleep from Human Genetics
PL Louis J. Ptáček
University of California San Francisco
On planet earth, organisms have evolved mechanisms to synchronize metabolic
and physiological functions and behavior with the approximately twentyfour hour light/dark cycle of a day. These rhythms touch on our cognitive
performance, metabolism, immune and cardiovascular function, sleep/wake, and
other behaviors. Advanced sleep phase syndrome (ASPS) is a common alteration
in the biological sleep- and wake-time preferences in humans and is common in
aging individuals. As we get older, we tend to fall asleep and wake earlier then
we did in youth. A dramatic autosomal dominant form of ASPS (Familial ASPS,
FASPS)—distinct from the ASPS of aging—was described by our group. We
now know that FASPS affects 0.3% of the general population. Recognition of
this phenotype has allowed genetic studies to identify and characterize causative
mutations and to generate animal FASPS models. Disruption of the circadian
clock has also been linked to numerous ailments including cancer, cardiovascular
diseases, depression, and learning disorders. Thus, the investigation of the human
circadian and sleep mechanism promises enormous benefits to our understanding
of human health and disease. Studies in one intriguing family with FASPS led to
identification of a mutation in casein kinase Iδ (CKIδ). Remarkably, the mutation
in this family segregates not only with FASPS, but also with Asthma and Migraine
with Aura suggesting the possibility that this mutation causes these other
phenotypes too. We have the strong anecdotal impression that other behavioral
phenotypes are more common in FASPS individuals although study of such
phenotypes in humans is extremely difficult. The mouse models of human FASPS
mutations that we’ve generated are a resource for beginning to test hypotheses
suggested by observations in humans. More recently, we have also recognized
and begun collecting families with Delayed Sleep Phase Syndrome (FDSPS) and
Familial Natural Short Sleep (FNSS). Identifying genes causing phase and sleep
phenotypes is beginning to shed light on the mysterious processes regulating
human sleep and this will have profound consequences for human health and
understanding and treatment of human diseases. Modeling human circadian and
sleep mutations in model systems is enabling parallel studies in humans carrying
92
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Distinguished Lecture
Monarchy Ballroom
Insights into Circadian Clock and Sleep from Human Genetics
Louis J. Ptáček (continued)
PL
such mutations with rodent and fly models to explore similarities and differences
between circadian clocks and sleep in different species. Ultimately such work
may lead to understanding of connections between human clock and various
diseases and behaviors and identification of novel therapeutic targets.
Dr. Ptácek’s laboratory focuses on genetic diseases of muscle, heart and brain
and hereditary variation of human sleep behavior. His group has cloned genes
causing many disease and behavioral phenotypes in humans. In addition, he and
his collaborators probe the biology underlying normal function of the encoded
proteins in the nervous system and pathophysiology of the mutant proteins in
human neurological diseases. To this end, they use cellular electrophysiology,
biochemistry, cell biology and animal modeling. He and Chris Jones identified
and characterized the first human families with a Mendelian circadian rhythm
variant. These individuals have an extreme “morning lark” phenotype and they
called this variant familial advanced sleep phase syndrome (FASPS). His group
and the group of Ying-Hui Fu have gone from the clinical and physiologic
characterization of this phenotype to the mapping and cloning of the causative
genes, biochemical study of the encoded proteins, and generation of animal
models. In addition, they have collected a large group of families with FASPS
and other circadian phenotypes, and these are being studied to identify additional
human circadian rhythm genes. They have also identified families with genetic
variants of sleep and begun to clone genes regulating the sleep hemostat. This
work represents a move from purely ‘disease genetics’ to the genetics of human
behavior. Insights into the proteins causing these disorders and traits will
ultimately lead to new insights into the normal function of the human nervous
system and mechanisms of disease. Ultimately, such insights will lead to new
therapies for treating patients with various neurological and other disorders.
93
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Monarchy
The Noradrenergic System as a
Therapeutic Target for Drug Dependence
Chair: Bernard Le Foll
Co-Chair: David Weinshenker
3:00 p.m.
PA
3:30 p.m.
4:00 p.m.
4:30 p.m.
Functional Neuroanatomy of Norepinephrine-Dopamine
Interactions within the Mesocorticcolimbic Reward System
David Weinshenker
Noradrenergic Alpha-1 Receptors as a Novel Target for the
Treatment of Nicotine Addiction
Bernard Le Foll
Preclinical Evidence for a Role of Noradrenergic Systems in
Addiction
George Koob
Results of a Pilot trial of the Alpha-1 Adrenergic Antagonist,
Prazosin for Alcohol Dependence
Tracy Simpson
5:00 p.m.
Discussant: Charles O’Brien
94
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Kona 4
Striving for the Correct Diagnosis of Mental Health Disorders
Chair: Alan Schatzberg
Co-Chair: Stephen Koslow
3:00 p.m.
What Will the New DSM-5 Provide for Us?
David Kupfer
3:30 p.m.
New Approaches to Psychiatric Diagnosis: The MIMH Research
Domain Criteria Project
Bruce Cuthbert
4:00 p.m.
4:30 p.m.
Using Biological and Cognitive Measures to Discriminate among
Depressive Subtypes
Alan Schatzberg
International Study to Predict Optimized Treatment for
Depression (iSPOT-D), A Randomized Clinical Trial: Rationale
and Protocol
Leanne Williams
5:00 p.m.
Discussant: John Rush
95
PA
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Kona 1-3
Genetic and Molecular Mechanisms of Normal Cognitive Aging
Chair: Venkata Mattay
Co-Chair: Terry Goldberg
PA
3:00 p.m. Aging-Associated Changes in the Human Brain Transcriptome
Vahram Haroutunian
3:30 p.m.
Protection against Cognitive Decline and Dementia by Longevity Genes
Yousin Suh
4:00 p.m.
Neuroimaging Genetic Influence in Normal Cognitive Aging:
The Role of Memory and Cognition Related Genes
Venkata Mattay
4:30 p.m.
Novel APOE4 Findings in Cognitively Healthy and
Compromised Aging Individuals
Terry Goldberg
5:00 p.m.
Discussant: David Michelson
96
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Kona 5
Memory Erasure: Mechanisms and Potential Utility in
Psychiatry
Chair: William Carlezon
Co-Chair: Michael Davis
3:00 p.m.
Memory as a New Therapeutic Target
Karim Nader
3:30 p.m.
Temporary, but not Permanent, Disruption of Fear Potentiated
Startle following PKMβ Inhibition of Amygdala
Michael Davis
4:00 p.m.
Disrupting Fear Memories: Retrieval, Reconsolidation and the
Passage of Time
Cristina Alberini
4:30 p.m.
Selectively Erasing a Fear Memory in Mice
Sheena Josselyn
5:00 p.m. Discussant: Roger Pitman
97
PA
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Kohala 3
Enteric Hormone Modulation of Cerebral Neurotransmission
and Eating Behaviors in Obesity
Chair: Robert Kessler
PA
3:00 p.m. Enteric Hormone Modulation of Cerebral Neurotransmission and Eating Behaviors in Obesity
Dianne Lattemann
3:30 p.m.
Impaired Striatal Akt Signaling disrupts Dopamine Homeostasis and increases Feeding
Aurelio Galli
4:00 p.m. Reward Mechanisms in Feeding and Addiction: Paradoxical Roles for Hypocretin (Orexin) Transmission
Jonathan Hollander
4:30 p.m.
PET Studies of Dopaminergic Neurotransmission in Obesity
Robert Kessler
5:00 p.m.
Discussant: Nicole Avena
98
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Queen’s 5-6
NMDA Receptor Complexes: A Point of Convergence
for Schizophrenia Candidate Pathways
Chair: Raquel Gur
3:00 p.m.
3:30 p.m.
4:00 p.m.
4:30 p.m.
Neuregulin1-ErbB4 Signaling Suppresses the Src Upregulation
of NMDA Receptors
Michael Salter
Rac 1-PAK Cascade: A Promising Drug Target for Synaptic
Deterioration in Mental Illness
Akira Sawa
Dysbindin-1 Reductions in Schizophrenia may affect Cognition
via Multiple Effects on NMDA Receptor Biology, including
Induction of Arc Expression
Konrad Talbot
N-methyl D-aspartate Receptor Complexes in Brains of
Schizophrenia Patients
Chang-Gyu Hahn
5:00 p.m.
Discussant: Daniel Javitt
99
PA
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Kohala 4
Adolescent Brains: The Constancy of Change
Chair: Ruben Gur
PA
3:00 p.m.
Clinical Studies during Adolescence: Autism and Bipolar Disorder
John Sweeney
3:30 p.m.
Anatomic MRI of the Developing Brain: Ages 3 to 30
Jay Giedd
4:00 p.m.
The Imagen Gene X Neuroimaging Study on Reinforcement-
Related Behavior in Adolescents: GWAS and Epigenetic Results
Gunter Schumann
4:30 p.m.
Sex Differences in Normative Developmental Trajectories of Brain Behavior
Ruben Gur
5:00 p.m.
Discussant: Daniel Pine
100
ACNP 50th Annual Meeting • Final Program
7:30 p.m. – 9:00 p.m.
Kona 1-3
Assessing Brain Developmental Trajectories from Infancy to
Adulthood
Chair: James Swanson
Moderator: Thomas Insel
Participants:
John Gilmore
Claudia Buss
Damien Fair
Jay Giedd
Xavier Castellanos
Raquel Gur
Linda Chang
Anders Dale
SG
7:30 p.m. – 9:00 p.m.
Kona 4
Ethical, Legal, and Social Challenges in Research
on Psychiatric Genetics
Chair: Paul Appelbaum
Moderator: Hank Greely
Participants:
Jennifer McCormick
Paul Appelbaum
Barbara Koenig
Laura Roberts
101
ACNP 50th Annual Meeting • Final Program
7:30 p.m. – 9:00 p.m.
Monarchy
PTSD Biomarkers Study Group
Chair: Alexander Neumeister
Co-Chair: Victoria Risbrough
Participants:
Charles Nemeroff
Thomas Neylan
Charles Marmar
Dewleen Baker
Scott Orr
Murray Stein
SG
7:30 p.m. – 9:00 p.m.
Queen’s 5-6
Can Vulnerability Markers identify Informative
Neurodevelopmental Abnormalities across the Spectrum of
Early Psychosis?
Chair: Kristin Cadenhead
Co-Chair: Diana Perkins
Moderator: Matcheri Keshavan
Participants:
Jean Addington
Barbara Cornblatt
Elaine Walker
Daniel Mathalon
Diana Perkins
Kristin Cadenhead
Larry Seidman
Tyrone Cannon
102
ACNP 50th Annual Meeting • Final Program
7:30 p.m. – 9:00 p.m.
Kona 5
Crisis in Psychiatric Drug Discovery: Solutions from Academia,
Government and the Advocacy Community
Chair: Mark Rasenick
Co-Chair: William Potter
Participants:
John Greden
Anand Pandya
Beth Hoffman
Jeffrey Nye
Patrick Kennedy
SG
7:30 p.m. – 9:00 p.m.
Kohala 4
Utilizing the NIH’s CTSA Network to Advance
Neuropsychopharmacology Research
Chair: Anantha Shekhar
Co-Chair: William Potter
Participants:
Kathleen Brady
John March
Srijan Sen
Linda Brady
Anantha Shekhar
103
ACNP 50th Annual Meeting • Final Program
7:30 p.m. – 9:00 p.m.
Kohala 3
The Alcohol Clinical Trial Initiatives (ACTIVE):
Progress Report and Feedback
Chair: Raymond Anton
Co-Chair: Henry Kranzler
Moderator: Henry Kranzler
Participants:
Raymond Anton
Henry Kranzler
Daniel Falk
Roger Meyer
Stephanie O’Malley
Bernard Silverman
SG
7:30 p.m. – 9:00 p.m.
Queen’s 4
Four Rodent Models of Psychosis: (Not) Lost in Translation
Chair: Herbert Meltzer
Participants:
Anthony Grace
Akira Sawa
Maria Karayiorgou
Bryan Roth
104
ACNP 50th Annual Meeting • Final Program
Notes
105
ACNP 50th Annual Meeting • Final Program
Notes
106
Tuesday Morning At A Glance
7:00 am – 8:30 am
Liaison Committee Meeting
Water’s Edge Ballroom
7:00 am – 8:30 am
American Journal of Psychiatry Board Meeting
7:00 am – 8:30 am
Membership Advisory Task Force Meeting
7:00 am – 8:30 am
NPPR Editors Meeting, Volumes 6&7
7:30 am – 8:30 am
ACNP Leadership & Institute Directors
Imari
Queen’s 4
Water’s Edge Boardroom
Donatonio’s
Mini Panel Sessions
8:30 am – 9:45 am
9:45 am – 11:00 am
Medication Discovery for Addiction: Translating
the Dopamine D3 Receptor Hypothesis
Monarchy
Vaccines, Viral Vectors, and Cocaine Addiction:
Neutralizing Cocaine before it gets to the Brain
Monarchy
Panel Sessions
8:30 am – 11:00 am
8:30 am – 11:00 am
8:30 am – 11:00 am
8:30 am – 11:00 am
Cortical Dopamine in Schizophrenia: Quantifying
Levels, Understanding Function
Kona 5
Synaptic Plasticity: From Adaptive Molecular
Mechanisms to Dysregulation in Psychiatric Disorders
Kohala 4
Neuroimaging Genomics: Discovering a Signal
in the Complexity of Genes, Brain and Behavior
Kona 1-3
Feast or Famine: Is Disordered Eating Related
to Disordered Reward?
Emerging Methods to Examine Fear Regulation
8:30 am – 11:00 am
Circadian Rhythms, Sleep Deprivation and
Mood Disorders
11:00 am – 12:30 pm
ACNP Corporate Liaison Luncheon
(by invitation only)
11:30 am – 1:30 pm
Data Blitz Session
11:30 am – 1:30 pm
Travel Awardee Research Presentations
11:30 am – 1:30 pm
Faculty Research Fellowship Presentations
Queen’s 5-6
Kohala 3
Donatonio’s
Kona 4
Kona 1-3
Kohala 4
Queen’s 5-6
PA-Panel MP-Mini Panel PL-Plenary SG-Study Group
Kohala 3
Tuesday
8:30 am – 11:00 am
Kona 4
Tuesday Afternoon At A Glance
1:30 pm – 3:00 pm
Issues in Ethics: The Perils and Pitfalls of
Biomedical Research: Historical and
Contemporary Perspectives on the Ethics
of Research
Kona 5
3:00 pm – 5:30 pm
Epigenetic Modifications in Development,
Aging and Mental Illness
Kona 4
3:00 pm – 5:30 pm
Molecular Mechanisms Informing PTSD
Risk, Treatment and Prophylaxis
Kona 5
Tuesday
Panel Sessions
3:00 pm – 5:30 pm
New Directions in Understanding the
Neurocircuitry of Choice, Value, and
Decision-Making
Kona 1-3
3:00 pm – 5:30 pm
A Convergence in Autism and Schizophrenia
Genetics: The Conundrum of Shared Risks and
Divergent Outcomes
Kohala 3
3:00 pm – 5:30 pm
Neurodevelopmental Pathology of Cortical
Interneurons in Schizophrenia: Is it the Journey
or the Destination that Matters?
Kohala 4
3:00 pm – 5:30 pm
Will We Have New Drugs or Not? Addressing
The Crisis in Neuropsychiatric Drug Discovery
Monarchy
3:00 pm – 5:30 pm
Toward a Neuroimmune-Medicated Subtype
of Autism Spectrum Disorders
5:30 pm – 7:30 pm
Poster Sessions II with Reception
Queen’s 5-6
Kohala 1-2
King’s & Grand Promenade
6:00 pm – 11:00 pm
ACNP Committee Chairs Waiting Room
Water’s Edge Ballroom
6:00 pm – 11:00 pm
ACNP Council Meeting
7:30 pm – 9:00 pm
Neuropsychopharmacology Editorial Board Water’s Edge Ballroom
Queen’s 4
PA-Panel MP-Mini Panel PL-Plenary SG-Study Group
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 9:45 a.m.
Mini Panel Session
Monarchy
Medication Discovery for Addiction: Translating
the Dopamine D3 Receptor Hypothesis
Chair: Amy Newman
8:30 a.m. Translational Approach to Dopamine D3 Receptor: From Mechanism of Action to Clinical Studies
Emilio Merlo Pich
8:55 a.m.
Buspirone: New Look at an Old Drug
Phil Skolnick
9:20 a.m.
Monkey Models of Stimulant Abuse: Effects of Dopamine D3-Selective Agonists, Partial Agonists and Buspirone
Michael Nadar
MP
107
ACNP 50th Annual Meeting • Final Program
9:45 a.m. – 11:00 a.m.
Mini Panel Session
Monarchy
Vaccines, Viral Vectors, and Cocaine Addiction:
Neutralizing Cocaine Before it gets to the Brain
Chair: Marilyn Carroll
8:30 a.m. Cocaine Vaccine: Promises vs. Reality
Thomas Kosten
8:55 a.m.
Steps Toward Cocaine Hydrolase Gene Therapy
Stephen Brimijoin
9:20 a.m.
Long Term Reduction of Cocaine-Seeking Behavior in Rats Treated with Cocaine Hydrolase Delivered by a Viral Vector
Marilyn Carroll
MP
108
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kona 5
Cortical Dopamine in Schizophrenia:
Quantifying Leads, Understanding Function
Chair: Anissa Abi-Dargham
Co-Chair: Holly Moore
8:30 a.m. Validation of [C-11]FLB 457 as a Tool to Measure Cortical Dopamine Release
Raj Narendran
9:00 a.m.
Decreased Cortical Dopamine Release in Schizophrenia: Evidence from in Vivo Imaging
Anissa Abi-Dargham
9:30 a.m.
Dysregulation of the Norepinephrine Transporter sustains Cortical Hypodopaminergia and Schizophrenia-Like Behaviors in Neuronal Rictor Null Mice
Aurelio Galli
10:00 a.m.
Developmental Disruption of Prefrontal Cortex Intereurons by Altered Dopamine Transmission during Adolescence
Kuei Tseng
10:30 a.m.
Discussant: Holly Moore
109
PA
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kohala 4
Synaptic Plasticity: From Adaptive Molecular Mechanisms to
Dysregulation in Psychiatric Disorders
Chair: R. Suzanne Zukin
Co-Chair: Carol Tamminga
PA
8:30 a.m. Mechanisms of LTP and LTD: Recent Advances
Robert Malenka
9:00 a.m.
Regulation of AMPA Receptor Function during Fear Memory and Erasure
Richard Huganir
9:30 a.m.
The Gene Silencing Factor REST and Maternal Deprivation Epigenetically Regulate the Switch in NMDA Receptor Phenotype during Brain Development
Suzanne Zukin
10:00 a.m.
Alterations in Hippocampal Learning and Memory Mechanisms in Schizophrenia
Carol Tamminga
10:30 a.m.
Discussant: Eric Nestler
110
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kona 1-3
Neuroimaging Genomics: Discovering a Signal in the
Complexity of Genes, Brain and Behavior
Chair: Raquel Gur
8:30 a.m. Genome-Wide Association Implicates FGF14 in Amygdala Volume and Fear Processing
David Glahn
9:00 a.m.
Imaging Genetics Validation of Molecular Interactions in Psychiatric Risk Pathways
Daniel Weinberger
9:30 a.m.
Epistasis and Epigenetic DNA Methylation are Involved in Risk for Schizophrenia Phenotypes
Alessandro Bertolino
10:00 a.m.
A Developmental Study Integrating Neuroimaging and Genomics
Raquel Gur
10:30 a.m.
Discussant: John Blangero
111
PA
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kona 4
Feast or Famine: Is Disordered Eating
Related to Disordered Reward?
Chair: Kathryn Cunningham
Co-Chair: Ralph DiLeone
8:30 a.m. Analysis of Brain Reward Circuits following Food-Restriction Reveals Common Glucocorticoid-Initiated Gene Expression Changes
Ralph DiLeone
9:00 a.m.
Nucleus Accumbens Serotonin (5-HT) 5-HT2C Receptor is Involved in Sensitivity to Obesogenic Food
Noelle Anastasio
PA 9:30 a.m.
Imaging of Brain Dopamine in Binge Eating Disorder
Gene-Jack Wang
10:00 a.m.
Individual Differences in Cue Reactivity: Food and Drugs
Harriet de Wit
10:30 a.m.
Discussant: Yavin Shaham
112
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Queens 5-6
Emerging Methods to Examine Fear Regulation
Chair: Kerry Ressler
8:30 a.m. Development and Expression of Fear Memories during Adolescence
Francis Lee
9:00 a.m.
Epigenetic Regulation of Gene Expression to Examine Mechanisms of Amygdala Plasticity and Fear Learning in Vivo and in Amygdala Primary Cultures
Kerry Ressler
9:30 a.m.
Optogenetic Investigation of Circuit Mechanisms of Anxiety and Anxiolysis
Karl Deisseroth
10:00 a.m.
Using Multi-Electrode Recording in Freely Moving Rats to Probe the Regulation of Fear Memory Formation and Extinction
Donald Rainnie
10:30 a.m.
Discussant: William Carlezon
113
PA
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kohala 3
Circadian Rhythms, Sleep Deprivation
and Mood Disorders
Chair: Ted Abel
Co-Chair: Colleen McClung
PA
8:30 a.m. Rhythms and Blues: How Circadian Genes Regulate Mood
Colleen McClung
9:00 a.m.
Circadian Gene and Sleep Modulation of Reward Circuitry: Implications for Vulnerability to Bipolar Disorder
Mary Phillips
9:30 a.m.
Neurobiological Consequences of Disrupted Sleep: Implications for Depression
Peter Meerlo
10:00 a.m.
Glutamatergic Neurotransmission and Synaptic Homeostasis in the Rapid Antidepressant Effect of Sleep Deprivation
Francesco Benedetti
10:30 a.m.
Discussant: David Kupfer
114
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Data Blitz Session
This new session is comprised of rigorously timed 5 minute presentations by 12
young investigators that are linked to posters scheduled for that same evening.
11:30 a.m.
DREADDed Decision-Making: Revealing a Role for the ‘Direct’
Pathway in Reward Preference
Susan Ferguson
11:40 a.m.
A Functional Role for Interleukin 6 in Susceptibility to Depression
Georgia E. Hodes
11:50 a.m.
The Impact of Placebo on IL-18 and its Relation to Analgesic
Expectation and Central µ-Opioid Receptor Activation
Alan R. Prossin
12:00 p.m.
Motivational Saliency Signal in Ventral Striatum is modulated
by Genetic Variation in the ARC Gene Region
Caroline Zink
12:10 p.m. Dopamine Transporter Knockdown Mice exhibit Poorer
Within-Session Risk Learning in a Mouse Iowa Gambling Task
Consistent with Bipolar Mania Patients
Jared W. Young
12:20 p.m. Evidence that Mutation in Neuregulin 1, a Schizophrenia
Susceptibility Gene, alters Glucose Tolerance in Animals
Nancy M. Bivens
12:30 p.m.
A Multi-Center Investigation of Folate plus B12 Supplementation
in Schizophrenia
Joshua L. Roffman
115
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
12:40 p.m. A Zebrafish Model for the Functional Analysis of Genes in Autism
Ellen J. Hoffman
12:50 p.m. 1:00 p.m.
1:10 p.m. 1:20 p.m.
Sensory and Motor Contributions to Visuomotor Impairments in
Individuals with Autism Matthew Mosconi
The Role of Orexin in Adverse Menopause-Associated “Hot
Flash” and Anxiety Symptoms Philip Johnson
Progression of Drug Cue-Induced Phasic Dopamine Release
from Limbic to Sensorimotor Striatum Mediates Action Selection
of Drug-Taking Behavior in a Rodent Model of Drug Addiction
Ingo Willuhn
Mechanisms underlying Hippocampal Dysfunction in
Schizophrenia and related psychotic disorders
Scott A. Schobel
PL
116
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
DREADDed Decision-Making: Revealing a Role for the ‘Direct’
Pathway in Reward Preference.
Tuesday, Poster #37
Susan Ferguson, Paul Phillips, Bryan Roth, John Neumaier
Seattle Children’s Research Institute
Background: A central feature of many neuropsychiatric disorders, such as drug
addiction and obsessive-compulsive disorder, is the development of aberrant
reinforcement learning and decision-making processes. Dysregulation of the
striatum is thought to contribute to these disorders; however, the striatum is a
heterogeneous structure containing distinct populations of GABAergic medium
spiny projection neurons(MSNs) that differ in their neuropeptide composition
and form two major efferent pathways. MSNs that contain the neuropeptides
dynorphin and substance P are part of the striatonigral, or ‘direct’, pathway whereas
MSNs that contain the neuropeptide enkephalin are part of the striatopallidal, or
‘indirect’, pathway. The roles of these specific striatal sub-types in reinforcement
learning and decision-making are not yet known. As a first step toward addressing
this question, we used a novel chemical-genetic approach to determine how
modulating activity of the striatonigral pathway would change preferences in a PL
high versus low reward decision-making task. Methods: Briefly, we developed viral vectors that use the preprodynorphin
promoter to target expression of hemagluttin-tagged Gi/o-coupled DREADD
(Designer Receptor Exclusively Activated by a Designer Drug) receptors or
hemagluttin-tagged Gs-coupled DREADD receptors to striatonigral neurons. Activation of Gi/o-DREADD receptors allows for transient reduction of neuronal
excitability whereas activation of Gs-DREADD receptors allows for transient
increases in neuronal excitability using the pharmacologically inert synthetic
ligand clozapine-N-oxide (CNO). Results: After viral infusion and DREADD receptor expression into the
dorsomedial striatum of Long Evans rats, we found that decreasing activity of
striatonigral neurons impaired the acquisition of a high-reward preference in a
decision-making task for small versus large magnitude natural rewards (i.e., 1
or 4 food pellets) whereas transiently increasing activity of striatonigral neurons
117
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
DREADDed Decision-Making: Revealing a Role for the ‘Direct’
Pathway in Reward Preference
Tuesday, Poster #37 (continued)
Susan Ferguson
produced the opposite effect. However, altering activity of striatonigral neurons
had no effect on performance in this decision-making task once the high-reward
preference was established. Discussion: These findings demonstrate that the ‘direct’ pathway is an important
modulator of decision-making processes related to reward choice. They also
support the idea that striatal dysregulation contributes to the development of
aberrant reinforcement learning and decision-making that is common among
neuropsychiatric disorders.
PL
118
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
A Functional Role for Interleukin 6 in Susceptibility to Depression
Tuesday, Poster #9
Georgia E. Hodes, Viktoria Steizhammer, Sam A. Golden, Daniel J. Christoffel,
Jane Magida, Wolfgang Kluge, Carol A. Tamminga, Subroto Ghose, Erik H.F. Wong, Chi-Ming Lee, Sabine Bahn, Scott J. Russo
Mt. Sinai School Medicine, New York
Background: The pro-inflammatory cytokine Interleukin-6 (IL-6) is increased
in patients with major depressive disorder (Dowlati et al., 2009). It is currently
unknown whether IL-6 levels are altered as a result of a depressive episode or
whether IL-6 is functionally involved in the etiology of depression. Methods: Using multiplex enzyme-linked immunosorbent assays (ELISA)
we examined circulating levels of cytokines in humans experiencing their first
depressive episode or those with chronic major depressive disorder (CMDD). We
then conducted further validation of blood and brain levels of IL-6 after repeated
social defeat stress, a mouse model of depression. We also tested whether
alterations of IL-6 levels in the Nucleus Accumbens (NAc) were sufficient to
induce susceptibility to stress. IL-6 was infused into the NAc and depressionlike behavior was examined following a one-day micro-defeat, a stress paradigm
that does not induce depression-like behavior in control animals. Additionally, to
further understand the mechanism of IL-6 induction in NAc, we examined gene
expression profiles of inflammatory signaling pathways in mice exposed to social
defeat and in post-mortem tissue from subjects with major depression. Results: We found a similar increase in circulating levels of IL-6 in both
susceptible mice exposed to repeated social defeat stress and in CMDD patients
(p < 0.05), but not in resilient mice or in patients experiencing a single depressive
episode (p > 0.05). Standard antidepressant treatment did not alter IL-6 levels in
either susceptible mice or CMDD patients (p > 0.05). A more detailed analysis of
the time course of IL-6 induction in susceptible mice showed a 150 fold increase
in blood levels of IL-6 30 min after their first social defeat compared to animals
that displayed resiliency to social stress (p < 0.05). We also found that IL-6 levels
were elevated in the blood (p < 0.05) and NAc (p < 0.05) of susceptible mice 48
hours after the last social defeat. Providing a functional role for this induction,
we found that infusion of IL-6 directly into the NAc increased susceptibility to a
119
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
A Functional Role for Interleukin 6 in Susceptibility to Depression
Tuesday, Poster #9 (continued)
Georgia E. Hodes
PL
micro-defeat (p < 0.05). Surprisingly, transcription of IL-6 and its receptors was
decreased in the NAc of both susceptible and resilient mice, and in postmortem
NAc from humans with depression (p < 0.05). These data indicate that the source
of elevated NAc IL-6 protein is not from local production and we are currently
examining what the source may be. Discussion: Based on these results, we feel that repeated social defeat stress has
strong validity as a model for CMDD, especially when examining the role of
cytokines in depression. Our mouse model indicates that elevations in IL-6 blood
levels immediately after a single stressful experience may be a good biomarker for
susceptibility to stress. A single IL-6 infusion in the NAc prior to a sub-threshold
micro-defeat induced depression associated behavior indicating a functional
role for IL-6 in susceptibility to stress. While IL-6 protein levels in the NAc
were increased in animals susceptible to the effects of social defeat stress, gene
expression data indicated that local transcription of IL-6 was decreased, which
suggests that the source is likely from the periphery or other brain structures. Overall, our data suggests that individual differences in the IL-6 response to a
stressful experience may mediate the development of depression. Given the higher
levels of IL-6 in the blood of patients with CMDD and the lack of regulation
by traditional antidepressants, IL-6 may be a novel target for drug development
with great potential for use in subsets of patient with dysregulated inflammatory
pathways.
120
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
The Impact of Placebo on IL-18 and its Relation to Analgesic
Expectation and Central μ-Opioid Receptor Activation
Tuesday, Poster #219
Alan R. Prossin, Steven S. Zalcman, Alisa E. Koch, Phillip L. Campbell, Jon-Kar
Zubieta Department of Psychiatry, University of Michigan
Background: Existing evidence implicates IL-1 family pro-inflammatory
cytokines (i.e. IL-1ß, IL-18) in their association with negative affective states (i.e. pain, depression). In animal models and humans, peripheral injection of IL-1ß
has been identified as having a hypernociceptive effects and as being associated
with negative affective state. These effects are blocked by peripheral injection
of an IL-1ß antagonist, IL-1 receptor antagonist (IL-1ra) in animal models. Such
effects (i.e. IL-1ß’s hypernociception and its reversal by IL-1ra) have been shown
to result in part through effects on endogenous opioid neurotransmission in animal
models. Another IL-1 family cytokine, namely IL-18, is structurally similat to
IL-1ß and induces pro-nociceptive cytokines, including IL-1ß. However, few
studies have investigated IL-18 for its nociceptive (or negative affective) effects. Placebos have been shown to exert powerful effects on mood and pain, albeit the
interface between placebo-induced neurobiological responses and inflammatory PL
mechanisms is poorly understood, and barely examined either in animal models
or in humans. Methods: We applied a standardized sustained pain model within a PET
neuroimaging paradigm using the μ-opioid receptor selective radiotracer [11C]
Carfentanil (CFN), to determine the relation between plasma IL-18 concentration
(as measured by standard ELISA techniques) and endogenous opioid
neurotransmitter responses to a pain-stress challenge in the presence or absence
of placebo administration (isotonic saline iv with expectations of analgesic
efficacy). Following PET scanning subjects rated their pain experience using the
McGill Pain Questionnaire and measures of pain intensity. Results: We observed a significant placebo induced reduction in IL-18 plasma
levels (p<0.001). Also, placebo-induced reductions on IL-18 plasma levels were
significantly correlated with the expected magnitude of placebo effect, as subjectively
rated by the volunteers (r=0.485,p<0.012). and placebo-induced activation of
121
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
The Impact of Placebo on IL-18 and its Relation to Analgesic
Expectation and Central μ-Opioid Receptor Activation
Tuesday, Poster #219 (continued)
Alan R. Prossin
μ-opioid neurotransmission bilaterally in the nucleus accumbens (R: xyz=-9,11,
-6; Z1,65=6.04; pcorr=0.000; L: xyz=15,10,-12; Z1,65=4.55; puncorr=0.000), and
unilaterally (i.e. left) in the amygdala (xyz=17,0,-27; Z1,65=5.77; pcorr=0.000),
hippocampus (xyz=25,-7,-21; Z1,65=4.31; puncorr=0.000), subgenual cingulate
(xyz=7,35,-10; Z1,65=4.03; puncorr=0.000), and the medial thalamus (xyz=4,
-10,2; Z1,65=4.62; puncorr=0.000).
Discussion: These findings are consistent with an effect of placebo administration
on pro-inflammatory cytokines in humans in the context of expectations of
analgesia, and relationships between those effects and a neurotransmitter system,
the endogenous opioid and μ-opioid receptors, involved in pain, stress and mood
regulation.
PL
122
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Motivational Saliency Signal in Ventral Striatum is Modulated
by Genetic Variation in the ARC Gene Region
Tuesday, Poster #75
Caroline F. Zink, Sam A. Colalillo, David N. Blitzer, M. Ryan Haynes, Kuan H. Wang, Daniel R. Weinberger
National Institute of Mental Health, NIH
Background: In humans, reward anticipation has been shown to reliably and
robustly evoke activation in the ventral striatum (vSTR). Using fMRI, we have
previously demonstrated that activation in the vSTR during reward anticipation
corresponds to motivational saliency, as cues associated with greater motivational
value, rather than reward value per se, evoke greater vSTR signal than cues
associated with less motivational value. Based on animal research, such a signal
in the vSTR is likely generated by dopamine neurotransmission and/or dopamine
modulation of glutamatergic neurotransmission; however, this information
cannot be ascertained using traditional fMRI techniques in humans. In the current
study, we used “imaging genetics” to investigate the influence of dopaminergic
and glutamatergic related genetic variations on the motivational saliency signal
in the vSTR. Methods: During fMRI (3T scanner), 80 healthy participants performed an
adapted Monetary Incentive Delay task, in which reward cues informed subjects
of upcoming target difficulty in order to manipulate motivation. On each trial,
subjects responded, via single button press, to a visually presented target. On
reward trials, each successful response resulted in the receipt of $2 (represented
visually). Just prior to target appearance, participants were shown one of two cues
predicting potential reward: one associated with relatively higher motivational
value (a shorter response window) and one associated with relatively lower
motivational value (a longer response window). A control cue was not predictive
of a reward or difficulty. The imaging data were subjected to an event-related,
random-effects analysis, with a particular interest focused on neural activity
associated cue presentation. Individual first level contrasts were created for the
main effect of motivational value (high motivational value cue > low motivational
value cue) and were then entered into a second-level regression analysis to assess
123
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Motivational Saliency Signal in Ventral Striatum is Modulated
by Genetic Variation in the ARC Gene Region
Tuesday, Poster #75 (continued)
Caroline F. Zink
PL
the influence of selected genetic variations in dopaminergic and glutamatergic
system genes on related neural activity. Gender, age, and IQ were entered into the
model as regressors of no-interest to remove these potential confounds. Statistical
maps were thresholded at p < 0.05, corrected for multiple comparisons across
voxels. Results: Participants responded significantly faster to the cues associated
with higher motivational value compared to lower motivational value and no
motivational value (non-reward predicting control cues). In concordance with
previous investigations, high motivational value cues elicited significantly greater
activation in the vSTR compared to cues associated with low motivational value. Using a regression analysis, we found that a single nucleotide polymorphism
(SNP) downstream, yet in close proximity, to the ARC gene (rs9324593),
predicted the vSTR response to motivational salience, with the G allele being
associated with significantly greater vSTR BOLD signal. Discussion: ARC protein expression is activity dependent and is involved in the
endocytosis of AMPA receptors. While the effect of the rs9324593 SNP on ARC
protein is still under investigation, these results highlight a genetic variation with
potential functionality in the glutamatergic system that may account for individual
differences in the neural coding of motivational value. It is striking that of the
genetic variations studied here, a genetic variation related to the glutamatergic
system, rather than the highly hypothesized dopaminergic system, accounted
for the most variability in vSTR reactivity to motivational value. These findings
shed light on the genetic influence on neural activity underlying the evaluation
of motivationally valuable stimuli, which is highly relevant to the elucidation of
the neurobiology of mental illnesses that involve deficits in the proper evaluation
of motivationally important stimuli, including schizophrenia and drug addiction.
124
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Dopamine Transporter Knockdown Mice exhibit Poorer
Within-Session Risk Learning in a Mouse Iowa Gambling Task
Consistent with Bipolar Mania Patients
Tuesday, Poster #35
Jared W. Young, Jordy van Enkhuizen, Mark A. Geyer
University of California, San Diego
Background: Patients with Bipolar Disorder (BD) mania exhibit a range of
symptoms that contribute to many of the difficulties they face throughout their
lives. During manic episodes, patients exhibit risk-taking behaviors that are
detrimental to their well-being, including high-risk gambling. This behaviorcan
be quantified using the Iowa Gambling Task (IGT) and is not currently treated
adequately, as evidenced by the fact that poor IGT performance is also observed
during periods of euthymia. The IGT requires subjects to learn within a session
to select options that provide lower reinforcement because there is less associated
risk and more gained overall. BD patients do not learn this rule to the same level
as healthy comparison subjects. Animal models of this impaired learning are
required in order to test for novel treatments for this symptom. We have described
dopamine transporter (DAT) knockdown (KD) mice that exhibit an aberrant profile
of exploratory behavior that is consistent with BD mania patients when assessed in
the mouse and human Behavioral Pattern Monitors (BPM) respectively. Moreover,
the reduced DAT levels in unmedicated subjects with BD support the etiological
validity of this model. To further examine the similarity of this model to mania,
we 1) replicated our findings of abnormal exploration in the BPM using mice
backcrossed onto the C57BL/6 strain, and 2) tested the risk learning behavior of
these mice in a mouse version of the within-session learning IGT. Methods: After testing in the BPM, male DAT wildtype (WT) and KD (n=28 and
31 respectively) littermates were trained to holepoke for a single food reward in
any 1 of 4 locations. The reward contingencies were altered for a single 60 min
IGT session whereby 2 locations provided 2 rewards but also long punishment
durations (flashing light), while the other 2 locations continued to provide only 1
reward but with short punishment durations. Holepokes into each location were
rewarded and punished randomly on similar schedules. Good, intermediate, and
125
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Dopamine Transporter Knockdown Mice exhibit Poorer
Within-Session Risk Learning in a Mouse Iowa Gambling Task
Consistent with Bipolar Mania Patients
Tuesday, Poster #35 (continued)
Jared W. Young
PL
poor learners were quantified by taking % good choices (% holepokes into the
low reward holes from total holepokes) of the third trial period from the % good
choices from the first trial period and stratifying them as 1) >0.5, 2) between
0.5 and -0.5, and 3) <0.5 standard deviations from the mean respectively. IGT
performance was analyzed using a repeated measures ANOVA with gene and
group as between-subjects factors and trial period as the within subject factor. BPM performance was analyzed using a one-way ANOVA with gene as the
between-subjects factor. Significant main effects and interactions were subjected
to Tukey post hoc analyses.
Results: DAT KD mice exhibited increased activity (F(1,52)=46.3, p<0.0001)
and exploration (F(1,52)=4.9, p<0.05), as well as more straight-line patterns of
movement (F(1,52)=6.0, p<0.05) in the BPM. When tested in the IGT, there were
more WT (39%) than KD (29%) mice in the good learners group, despite % good
choices not differing between the 3 groups during trial period 1 (F(2,53)=1.5, ns). Although both WT (F(2,20)=13.3, p<0.0001)and KD (F(2,16)=11.0, p<0.005)
mice exhibited risk-related learning in the IGT, WT mice exhibited a trend for
more % good choices when compared with KD mice (F(1,18)=3.9, p=0.064). The
% good choices of WT and KD mice in the intermediate (F(1,21)=2.8, p=0.11)
and poor (F<1, ns) groups did not differ significantly.
Discussion: Mice with reduced expression of the DAT exhibit poor risk-related
learning in a mouse IGT consistent with subjects with BD in the human version
of the IGT. The increased risk preference of these mice in a within-session risklearning paradigm supports their use as a model of BD. Despite changing the
background strain of these mice (C57BL/6 compared with 129/S previously),
the DAT KD mice continued to exhibit an abnormal pattern of exploration in
the mouse BPM consistent with that of patients with BD mania in the human
BPM. These data provide further support for the use of these mice in testing novel
compounds to treat BD mania.
126
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Evidence That Mutation in Neuregulin 1, a Schizophrenia
Susceptibility Gene, Alters Glucose Tolerance in Animals
Tuesday, Poster #21
Nancy M. Bivens, Jay A. Gingrich
Columbia University/NYSPI
Background: Neuregulin 1 (Nrg1) alleles have been associated with schizophrenia
and Nrg1 mutant animals show psychosis related phenotypes. Nrg1 is an EGFlike growth factor implicated in muscle glucose metabolism as well as in brain
development and synaptic function. Nrg1 and its receptor ErbB4 have recently
been described as showing signals of positive selection in human populations,
although it is unclear whether this would relate to energy balance, neuronal
functions, or other growth factor related functions of Nrg1. We hypothesized that
Nrg1 mutant animals would show impaired glucose tolerance. This would have
relevance for patients with schizophrenia, as diabetes is very common, likely due
to effects of antipsychotic medications. Methods: Male animals heterozygous for Nrg1 Immunoglobulin domain isoform
null mutation (Nrg1IGhet) were fed a normal diet and tested as adults. The mutant
animals were on a c57Bl/6 background, and were tested against wild-type (WT) PL
littermate controls. Each group consisted of eight to ten animals. Blood glucose
was tested with a clinical glucometer immediately prior to and 30, 60, and 120
minutes after i.p. administration of a 1.5 g/kg glucose load. Fasting glucose was
also recorded for animals treated chronically (two months) with clozapine at 10
mg/kg in drinking water (a blood level of 59 ng/ml Norclozapine). Results: Contrary to the hypothesized result, Nrg1IGhets showed improved
glucose tolerance with an average maximum blood glucose of 165 mg/dl at 30
minutes after glucose load, while wild type littermates had a max of 220 mg/dl
at 30 minutes after glucose load (p< .05). There were no significant differences
between WT and heterozygous animals for baseline fasting glucose. There were
also no significant differences in baseline fasting glucose between animals treated
chronically with clozapine and those treated with vehicle, regardless of genotype. Discussion: This result suggests that Nrg1 genotypes may impact risk for diabetes,
for instance, reducing risk in low expression situations. It also provides alternate
127
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Evidence That Mutation in Neuregulin 1, a Schizophrenia
Susceptibility Gene, Alters Glucose Tolerance in Animals
Tuesday, Poster #21 (continued)
Nancy M. Bivens
frameworks for interpreting the psychosis and seizure related phenotypes found
both in Nrg1 hypomutant animals and in Nrg1 overexpression mutants. It is of
particular relevance to understanding neurological phenotypes produced by acute
peripheral administration of recombinant Nrg1, as a strong immediate effect on
blood glucose levels may mediate many downstream effects. Nrg1 effects on
blood glucose metabolism may also be of interest in determining populations at
higher risk for side effects of antipsychotic medications.
PL
128
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
A Multi-Center Investigation of Folate plus B12
Supplementation in Schizophrenia
Tuesday, Poster #64
Joshua L. Roffman, Steve Lamberti, Eric Achtyes, Eric A. Macklin, Gail Galendez,
Lisa Raeke, Noah J. Silverstein, Dan Tuinstra, Michele Hill, Donald C. Goff
Massachusetts General Hospital / Harvard Medical School
Background: Negative symptoms, which include apathy, impoverished speech,
flattened affect, and social withdrawal, cause substantial morbidity and functional
impairment among patients with schizophrenia. These problems are compounded
by the lack of effective treatments for negative symptoms. Previous work from
our group and others suggests a link between negative symptom severity and
altered folate metabolism in schizophrenia. Moreover, a common genetic variant
in MTHFR, which plays a key role in folate metabolism, may contribute to this
pattern: the hypofunctional 677T allele of MTHFR, each copy of which reduces
MTHFR activity by 35%, has been associated with increased schizophrenia
risk and specifically with increased severity of negative symptoms and related
neurocognitive dysfunction. A previous pilot investigation by our group suggested
that folate supplementation may improve negative symptoms in schizophrenia, PL
but only among patients who carried the 677T allele. Here, we conducted a large
multi-site investigation to determine whether folate plus B12 supplementation,
by itself and in concert with MTHFR 677C>T genotype, influenced symptom
severity among medicated schizophrenia patients. Methods: We enrolled outpatients with schizophrenia at three sites (Massachusetts
General Hospital, University of Rochester, and Michigan State University). Patients were taking stable regimens of antipsychotic medication upon enrollment
and were continued on their medications throughout the study. A total of 140
patients were randomized, double-blind, in a 2:1 ratioto receive 16 weeks of daily
treatment with either 2 mg folic acid plus 400 mcg B12 or placebo. Fasting serum
folate values and clinical measures including the Scale for the Assessment of
Negative Symptoms (SANS) and Positive and Negative Syndrome Scale (PANSS)
were obtained at screening and at 2, 4, 8, 12, and 16 week follow-up visits. For
data analysis, linear mixed models that included treatment status (active versus
129
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
A Multi-Center Investigation of Folate plus B12
Supplementation in Schizophrenia
Tuesday, Poster #64 (continued)
Joshua L. Roffman
PL
placebo), MTHFR genotype (C/C versus T allele carrier), and baseline folate
levels were used to examine changes in SANS total score, PANSS total score,
and PANSS positive symptoms over time. Alpha (2-tailed) was set at 0.05.
Results: Preliminary analyses are as follows, with additional analyses to
be presented at the meeting. The two treatment groups did not differ on any
demographic or clinical variable at baseline. Sixteen week retention was 78%. Serum folate levels rose significantly in the active treatment group. SANS total
scores improved significantly in the folate group and significantly more compared
to the placebo group, which did not show a change in negative symptoms. PANSS
total scores improved significantly in the folate group, but not significantly more
compared to placebo. No effects of treatment were seen for PANSS positive
symptoms. MTHFR genotype influenced each main outcome variable. For SANS,
genotype effects differed by treatment group, as only T allele carriers showed
significant improvement compared to placebo. For PANSS total and PANSS
positive symptoms, C/C patients improved significantly across both treatment
groups on average and significantly more than T carriers, who did not improve,
regardless of treatment. Discussion: Folate plus B12 supplementation confers a specific benefit for
negative symptoms of schizophrenia, for which no other treatment is available. Confirming our hypothesis and replicating previous results from a smaller
sample, these effects were especially pronounced in patients who carried the lowfunctioning 677T variant of MTHFR, which has previously been associated with
increased negative symptom severity and related cognitive impairment. Studies of
wider-scale implementation of folate plus B12 supplementation in schizophrenia
are warranted, as are prospective investigations of whether folate and B12 status
influence schizophrenia risk, especially among individuals who are genetically
predisposed to altered folate metabolism.
130
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
A Zebrafish Model for the Functional Analysis of Genes in Autism
Tuesday, Poster #1
Ellen J. Hoffman, Antonio Giraldez, Matthew State
Yale University
Background: A critical challenge in the genetics of neuropsychiatric disorders
is distinguishing deleterious rare mutations from neutral variants, as rare
sequence and structural variants have been identified throughout the genomes
of both affected and unaffected individuals, including at candidate gene loci. The ability to distinguish rare functional from rare neutral variation is critical
for confirming the association of risk genes carrying rare alleles. For this reason,
we propose to develop an in vivo model that will allow us rapidly to differentiate
mutations that alter the function of susceptibility genes from neutral rare variants. This novel approach capitalizes on critical advantages of zebrafish, including
visualization of the developing nervous system in transparent embryos, ease of
genetic manipulation, and large progenies that facilitate the conduct of largescale pharmacologic screens. Therefore, we generated zebrafish knockouts of
the ASD risk gene, CNTNAP2, using the emerging technology of zinc finger
nucleases (ZFN). We anticipate that CNTNAP2 will be particularly informative PL
in this regard, as homozygous disruption of CNTNAP2 by a single base pair
deletion in the Old Order Amish population causes a monogenic syndrome that is
highly associated with ASD. In addition, the State laboratory identified a de novo
chromosome 7q inversion disrupting CNTNAP2 in a child with cognitive and
social delay. Our goals in developing this model are: 1) to leverage any distinctive
reproducible and quantifiable phenotype for forward genetic studies that will
help to elaborate conserved molecular mechanisms and pathways involving these
susceptibility genes; and 2) to test the relative ability of the wild type human
gene compared to constructs containing rare mutations identified in affected and
unaffected individuals to rescue the identified phenotype. Methods: We identified the zebrafish orthologs of the human CNTNAP2 gene
by conducting a search of the zebrafish genome (Zv7) in the National Center for
Biotechnology Information (NCBI) database. We analyzed the expression patterns
of these paralogs in zebrafish embryos at 30 and 48 hours post fertilization by in
131
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
A Zebrafish Model for the Functional Analysis of Genes in Autism
Tuesday, Poster #1 (continued)
Ellen J. Hoffman
PL
situ hybridization. We utilized ZFN directed against exons 2 and 3 of CNTNAP2a
and CNTNAP2b, respectively, to generate targeted germline deletions in each
gene. Founders were generated by injecting mRNA encoding ZFN (SigmaAldrich) targeting either CNTNAP2a or 2b into embryos at the one-cell stage. Founders were identified by screening the progeny of incrosses of ZFN-injected
adult fish by PCR followed by high-resolution fragment analysis. Founders were
outcrossed to wild-type fish, and the heterozygous CNTNAP2a and 2b knockouts
were incrossed to generate viable homozygous knockouts. Results: We identified two zebrafish orthologs of CNTNAP2, CNTNAP2a and
2b. In situ analysis of these transcripts between 30 and 48 hours after fertilization
revealed expression of both paralogs in the CNS, predominantly in the midbrain
and hindbrain. These paralogs demonstrate distinct yet partially overlapping
expression patterns. Utilizing ZFN targeting each gene, we generated multiple
zebrafish founders with deleterious germline mutations in both CNTNAP2a and
2b and outcrossed these founders to wild-type fish, producing viable heterozygous
CNTNAP2a and 2b knockouts. These mutations are predicted to be damaging as
they occur early in the coding regions of each gene and produce a frameshift,
resulting in a premature stop codon and truncation of the protein in or immediately
after the N-terminal discoidin domain. In total, we have successfully generated
zebrafish founders with deleterious germline mutations in both CNTNAP2
paralogs. We are currently conducting a battery of morphological and behavioral
assays to identify quantifiable phenotypes in CNS structure and larval neural
circuits in mutant fish. Discussion: Our experiments lay the foundation for the use of zebrafish as
a model system for elucidating the function of susceptibility genes in ASD. We have identified the zebrafish orthologs of the ASD susceptibility gene,
CNTNAP2, demonstrated its expression in the zebrafish CNS, and successfully
generated the first ZFN-induced deletions in each CNTNAP2 paralog. This model
has tremendous promise for illuminating common pathways involving ASD
132
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
A Zebrafish Model for the Functional Analysis of Genes in Autism
Tuesday, Poster #1 (continued)
Ellen J. Hoffman
susceptibility genes and rapidly assessing the functional consequences of rare
sequence variation in a risk gene. Future applications of this research include
large-scale pharmacological screens to identify novel therapeutic targets aimed at
the mechanisms underlying the core deficits of ASD.
PL
133
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Sensory and Motor Contributions to Visuomotor Impairments
in Individuals with Autism
Tuesday, Poster #76
Matthew W. Mosconi, Suman Mohanty, Lauren Schmitt, Edwin H. Cook, David
E. Vaillancourt, John A. Sweeney
University of Texas Southwestern Medical Center
PL
Background: Sensorimotor disturbances are present in the majority of individuals
with autism. It remains unclear whether these deficits reflect impaired processing
of sensory feedback for action planning, or fundamental deficits in motor control. Methods: Twenty-six individuals with autism and 26 age- and IQ-matched
healthy controls performed sustained precision grip force tasks in which the
amplitude of the required target force (motor manipulation) and the precision of
visual feedback (sensory manipulation) each were varied. They viewed a white
bar that moved upwards with increased grip force toward a fixed green target
bar. Subjects were instructed to sustain a constant force in order to stabilize the
white bar at the level of the green bar. During the motor manipulation, the green
target bar was set to 5, 25, 45, 65 or 85% of individual subjects’ maximum force
contraction. During the sensory manipulation, the vertical distance the white bar
moved per Newton of force applied was set to visual angles of .02, .06, .19, .62,
2.02, 6.66 and 21.13 deg. When the visual angle was small, the white bar moved
a smaller distance for every Newton of force applied, increasing the information
fidelity of sensory feedback. All trials were 15 sec in duration and were followed
by 15 sec of rest. Subjects completed dominant and non-dominant hand testing
separately. Results: The mean force did not differ between subjects with autism compared to
controls. However, subjects with autism showed reduced control of their motor
output as demonstrated by greater force variability during the trial. This impairment
was more robust at greater force amplitudes, especially for the non-dominant
hand. Changes in the precision of sensory feedback did not affect force control
impairments in individuals with autism; force variability was increased to similar
degrees across visual angles in individuals with autism compared to controls. Increased variability of sustained force was associated with clinical ratings of
communication impairment and motor stereotypies in individuals with autism. 134
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Sensory and Motor Contributions to Visuomotor Impairments
in Individuals with Autism
Tuesday, Poster #76 (continued)
Matthew W. Mosconi
Discussion: We examined whether deficits in visuomotor control in autism result
from disruptions to sensory feedback processing and/or feedforward motor
systems. Our results provide clear evidence that visuomotor impairments are due
to deficits in producing motor output, and this deficit appears to be independent
of the quality of sensory feedback. Further, motor impairments appear to be
related to altered development of language skills and stereotypies, indicating
a relationship with core clinical features of the disorder. This pattern of motor
deficit implicates dysfunction of lobules V-VI and Crus I/II of the cerebellum
which generate efferent motor commands to multiple cortical regions, including
motor, premotor, and parietal cortices. Our finding that changes in the quality of
visual feedback do not affect the degree of motor control impairment in autism
suggests that motor cortices which scale their activity according to the precision of
visual feedback may be relatively spared. Combined with findings from multiple
postmortem studies of autism documenting reduced Purkinje cell size and density,
these results suggest that cerebellar abnormalities may underlie the dyspraxia and
poor fine motor control that are present in the majority of individuals with this
disorder.
135
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
The Role of Orexin in Adverse Menopause-Associated “Hot
Flash” and Anxiety Symptoms
Tuesday, Poster #218
Philip Johnson, Lauren Federici, Stephanie Fitz, Todd Skaar, Janet Carpenter,
Anantha Shekhar
Indiana University School of Medicine
PL
Background: Menopause is a condition in which estrogen levels are severely
depleted which leads to a cluster of adverse menopausal symptoms such as
cutaneous vasomotor/sudomotor “hot flashes”, anxiety, sleep disturbances, and
appetite changes (Freeman et al., 2005; Seritan et al., 2010). Currently, estrogen
replacement therapy is the first line treatment for menopausal symptoms. However, it is no longer acceptable because of shifts in its risk (e.g., cancer)benefit ratio uncovered by the Women’s Health Initiative study (Rossouw et
al., 2002). Therefore, there is a need for non-hormonal therapies to reduce the
incidence of adverse menopausal-related symptoms. Unfortunately, the scientific
understanding of menopausal symptoms is limited and the few non-hormonal
therapies that exist are much less effective than estrogen replacement and have
adverse side effects (Nelson et al., 2006). Although the understanding of the
neural circuits involved in menopausal symptomology is not clear, it is commonly
accepted that the hypothalamus plays a critical role (Miller and Li, 2004). For
instance, menopausal symptoms are clearly induced by estrogen withdrawal and,
within the brain, estrogen receptors are highly expressed and fairly concentrated
in the hypothalamus (Laflamme et al., 1998). Furthermore, the hypothalamus
plays a critical role in setting the thermoneutral zone (Guyton, 1976), and
postmenopausal women have a reduced thermoneutral zone (i.e., lower thresholds
of ambient temp increases to elicit hot flashes) (Freedman and Krell, 1999). In
1998, a neuropeptide called orexin (ORX: also known as hypocretin) was found
to be exclusively synthesized in the perifornical hypothalamus (De Lecea et al.,
1998 and Sakurai et al., 1998) that has long been known to play a critical role in
wake-promotion, thermoregulation, and anxiety (Ferguson and Samson, 2003;
Sakurai, 2007), which are all components of menopausal symptoms. Recently
our lab determined that a hyperactive ORX system is linked to an animal model
136
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
The Role of Orexin in Adverse Menopause-Associated “Hot
Flash” and Anxiety Symptoms
Tuesday, Poster #218 (continued)
Philip Johnson
of panic disorder and in patients with panic symptoms (Johnson et al., 2010,
Nature Medicine). Similarly, in female rats ORX expression in the hypothalamus
is highest when estrogen levels are low (Porkka-Heiskanen et al., 2004). Methods: In order to test this hypothesis, we removed the ovaries (ovariectomy:
OVEX) in female rats to model an acute menopausal state then surgically
implanted radiotelemetry probes to measure core body and tail temp. Next, we
created a novel model of menopause-induced “hot flashes” utilizing peripheral
vasodilators that reliably result in tail-flushing response in female rats, utilizing
either an acute CO2 inhalation-induced or yohimbine-induced vasodilation.
Results: We were able to demonstrate that submaximal doses of these compounds
were able to induce profound and significantly greater tail flushes in OVEX rats
compared to SHAM rats. Utilizing these and other accepted models of anxiety and
autonomic responses, we determined that pretreating OVEX rats with a systemic
injection of a centrally active ORX 1 receptor antagonist: 1) blocked OVEXinduced anxiety behavior; and 2) blocked an exacerbated “hot flash”-associated
increase in tail flushes and temperature following a clinically relevant “hot flash”
provocation stimulus.
Discussion: These results clearly demonstrate preclinical evidence that orexin
antagonists may be very effective therapies for post-menopausal hot flashes. This
is consistent with clinical data demonstrating that compared to reproductive female
controls, menopausal women had 300% higher ORX levels in their cerebrospinal
fluid and these levels were restored to control levels following estrogen
replacement (El-Sedeek et al., 2010). Therefore, loss of normal inhibitory control
by estrogens of the ORX system may lead to menopausal-related symptoms, and
ORX antagonists could constitute a potential novel treatment strategy for adverse
menopausal symptoms.
137
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Progression of Drug Cue-Induced Phasic Dopamine Release
from Limbic to Sensorimotor Striatum Mediates Action
Selection of Drug-Taking Behavior in a Rodent Model of Drug
Addiction
Tuesday, Poster #36
Ingo Willuhn, Barry J. Everitt, Paul E.M. Phillips
University of Washington
PL
Background: Dopamine neurotransmission in the ventral striatum is strongly
implicated in the acute reinforcing effects of drugs of abuse. After repeated drug
intake, the dorsolateral striatum is thought to become increasingly involved
in the control of drug taking and the automation of this behavior. Dopamine
neurotransmission in the dorsolateral striatum is potentially regulated by
ventral striatal circuitry via serial striatonigrostriatal connection. Therefore,
we simultaneously characterized phasic dopamine signaling in these two brain
regions using an animal model of drug addiction. Changes in dopamine release
were measured over the course of weeks in rats self-administering cocaine. Furthermore, we tested whether dopamine release in the dorsolateral striatum is
dependent upon ventral striatal circuitry. Finally, we blocked dopamine signaling
in the dorsolateral striatum to further investigate its role in the control of drug
taking. Methods: Multiple carbon-fiber microelectrodes for fast-scan cyclic voltammetry
or guide cannulas for microinfusion of the dopamine receptor antagonist alphaflupenthixol were chronically implanted in the striatum of rats bearing indwelling
intravenous catheters for drug self-administration. Animals had access to cocaine
for one hour per day for 20 days. During a self-administration session, a nose
poke into the active hole elicited a cocaine infusion (FR1, 0.5 mg/kg/infusion)
that was accompanied by a 20-second presentation of an audiovisual stimulus
(drug cue). Additional responses during this time-out period or nose pokes into
the inactive hole (control) were without consequences. Results: Throughout self-administration training, we observed phasic dopamine
release in the ventral striatum associated with contingent and non-contingent
presentation of the drug cue. In contrast, cue-related dopamine signals in the
138
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Progression of Drug Cue-Induced Phasic Dopamine Release
from Limbic to Sensorimotor Striatum Mediates Action
Selection of Drug-Taking Behavior in a Rodent Model of Drug
Addiction
Tuesday, Poster #36 (continued)
Ingo Willuhn
dorsolateral striatum developed only during later stages of training. Unilateral
lesion of the ventral striatum with quinolinic acid inhibited the development of such
dorsolateral dopamine signals in the ipsilateral, but not contralateral, hemisphere
without affecting drug intake. Bilateral blockade of dopamine receptors in the
dorsolateral striatum with alpha-flupenthixol increased drug intake both early in
training, before dorsolateral dopamine signaling were detected, as well as later in
training, when dopamine signals were present. However, the efficiency of drug
intake (successful / total nose pokes) was affected at the late time point only. Discussion: Our results demonstrate that phasic dopamine signaling in the
striatum in response to the presentation of drug cues is dynamic and region
specific, developing in the ventral then dorsolateral striatum sequentially. This
progression of dopamine signaling from limbic to sensorimotor regions of the
striatum requires intact ventral striatal circuitry and plays a role in the action
selection of drug-taking behavior. Overall, these data implicate recruitment of
sensorimotor striatal circuitry for dopamine-mediated encoding of drug cues in
the development of long-term efficient performance of the drug-taking response
and, thus, such recruitment may contribute to automated, habitual drug intake
observed in addicts.
139
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Mechanisms Underlying Hippocampal Dysfunction in
Schizophrenia and Related Psychotic Disorders
Tuesday, Poster #147
Scott A. Schobel, Nashid Chaudhury, Cheryl M. Corcoran, Martin Styner,
Beatriz Paniagua, Jeffrey A. Lieberman, Holly Moore, Scott A. Small
University Medical Center, New York and New York State Psychiatric Institute
PL
Background: Hippocampal hypermetabolism characterizes schizophrenia and
related psychotic disorders. Here, we hypothesized that elevations in extracellular
glutamate underlie disease-associated increases in hippocampal metabolism in
early stages of psychotic disorder and that elevations in extracellular glutamate
drive hippocampal tissue loss upon progression to first episode psychosis. To test
this hypothesis, we exploited a high-resolution variant of functional magnetic
resonance imaging (fMRI) that can map hippocampal subregional metabolism
in patients and in animal models. To translate observed functional imaging
abnormalities into underlying neurochemical mechanisms, we applied a glutamate
biosensor to measure evoked changes in extracellular glutamate in mice exposed
to acute ketamine challenge (30mg/kg), a pharmacological condition that reliably
produces positive, negative, and cognitive symptoms of acute psychosis in
humans. Methods: High-resolution functional and structural MRI in a prospective
longitudinal cohort of individuals at clinical risk for psychotic disorders imaged
at baseline (n=25) and at 2.5 years prospective follow-up for clinical outcomes
(n=20) and a combined high-resolution structural and functional MRI/glutamate
biosensor approach in C57b6 WT mice exposed to acute, as well as chronic
ketamine challenge (30mg/kg). Results: Using this cross-species imaging approach, we observed that as compared
to individuals at clinical risk for psychosis who did not progress to psychosis
over 2.5 years clinical follow-up, those individuals who did progress to psychotic
disorders exhibited hypermetabolism in the CA1 subfield (F1,19=6.2, p=.03) and
subiculum (F 1,19=5.8, p=.03), replicating an imaging profile we have previously
described in schizophrenia. Hippocampal hypermetabolism at baseline in left
anterior CA1 is associated with subsequent hippocampal volume reduction in
140
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Data Blitz Session
Kona 4
Mechanisms Underlying Hippocampal Dysfunction in
Schizophrenia and Related Psychotic Disorders
Tuesday, Poster #147 (continued)
Scott A. Schobel
progressors to first episode psychosis (r=.7, p=.003). Morphometric shape
change localizes this volume reduction to the left anterior CA1 and subiculum
subregions. In mice, with acute ketamine challenge (30 mg/kg, i.p.), we again
observed a selective increases in metabolism within the CA1 subfield (F4,13=4.1,
p=.02) and subiculum (F4,13=3.8, p=.03). Second, using an in vivo glutamate
biosensor, we found that acute ketamine challenge (30mg/kg i.p.) selectively
produced increases in extracellular glutamate in the CA1 (t11=2.4, p=.03) and
subiculum (t10=2.5, p=.03); there were no changes observed in medial entrorhinal
cortex or dentate gyrus. Pretreating mice over five-days with glutamate-reducing
agents LY379268 (10mg/kg), gabapentin (600mg/kg), or lamotrigine (10mg/kg)
showed that relative to saline pretreatment, each drug pretreatment significantly
decreased the post-ketamine extracellular glutamate response (LY379268:
t10=2.4, p=.04, lamotrigine: t10=2.9, p=.02, gabapentin: t10=3.1, p=.01); as well
as the evoked CBV response (LY379268: t8=9.1, p<.001, gabapentin: t8=3.9,
p=.004, lamotrigine: t7=1.9 p=.09). Discussion: Our study results confirm that hypermetabolism in the CA1 subfield
and the subiculum are characteristic of schizophrenia and related psychotic
disorders, and are inducible by acute ketamine challenge in rodents using the
same imaging paradigm. Moreover, the pharmacological model shows that this
hypermetabolism is mediated by increases in extracellular glutamate, and can
be blocked by diverse glutamate-limiting therapeutic strategies. Longitudinal
imaging results in patients across the transition to psychosis suggest that
hippocampal hypermetabolism drives a pattern of hippocampal volume loss
that is maximal in the left anterior body of the structure. These findings have
immediate implications for a glutamate-limiting preventive therapeutic strategy
in patients at clinical high risk for psychotic disorders who exhibit hippocampal
hypermetabolism.
141
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kona 1-3
Travel Awardee Research Presentations
Basic Neuroscience of Addiction
Chair: Stephanie O’Malley
11:30 a.m.
12:10 p.m.
12:50 p.m.
Orexin Mediates Yohimbine Actions in BNST and
Impaired Extinction of Cocaine Place Preference through a
Norepinephrine-Independent Process
Kelly L. Conrad
Csnk1e is a Genetic Regulator of Sensitivity to Psychostimulants
and Opioids
Camron D. Bryant
The Glial Modulator Propentofylline impairs Reinstatement in a
Rat Model of Cocaine Abuse
Kathryn J. Reissner
PL
142
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kona 1-3
Orexin mediates Yohimbine Actions in BNST and
Impaired Extinction of Cocaine Place Preference through a
Norepinephrine-independent Process
Kelly L. Conrad
Vanderbilt University Medical Center
Background: The alpha2 adrenergic receptor (a2-AR) antagonist yohimbine
is a widely used tool for the study of anxiogenesis and stress-induced drugseeking behavior. We previously demonstrated that yohimbine produces both a
paradoxical depression of excitatory transmission in the bed nucleus of the stria
terminalis (BNST) along with an impairment of extinction of cocaine conditioned
place preference (cocaine CPP) that are independent of (a2-AR) signaling. The
target of yohimbine that mediates these actions, however, is unknown.
Methods: We utilized whole-cell patch clamp and field potential recordings in
the BNSTex vivo to examine the effects of yohimbine (30-50 uM), orexin A
(100 nM), orexin 1 receptor (Ox1R) antagonist (SB-334867; 2-5 uM), orexin 2
receptor (Ox2R) antagonist (JNJ-10397049), and a novel dual OxR antagonist
MTBDQ (1uM), and the norephinephrine transport blocker reboxetine (100
nM) on excitatory transmission in wild-type and prepro-orexin knockout mice
PL
(Ox-KO). The ability of yohimbine to directly activate Ox1R was investigated
utilizing an Ox1R expressing stable cell line. To examine the potential behavioral
implications of these findings, the ability of OxR antagonists to alter yohimbine
impairment of cocaine CPP extinction was investigated
Results: Yohimbine-induced depression of excitatory transmission in the BNST
is blocked by two distinct OxR antagonists, absent in Ox-KO mice, and mimicked
by exogenous orexin A application but not blockade of norepinephrine uptake. Moreover, we show that the action of yohimbine is not through direct activation
of Ox1R, suggesting yohimbine facilitates orexin release. Behaviorally, we find
that yohimbine-induced impairment of cocaine CPP extinction is also blocked by
an Ox1R antagonist.
Discussion: These data describe a major new mechanism for orexin action
on excitatory anxiety circuits, and show that significant behavioral actions of
yohimbine may be directly dependent upon orexin signaling and independent of
norepinephrine.
143
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kona 1-3
Csnk1e is a Genetic Regulator of Sensitivity to Psychostimulants
and Opioids
Camron D. Bryant
University of Chicago
PL
Background: Csnk1e, the gene encoding casein kinase 1-epsilon, has been
implicated in sensitivity to amphetamines. We previously identified a quantitative
trait locus (QTL) in mice for methamphetamine (MA)-induced locomotor activity
near Csnk1e. Additionally, CSNK1E genetic variants have been associated with
amphetamine euphoria and heroin addiction in humans. The casein kinase 1
(CK-1) family phosphorylates dopamine- and cyclic adenosine monophosphateregulated neuronal phosphoprotein-32 (DARPP-32) and co-administration of the
casein kinase 1-delta (Csnk1d)-preferring inhibitor PF-670462 with amphetamines
inhibits DARPP-32 phosphorylation and the locomotor stimulant response. This
suggests that CK-1 acts via the DARPP-32 pathway to influence psychostimulant
sensitivity. The objective of this study was two-fold. First, we wished to narrow
the QTL near Csnk1e that influences MA sensitivity and test for its relevance in
opioid sensitivity. Second, we wished to directly test the hypothesis that Csnk1e
regulates sensitivity to psychostimulants and opioids using knockout mice and
selective pharmacological inhibition.
Methods: We conducted a genome-wide QTL mapping study of MA-induced locomotor activity (2 mg/kg, i.p.) in C57BL/6J (B6) x DBA/2J (D2)-F2 mice
and a highly recombinant F8 advanced intercross line. We also generated
and phenotyped B6.D2Csnk1e and D2.B6Csnk1e reciprocal congenic lines
capturing Csnk1e (78-86.8 Mb and 78.7-81.6 Mb, respectively; Csnk1e = 79.25). B6.D2Csnk1e mice were also tested for sensitivity to the mu opioid receptor
agonist fentanyl (0.2 mg/kg, i.p.). Additionally, mice harboring a null allele of
Csnk1e were tested for MA-induced locomotor activity. Last, we tested the effect
of the Csnk1e-selective inhibitor PF-4800567 (40 mg/kg, i.p.) on the locomotor
stimulant response to methamphetamine, fentanyl, or saline.
Results: We identified a QTL on chromosome 15 for MA sensitivity that contained
Csnk1e (63-86 Mb; Csnk1e = 79.25 Mb) and further narrowed the locus to 3 Mb
using reciprocal congenic mice (78.7-81.6 Mb). This locus also affected fentanyl
sensitivity. Csnk1e knockout mice showed an increase in basal and MA-stimulated
144
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kona 1-3
Csnk1e is a Genetic Regulator of Sensitivity to Psychostimulants
and Opioids (continued)
Camron D. Bryant
locomotor activity. The selective Csnk1e inhibitor PF-4800567 also produced
an increase in MA- and fentanyl-induced locomotor activity but did not have
any effect by itself. Interestingly, the enhancement of MA sensitivity with PF4800567 depended on the genotype of congenic mice, providing further support
that genetic variation in Csnk1e regulates the response to MA.
Discussion: These results show that a narrow genetic locus that contains Csnk1e
is associated with differences in sensitivity to MA and fentanyl. Interestingly, the
precise locus has also been identified for variation in ethanol consumption in mice. The convergence of a QTL on this locus for the response to psychostimulants,
opioids, and ethanol implicates gene(s) acting via dopaminergic mechanisms. Gene knockout and selective pharmacological inhibition of Csnk1e define its
role as a negative regulator of sensitivity to psychostimulants and opioids. This
suggests that our previous findings regarding the Csnk1d-preferring inhibitor PF670462 were mediated by Csnk1d and that the two isoforms exhibit opposing
control over the response to drugs of abuse. In support of this hypothesis and PL
similar to the results with Csnk1e knockout mice, overexpression of Csnk1d in the
forebrain has been shown to increase basal and amphetamine-induced locomotor
activity. Future studies will examine the role of DARPP-32 and cell type-specific
expression of Csnk1e and Csnk1d in regulating sensitivity to drugs of abuse as
well as their role in motivational behaviors such as pain and reward. 145
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kona 1-3
The Glial Modulator Propentofylline impairs Reinstatement in a
Rat Model of Cocaine Abuse
Kathryn J. Reissner
Medical University of South Carolina
PL
Background: Accumulating evidence indicates that activation of glial cells is
a cellular feature associated with drug abuse. Noncontingent administration of
cocaine leads to upregulation of markers for glial activation, including GFAP and
vimentin, 3 weeks after one week of daily cocaine administration. Moreover,
glial high affinity glutamate transporters GLT-1/EAAT2 and GLAST/EAAT1
are also chronically downregulated following cocaine self-administration and
extinction training. Because these observations occur several weeks following
withdrawal, the findings collectively suggest that glial reactivity may represent a
long-lasting cellular adaptation following cessation of cocaine use, and may thus
contribute to mechanisms of relapse. As an initial step to test this hypothesis,
we have employed the glial modulator propentofylline in a rat reinstatement
model of cocaine abuse. Propentofylline is a xanthine derivative with numerous
cellular functions, including adenosine uptake inhibition and phosphodiesterase
inhibition. Propentofylline has also been shown to reverse markers for reactive
gliosis following nerve injury, including changes in expression of GFAP, GLAST,
and GLT-1.
Methods: Following intrajugular catheterization and recovery from surgery, male
Sprague-Dawley rats were trained to self-administer cocaine on an FR1 schedule
during 2h sessions each day. A lever press resulted in a cocaine infusion (0.2 mg
per infusion), as well as presentation of light and tone drug-paired cues. Criteria
for self-administration was a minimum of 10 sessions receiving 10 infusions or
more. Following self-administration, rats entered an extinction training phase
for 2 weeks, during which a lever press no longer resulted in drug or drugpaired cues. During the last 6 days of extinction, rats received an injection of
propentofylline (10 mg/kg, i.p.) or saline 30 min prior to the extinction session. One final injection was also given 30 min prior to a cue-prime reinstatement test. In a separate series of experiments, one injection of propentofylline (10 mg/kg or
25 mg/kg, i.p.) or saline was given 30 min prior to a reinstatement test only.
Results: Animals that received chronic propentofylline treatment during the last
6 days of extinction and prior to a reinstatement test reinstated significantly less
146
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kona 1-3
The Glial Modulator Propentofylline impairs Reinstatement in a
Rat Model of Cocaine Abuse (continued)
Kathryn J. Reissner
than animals receiving saline only, as measured by number of active lever
presses (experiment 1). However, acute propentofylline given once prior to
a reinstatement test was without effect at 10 mg/kg or 25 mg/kg (experiment
2). Animals receiving chronic propentofylline or saline (experiment 1) were
allowed to re-enter extinction training following reinstatement without continued
treatment, for a second test one week later. Number of active lever presses
was not significantly different between animals receiving propentofylline or
saline in the second reinstatement test, indicating that continued treatment with
propentofylline is necessary for an enduring effect on reinstatement.
Discussion: Results presented here indicate that chronic administration of the
glial modulator propentofylline can impair drug seeking behavior. These findings
indicate that reactive gliosis may represent a credible target for pharmacotherapy
for cocaine abuse. Ongoing studies are designed to measure changes in protein
expression for glial markers known to be affected by cocaine abuse which are
also identified targets for propentofylline, including GFAP, GLT-1, and GLAST. 147
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kohala 4
Travel Awardee Research Presentations
Basic Neuroscience of Depression, Anxiety & Stress
Chair: Xavier Castellanos
11:30 a.m.
Differential Role of ΔFosB in the Prefrontal Cortex in CCK
Sensitivity and Vulnerability to Stress
Vincent Vialou
12:10 p.m.
Modulation of Adult Hippocampal Neurogenesis through HPA
Axis Activity determines the Divergent Effects of Distress and
Eustress on Affective Disorders
Michael L. Lehmann
12:50 p.m.
Optical Activation of Nucleus Accumbens Neurons modulates
Depression- and Anxiety-Like Behaviors
Mary Kay Lobo
PL
148
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kohala 4
Differential Role of ΔFosB in the Prefrontal Cortex in CCK
Sensitivity and Vulnerability to Stress
Vincent Vialou
Mount Sinai School of Medicine
Background: Chronic social defeat stress in mice induces long-lasting
abnormalities, including social avoidance, which can be normalized by chronic,
not acute, antidepressant treatment (Berton et al., 2006). However, a significant
proportion of defeated mice avoid these abnormalities (Krishnan et al., 2007),
allowing a look at mechanisms of resilience in addition to vulnerability. Recently, we observed decreased expression of markers of neuronal activity
in the medial prefrontal cortex (mPFC) of susceptible mice (Covington et al.,
2010), and found that optogenetic stimulation of the mPFC of susceptible mice
reversed their depression-like symptoms (Covington et al., 2010). A potential
molecular mechanism underlying the susceptible phenotype could be the release
of cholecystokinin (CCK) in the mPFC, which has been shown to be released
during social stress and to mediate an anxiogenic response.
Methods: We first quantified ΔFosB levels by immunohistochemistry, a marker
implicated in the persistent neuronal alterations induced by chronic exposure to
stress. In order to evaluate its role in sensitivity to stress, we used viral-mediated PL
gene transfer into different regions of the PFC. We used quantitative PCR to
evaluate CCK-B receptor, a known target gene of ΔFosB, after chronic social
defeat and after ΔFosB manipulation. Finally, we tested the antidepressant effect
of an infusion of CI-988 (1ng), a CCK-B antagonist, into the mPFC of susceptible
mice.
Results: We show here that social defeat induces long-term molecular adaptations
differentially in susceptible vs. resilient mice in the mPFC as well as in the
orbitofrontal cortex (OFC). Resilient mice show a greater induction of ΔFosB
in the OFC, while susceptible mice show greater induction in the mPFC. These
results suggest that ΔFosB induction after chronic stress in these two brain regions
regulates specific neural pathways that contribute to the susceptible and resilient
phenotype, respectively. Preliminary evidence indicates that ΔFosB overexpression
in the OFC reduces immobility in the forced swim test, an antidepressantlike effect. We are currently testing whether ΔFosB overexpression in the
149
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kohala 4
Differential Role of ΔFosB in the Prefrontal Cortex in CCK
Sensitivity and Vulnerability to Stress (continued)
Vincent Vialou
PL
OFC of susceptible mice promotes resilience. Conversely, ΔFosB overexpression
in the mPFC increased social avoidance induced by social stress, a pro-depressant
effect. To verify that CCK-B is implicated in the behavioral deficits induced
by social stress, we infused CI-988 in the mPFC of susceptible mice. This was
sufficient to reverse the social avoidance induced by social defeat. We are currently
testing the binding of ΔFosB at the CCK-B promoter gene after social defeat.
Discussion: Long-term alterations in the mPFC mediate the behavioral
abnormalities observed after social defeat. We identified ΔFosB as mediator
of the depressive-like phenotype induced by chronic stress. Blockade of CCK
actions in the mPFC promotes social interaction, an antidepressant-like effect. Conversely, increased ΔFosB levels in the OFC after social defeat promotes
resilience, revealing a differential role for ΔFosB in OFC and mPFC. Together,
these experiments complement our earlier demonstration that ΔFosB acting in
the nucleus accumbens reward circuitry mediates resilience and antidepressant
responses (Vialou et al., 2010), and will provide a better understanding of the role
of ΔFosB in the pathophysiology of depression and its treatment.
150
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kohala 4
Modulation of Adult Hippocampal Neurogenesis through HPA
Axis Activity determines the Divergent Effects of Distress and
Eustress on Affective Disorders
Michael L. Lehmann
National Institutes of Health
Background: In animals, complex environments and physical exercise have
been shown to reduce depressive behavior, increase hippocampal neurogenesis,
and improve cognitive function. Previously, we showed that environmental
enrichment facilitated the recovery from social defeat stress and enhanced stress
resiliency. In a social conflict paradigm, repeated social defeat stress consistently
yields a submissive phenotype and is an ethological means of inducing depression
in mice. Social defeat stress (SD) and enriched environments (EE) are both strong
modulators of hypothalamic-pituitary-adrenal (HPA) axis function. However, they
have divergent effects on the HPA axis. SD stress is a negative stressor (distress)
and has been shown to induce HPA axis dysfunction, whereas physical exercise
and enriched housing—both of which are positive stressors (eustress)—render
the HPA axis more adaptive. We hypothesized that the persistent arousal of HPA
activity by SD contributes to the pathogenesis of depression. We also hypothesized
that HPA activity during EE is requisite for the restoration of normal phenotype
in depressed mice. Because the hippocampus provides inhibitory control on
HPA axis output, and because hippocampal neurogenesis has been implicated
in the pathology of depression, we also hypothesized that divergent outcomes
of eustress and distress exposure are hippocampal neurogenesis-dependant. We
designed three experiments to test these hypotheses.
Methods: We first examined if adrenalectomy (ADX) coupled with basal
corticosterone (CORT) replacement would alter how animals respond to SD. We
exposed ADX and sham-operated male C57BL6/J mice to 14 days of chronic SD
and measured the expression of maladaptive behaviors. We also examined the
survival of newborn hippocampal neurons during SD. Next, we examined the
involvement of adrenal glucocorticoids in the restorative effects of environmental
enrichment (EE) in defeated mice. We hypothesized that HPA activity during
EE are required for behavioral recovery after defeat stress. Male mice were
exposed to SD for 2 weeks then either ADX (and basal CORT-replaced) or sham
151
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kohala 4
Modulation of Adult Hippocampal Neurogenesis through HPA
Axis Activity determines the Divergent Effects of Distress and
Eustress on Affective Disorders (continued)
Michael L. Lehmann
PL
operated. Mice were then housed in EE for 3 weeks and subsequently examined
for affective behavioral disorders. Lastly, we tested if decreased neurogenesis
contributes to the etiology of SDinduced depression or if it is downstream from
changes in behavior. We hypothesized that SD results in depressive-like behavior
through a glucocorticoid-induced decline in surviving hippocampal neurons
and the protective effects of ADX in distress would not persist in mice with
conditionally suppressed neurogenesis (NG-). Suppression is achieved through
the administration of ganciclovir to the diet of GFAP-HSVtk transgenic mice. NGmice and their wildtype (WT) littermate controls were ADX and exposed to
defeat stress as described in the first experiment.
Results: Mice ADX prior to social defeat showed decreased anxiety- and
depressive-like behaviors and increased survival of adult born hippocampal
neurons compared to sham-operated mice. During behavioral recovery after SD,
sham-operated mice housed in EE showed decreased expression of anxiety- and
depressive-like behaviors and increased survival of adult born neurons. However,
if we ADX defeated mice prior to EE, the beneficial effects of EE disappeared,
and hippocampal neurogenesis was decreased. Lastly, while ADX conferred
stress resiliency to the WT mice, ADX NG- mice lacked resiliency and developed
depressive behavior.
Discussion: The current experiments show a strong correlation between survival
of adult-born hippocampal neurons, adrenal corticosteroids, and expressions
of affective disorders. For instance, HPA axis stimulation during EE promotes
behavioral recovery in defeated mice and ADX prevents behavioral depression
from social defeat. Both manipulations promote the survival of adultborn
hippocampal neurons; HPA-activity during EE increases survival and ADX before
SD protects against decreased survival. Decreased hippocampal neurogenesis
and dysregulation of the HPA axis are implicated in the etiology of depression. The finding of the present study—demonstrating that ADX protects WT mice
from the detrimental effects of SD but did not protect mice with conditionally
152
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kohala 4
Modulation of Adult Hippocampal Neurogenesis through HPA
Axis Activity determines the Divergent Effects of Distress and
Eustress on Affective Disorders (continued)
Michael L. Lehmann
suppressed neurogenesis (NG-)— suggests that SD causes depressive behavior
through CORT-induced decreases in hippocampal neurogenesis. Because ADX
did not afford protection from defeat in NG- mice, this suggests that decreased
hippocampal neurogenesis—even in the absence of high-levels of CORT—is
sufficient to eliminate stress-resilience in mice.
PL
153
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Kohala 4
Optical Activation of Nucleus Accumbens Neurons modulates
Depression- and Anxiety-Like Behaviors
Mary Kay Lobo
University of Maryland School of Medicine
PL
Background: The nucleus accumbens (NAc) plays a crucial role in regulating
mood, including maladaptive behaviors seen in depression and anxiety disorders. Recently, deep brain stimulation (DBS) of the nucleus accumbens was shown to
alleviate depression and anxiety symptoms in patients suffering from treatment
resistant depression. Methods: We employ optogenetic technologies to apply repetitive (5 days)
high frequency (130Hz) and low frequency (10Hz) stimulation to NAc neurons,
using adeno-associated viruses (AAVs) expressing Channelrhopsin-2 (ChR2), in
animals exhibiting social avoidance (a depression-like behavior) after chronic
social defeat stress. Additionally, we optogenetically activate specific NAc
neuronal subtypes with a low frequency stimulation (10Hz) during acute moodrelated behaviors including tail suspension test (TST–a form of stress) and
elevated plus maze (EPM–a measure of anxiety).
Results: Repetitive high frequency, but not low frequency, ChR2 activation of
NAc neurons reversed the social avoidance seen after chronic social defeat stress. Furthermore, low frequency stimulation of dopamine receptor 1 (D1)-containing
NAc projection neurons during acute mood-related behaviors decreased time
spent immobile during the TST and increased time in the open arms in the EPM.
Discussion: These results provide insight into the mechanism of DBS stimulation
in the NAc, since directly activating NAc neurons repetitively with high frequency
stimulation can alleviate depression symptoms displayed after chronic social
defeat stress. Additionally, we show that activation of D1-containing NAc neurons
is important for mediating acute mood-related behaviors, since activation of these
neurons has antidepressant- and anxiolytic-like effects.
154
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Queen’s 5-6
Travel Awardee Research Presentations
Clinical and Translational Research
Chair: Scott Rauch
11:30 a.m.
12:10 p.m.
12:50 p.m.
Essential Role of Ventral Tegmental Area Dopamine Neurons in
Mediating the Induction and Rapid Reversal of Depression-Like
Behaviors
Ming-Hu Han
Evidence from Mouse and Man for a Role of Neuregulin 3 in
Nicotine Dependence
Jill R. Turner
Changes in Figural Memory Performance and fMRI Activity
across the Adult Age Span
Sharna Jamadar
PL
155
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Queen’s 5-6
Essential Role of Ventral Tegmental Area Dopamine Neurons in
Mediating the Induction and Rapid Reversal of Depression-Like
Behaviors
Ming-Hu Han
Mount Sinai School of Medicine
PL
Background: The surprising rapid clinical effect of deep brain stimulation in
depressed patients supports the notion that depression is a neural circuit disorder. In contrast, less is known if depression-like behaviors can be induced or reversed
rapidly in animal models. Our previous ex vivo and in vivo studies showed that
chronic social defeat consistently increased the firing rate and phasic firing events
of ventral tegmental area (VTA) dopamine (DA) neurons in the brain reward
circuitry of susceptible mice, but not of the resilient subgroup (Cell 2007; J
Neurosci 2010). However, the physiological function of this increased firing
and its ionic mechanisms, as well as potential therapeutic targets, remain to be
elucidated.
Methods: Susceptible and resilient mice were segregated following a chronic
(10-day) social defeat paradigm, a well-established model of depression. Utilizing
optogenetic techniques – Cre-dependent expression of channelrhodopsin-2
(ChR2) in tyrosine hydroxylase (TH)-Cre mice, phasic firing was induced by
high frequency optoactivation of ChR2 specifically in VTA DA neurons, and
behavioral tests (social interaction and sucrose preference) were performed in
behaving mice. The ionic mechanisms that underlie the hyperactivity of these
neurons were also examined in TH-GFP mice by use of electrophysiological
techniques.
Results: Consistent with our earlier findings, we found that optoactivation of
ChR2 (mimicking phasic firing) during sub-threshold defeat, a paradigm that does
not induce depressive-like behaviors in normal mice, increased social avoidance
behavior and reduced sucrose intake. Importantly, the same optoactivation of
these neurons during social interaction tests instantly induced similar susceptible
behaviors in both resilient mice and the mice treated with sub-threshold defeat. These findings indicate that the phasic firing of VTA DA neurons encodes, and is
tightly linked to, the susceptible phenotype. Next, toward understanding the ionic
mechanisms of the higher VTA DA neuron firing, we previously found that the
156
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Queen’s 5-6
Essential Role of Ventral Tegmental Area Dopamine Neurons in
Mediating the Induction and Rapid Reversal of Depression-Like
Behaviors (continued)
Ming-Hu Han
current of Ih (hyperpoplarization-activated cation channel), an important channel
in the transition between tonic andphasic firing patterns, was upregulated in
susceptible mice and that local infusion of Ih inhibitors ZD 7288 and DK-AH 269
into the VTA normalized depression-like social avoidance in susceptible mice
within one hour after the infusion. This rapid effect is very different from classic
antidepressants that take weeks to show treatment efficacy. Surprisingly, in our
ongoing studies, we also observed that the antidepressant effect induced by a
single-dose infusion of Ih inhibitor DK-AH 269 lasted at least two weeks. More
importantly, this long-lasting effect was also seen with a single intraperitoneal dose
of DK-AH 269. Following these studies, we also found that chronic defeat induced
a larger increase in Ih current in resilient mice than in the susceptible subgroup. This finding suggests that the force driving the pathological hyperactivity persists
in resilient mice, and that additional compensatory ionic mechanisms such as K+
channels are necessary to drive the higher firing back to normal levels in resilient
PL
mice. Consistently, our data showed that K+ currents were significantly increased
selectively in resilient mice, which is consistent with our earlier microarray data. In addition, we found that local infusion of the K+ (KCNQ) channel activator
flupirtine into the VTA had similar rapid antidepressant effects as seen with Ih
inhibitors. We are now investigating the effect of systemic administration of this
activator and others.
Discussion: Here we demonstrate that optogenetics is a powerful tool to
precisely imitate stress-induced physiological adaptations and reliably define
VTA DA neurons in the brain reward circuit as a therapeutic target. Interestingly,
optogenetically-induced phasic firing rapidly induces avoidance behavior during
interaction tests, even in resilient mice. Our data also strongly support that
Ih channels are one of the passive pathological ion mechanisms that underlie
the susceptible firing increase, while K+ channels are an important active ion
mechanism that drives the pathological hyperactivity back to normal levels in
resilient mice. Importantly, passive ion channel inhibitors and active ion channel
157
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Queen’s 5-6
Essential Role of Ventral Tegmental Area Dopamine Neurons in
Mediating the Induction and Rapid Reversal of Depression-Like
Behaviors (continued)
Ming-Hu Han
activators that inhibit the higher pathological firing of VTA DA neurons are
both antidepressant and pro-resilient. Our studies provide fundamentally novel
drug targets for rapid or long-lasting antidepressants, which are mechanistically
distinct from predominant monoamine-based medications. PL
158
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Queen’s 5-6
Evidence from Mouse and Man for a Role of Neuregulin 3 in
Nicotine Dependence
Jill R. Turner
University of Pennsylvania
Background: Smoking is the largest preventable cause of death and disease
in the United States, with about 46 million U.S. adults currently smoking [1]. Though there are effective medications approved by the FDA to treat nicotine
addiction, roughly 80% of smokers treated with these approaches relapse within
one year [2]. Interestingly, failed smoking cessation has been shown to have
genetic contributions [3-5]. One protein associated with mechanisms of both
gene regulation and nicotine response is the transcription factor CREB. To further
investigate mechanisms underlying nicotine dependence, the current study sought
to identify downstream targets of CREB and their regulation following treatment
with nicotine as well as during 24h withdrawal. Methods: Using functional genomic approaches (chromatin immunoprecipitation
(ChIP) and whole genome sequencing), CREB targets were identified following
chronic nicotine administration and withdrawal. GLITR (GLobal Identifier of
Target Regions) analysis was performed to assess potential target regions. Two
independent biological replicates were used to validate genomic targets using PL
ChIP, quantitative PCR and Western blotting. Validated genomic targets were then
assessed for single nucleotide polymorphisms (SNPs) in the clinical population.
Results: Chronic nicotine and withdrawal differentially modulates CREB binding
to the gene for Neuregulin 3 (NRG3). Quantitative PCR and Western blot analysis
of saline, nicotine, and nicotine withdrawal groups in two biological replicates
corroborate this finding, with NRG3 increases in both mRNA and protein following
nicotine treatment and withdrawal (p=0.008). Single nucleotide polymorphisms
(SNPs) across NRG3 were examined for association with prospective smoking
cessation among 595 smokers of European ancestry treated with transdermal
nicotine in two independent cohorts. Individual SNP and haplotype analysis
support association of NRG3 SNPs and smoking cessation success; however, the
function of these specific SNP markers in NRG3 is unknown. Discussion: These data suggest a role for NRG3 in nicotine dependence and
withdrawal. NRG3 is a neural-enriched member of the EGF family, and a specific
159
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Queen’s 5-6
Evidence from Mouse and Man for a Role of Neuregulin 3 in
Nicotine Dependence (continued)
Jill R. Turner
ligand for the receptor tyrosine kinase ErbB4 [6]. Of interest, genetic variation in
NRG3 has recently been implicated in risk susceptibility for schizophrenia [6-8]. Future studies in genetically modified mice for the NRG3 gene and its cognate
receptor, ERBB4, will investigate behavioral and molecular changes associated
with nicotine treatment and withdrawal. PL
160
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Queen’s 5-6
Changes in Figural Memory Performance and fMRI Activity
across the Adult Age Span
Sharna Jamadar
Olin Neuropsychiatry Research Center, Institute of Living
Background: Older adults perform worse than younger adults on tests of
recognition memory. This decreased performance is often accompanied by
changes in fMRI activity, both in task-related regions that are activated by young
participants, and in additional regions not activated in young participants. This
over-recruitment of neural activity has been attributed to either compensatory
processes or dedifferentiation of function. The vast majority of studies of
cognitive aging compare young (~20-30yrs) and old (~60-70yrs) participant
groups, however this artificial categorizing of age groups ignores the continuous
nature of age as a variable, and also can inflate estimates of age relations because
the variance associated with middle-age is ignored. In this study, we examined
changes in figural memory performance and fMRI activity in a large sample of
adults aged 17-81yrs. The figural memory task is a visual recognition task that
uses stimuli that are resistant to verbal encoding and has been developed for the
study of cognitive aging, however to date no fMRI study has been conducted with
the task, nor has changes in performance/neural activity been examined across PL
the adult age range. Methods: Two hundred and thirty five individuals aged 17-81yrs (mean 35yrs,
127 female) participated in the study. Ages 18-20 were oversampled, with ages
21-81 sampled with a frequency of around 25 per 10yr age bin. Task stimuli
were black line drawings that were resistant to verbal encoding presented in two
phases. In the Encoding phase, participants silently examined 20 Target stimuli. In the recognition phase, 20 Target and 20 Distractor stimuli were presented, and
participants pressed a button with either their index or middle finger to indicate if
they had seen it previously or not, respectively. Results: Reaction time (RT) increased linearly for Hit Targets (i.e. true positive
responses) with age; RT for Miss Target (false negative) and Hit Distractors (false
positive), Distractor error rate, and response bias showed a quadratic relationship
with age, such that RT increased until around 30yrs, plateaued, then decreased
around 60-yrs of age. So, the youngest and oldest participants were faster, made
161
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Travel Awardee Research Presentations
Queen’s 5-6
Changes in Figural Memory Performance and fMRI Activity
across the Adult Age Span (continued)
Sharna Jamadar
PL
fewer errors and showed a more conservative response bias than participants in the
median age ranges. During Encoding, participants activated a distributed bilateral
network encompassing frontal, parietal, lateral and medial temporal and occipital
cortices. fMRI activity during Encoding did not correlate with age. To determine
if Encoding differed between remembered vs. forgotten trials, we compared
Encoded-Remembered to Encoded-Forgotten trials. Encoded-Remembered trials
showed increased activity in bilateral occipital cortex and reduced activity in
precuneus, right inferior parietal and temporal cortex. This activity also did not
correlate with age. For Hit Targets, a distributed network encompassing premotor
(PM), subcortical, lateral and medial temporal (LatTEMP, MedTEMP), parietal
and occipital regions was activated. Activity in PM, cingulate and parahippocampal
gyrus (PHG) were linearly positively correlated with age; additional non-task
related activity in prefrontal (PFC), PM, parietal, LatTEMP, MedTEMP and
occipital regions was quadratically correlated with age. A similar network was
activated for Hit Distractors; activity in PFC, PM, putamen, PHG, parietal and
LatTEMP cortex were positively linearly correlated with age, additional nontask activity in PFC, PM, LatTEMP, MedTEMP and occipital regions were
quadratically related to age. Miss Targets activated a similar network to Hit
Targets. fMRI activity during Miss Targets did not correlate with age. Discussion: Behaviorally, younger and older participants were more likely to
be faster, make fewer errors and respond ‘no’ when uncertain. The changes in
performance in our youngest age range are likely to reflect continued maturation
of the brain that continues into the late 20s. Older adults showed greater activation
of task-related regions for Hit Targets and Distractors, and also showed differential
recruitment of additional non-task related brain regions. Together with the finding
that older participants performed better than middle aged participants on the task,
this pattern of fMRI activity suggests that older adults successfully compensated
for age-related changes in function of the brain.
162
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Faculty Research Fellowship Presentations
Kohala 3
Faculty Research Fellowship Presentations
Chair: Carrie Bearden
11:30 a.m. 12:10 p.m.
12:50 p.m.
Connecting NMDA Receptor signaling to Intellectual Disability
and Autism
Gavin Rumbaugh
Neuropsychological Tests as Predictors of Fear Conditioning and
Extinction
Karen G. Martinez
The Role for NDEL1 in nNOS Signaling: Implications for
Cortical Development and Prefrontal Cortex-mediated Cognitive
Behaviors
Atsushi Kamiya
PL
163
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Faculty Research Fellowship Presentations
Kohala 3
Connecting NMDA Receptor signaling to Intellectual Disability
and Autism
Gavin Rumbaugh
The Scripps Research Institute
PL
Background: Disruptions to the molecular mechanisms controlling glutamatergic
synapse structure and function are believed to underlie certain neurodevelopmental
disorders, such as Intellectual Disability (ID), Autism Spectrum Disorder (ASD)
and schizophrenia. Deleterious mutations in several synaptic proteins have
been liked to these disorders, though the impact of such mutations on neural
development and cognitive maturation is unclear. Recently, haploinsufficiency
of the gene that encodes the essential synaptic RasGAP, SynGAP, was shown
to cause ID in 3-4% of sampled patients. CNVs of this gene are linked to ASDs
and SynGAP expression levels are altered in elderly patients with schizophrenia. Because SynGAP is exclusively localized to forebrain dendritic spines, where it
directly regulates G-protein signaling controlling glutamatergic synapse structure
and function, the neurodevelopmental outcome of SynGAP1 mutations may
provide valuable insight into the patho-neurobiology underlying these disorders. Methods: The mice used for in this study were a mixed genetic background of
129sv/ev (Taconic) and c57/B6J (Jackson Labs). Electrophysiological studies
were carried out in mice at three different age groups: PND8-9, PND14-16,
PND>60. Two-photon imaging of dendritic spines in acute slices was performed
by exciting eGFP in SynGAP Het mice crossed to THY1-GFPm mice. Laser
photo-stimulation was performed by uncaging glutamate in the dentate gyrus and
then measuring spread of activity using a voltage sensitive dye. Dentate gyrus
function was assessed by mouse performance in a memory test designed to test
pattern separation.
Results: We show that excitatory synapses in the hippocampus of SynGAP1
haploinsufficient mice develop at an accelerated rate starting in the second
postnatal week. The changes in synaptic function were specific to glutamatergic
synapses and were caused by elevated AMPAR function. There were no observed
alterations to intrinsic neuronal properties or inhibitory currents onto glutamatergic
neurons. At this same point in development, SynGAP mutants exhibited enhanced
excitability of the hippocampal tri-synaptic circuit. Laser photo-stimulation of
164
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Faculty Research Fellowship Presentations
Kohala 3
Connecting NMDA Receptor signaling to Intellectual Disability
and Autism (continued)
Gavin Rumbaugh
the dentate gyrus resulted in a more than 8-fold increase in signals reaching CA1
from Hets compared to WT mice. In addition, SynGAP P16 SynGAP Hets also
exhibited an enhanced seizure threshold and audiogenic seizures, while also
exhibiting behavioral abnormalities related to hippocampal dysfunction. These
developmental disruptions had persistent consequences because adult mutants
performed poorly on a test for pattern separation, a memory test designed to
assess hippocampal function during memory encoding and retrieval.
Discussion: We conclude that SynGAP expression restricts the functional
maturation of glutamatergic synapses by tempering AMPAR accumulation at
postsynaptic sites. This negative tuning of glutamatergic synapses by SynGAP
appears essential for maintaining the balance of neural excitability in developing
hippocampal networks, which has implications for the organization of connected
brain regions and the emergence of cognitive ability. SynGAP tempers synaptic
function by down-regulating signals in dendritic spines that promote plasticity. Thus, these studies provide a neurobiological link connecting a disease-causing
genetic mutation to the abnormal development of neural circuits that underlie PL
human intellectual ability.
165
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Faculty Research Fellowship Presentations
Kohala 3
Neuropsychological Tests as Predictors of Fear Conditioning and
Extinction
Karen G. Martinez
University of Puerto Rico
PL
Background: Anxiety disorders are characterized by specific emotions, thoughts
and physiological responses. The psychological symptoms of anxiety disorders
are usually measured with self-report scales such as Beck Anxiety Inventory
(BAI) and the State-Trait Anxiety Inventory (STAI), which asses fear (thoughts
of threat, perception of physiological arousal) and anxiety (worry, avoidance,
muscle tension)[1]. Cognitive function, assessed with neuropsychological
tests like the Emotional Stroop, shows increased attention to threat in anxiety
patients[1]. Anxiety patients also show a characteristic psychological profile
consisting of high neuroticism and low extraversion[2]. Physiological responses
can be assessed at baseline, or in response to a sensory stimulus that has been
paired with a mild shock, a process known as fear conditioning. Little is known
about the relationship between psychological/cognitive symptoms of anxiety and
physiological responses. We studied this relationship in healthy subjects. Methods: Forty-seven healthy adults living in San Juan, Puerto Rico (31 female
and 16 male aged 21-54) were administered a series of tests that included the
NEO-PI personality inventory, State-Trait Anxiety Inventory (STAI), Beck
Anxiety Inventory (BAI), the Multi-Source Interference Task (MSIT, a counting
interference task), the Wisconsin Card Sorting Test (WCST) and an Emotional
Stroop Task (EST) which required subjects to identify the color of words that had
either neutral or emotional significance. All tests were administered in Spanish
and validated for Puerto Rico. Subjects were then trained in an established fear
conditioning and extinction paradigm (Milad et al., 2005), which assessed the
SCR to pictures of a colored light associated with shock (CS+, CS-), and to the
room in which the light CS was presented (context). On day 1, subjects received
habituation, conditioning and extinction trials. On day 2, subjects were tested for
memory of extinction (recall), and contextual renewal. The extent to which tests
predicted the physiological responses was assessed with individual regressions,
and then with predictive models using a combination of tests (multiple linear
regressions). Adjusted R2 were used to assess the predictive power of the models
outside of our own sample.
166
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Faculty Research Fellowship Presentations
Kohala 3
Neuropsychological Tests as Predictors of Fear Conditioning and
Extinction (continued)
Karen G. Martinez
Results: While individual tests showed poor prediction of fear learning and
extinction, a combination of tests yielded significant predictive models. We
were able to predict the response to the CS+ during conditioning and renewal
using combinations of the STAI, BAI, NEO-PI, MSIT, and sex (Cond:R2=0.304,
p<0.01; Renewal: R2=0.263, p<0.001). Thus, simple psychological tests were
sufficient to predict as much as 30% of the variance of the fear responses in our
sample. We were also able to explain the variance of baseline skin conductance
using extraversion scores.
Discussion: We assessed the relationship of several tests related to the anxiety
phenotypes with physiological indices, both at baseline and during various
phases of experimental fear conditioning. While we were unable to predict
extinction learning or extinction recall, we were able to predict 26-30% of SCR
variance in conditioning memory and renewal, both indicators of the strength
of conditioning. We also were able to explain 16% of the variance in baseline
SCL with extraversion alone. We developed a simple battery of psychological
tests capable of predicting approximately 30% of variance in experimental fear
conditioning and renewal. Predicting conditioned fear responses could help
identify people at risk for anxiety disorders and/or serve as a marker for relapse.
167
PL
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Faculty Research Fellowship Presentations
Kohala 3
The Role for NDEL1 in nNOS Signaling: Implications for
Cortical Development and Prefrontal Cortex-mediated
Cognitive Behaviors
Atsushi Kamiya
Johns Hopkins University School of Medicine
PL
Background: Higher brain function and behavior are influenced by neuronal
circuit formation during brain development. Many genetic risk factors for
schizophrenia, such as Disrupted-in-Schizophrenia-1 (DISC1) and neuronal nitric
oxide synthase (nNOS), have key roles in neurodevelopment. Consequently,
disturbances in brain development are suggested to underlie the pathology of
such devastating conditions. Although roles for these factors have been reported
at the molecular level, there are limited studies on whether they act in common
molecular pathways that contribute to disease pathology. In this study, we
explored the role of NudE-like 1 (NDEL1), a schizophrenia-associated protein
interactor of DISC1, in nNOS signaling for the development of the prefrontal
cortex and resultant behaviors. Given that nNOS and NDEL1 are highly expressed
in the cortical plate of developing cerebral cortex, NDEL1 may function as a
downstream effecter of nNOS signaling for cortical development, which may
also contribute to the NO-mediated establishment of neuronal circuits responsible
for long-lasting behaviors. Method: We examine the role of S-nitrosylation of NDEL1 via nNOS signaling
for cortical development and their underlying molecular mechanisms by using
cortical neuron cultures with RNAi approach and brains from nNOS KO mice. To manipulate NDEL1 function in the developing cerebral cortex, we use a Cre/
loxP-mediated inducible expression system with in utero electroporation. Result: We found that NDEL1 is S-nitrosylated in nNOS signaling. S-nitrosylation
of NDEL1 is required for dendritic development. We also found the interaction
of NDEL1 with nNOS, which is mediated by DISC1. Furthermore, our data from
the behavioral characterization of nNOS KO mice suggest that prefrontal cortexmediated cognitive functions are impaired in nNOS KO mice. Discussion: We are currently exploring the impact of nNOS/NDEL1 signaling in
vivo by in utero electroporation with an inducible gene expression system. With
this technique, we can dissect the temporal requirement for the studies of NDEL1
168
ACNP 50th Annual Meeting • Final Program
11:30 a.m. – 1:30 p.m.
Faculty Research Fellowship Presentations
Kohala 3
The Role for NDEL1 in nNOS Signaling: Implications for
Cortical Development and Prefrontal Cortex-mediated
Cognitive Behaviors (continued)
Atsushi Kamiya
in nNOS signaling in cortical development as well as explore the molecular basis
of disease animal models for further testing of resultant behaviors. Our results
will provide us with important clues for the possible involvement of nNOS/
NDEL1 signaling for cortical development and resultant behaviors, including
cognitive function. PL
169
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Issues in Ethics
Kona 5
Issues in Ethics
The Perils and Pitfalls of Biomedical Research: Historical and
Contemporary Perspectives on the Ethics of Research
Chair: Ellen Frank
Co-Chair: Jeffrey Lieberman
1:30 p.m.
Ports, Patches and Implants: The Ethics of Surgical Interventions to Modify the Brain
Arthur Caplan
2:15 p.m.
Animal Research in Neuropsychopharmacology: What are the Critical Ethical Issues?
David Jentsch
2:35 p.m.
The Process of Informed Consent: The Perspectives of a Clinical
Investigator on the Past and Future
Nina Schooler
PL
170
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Issues in Ethics Plenary
Kona 5
Ports, Patches and Implants: The Ethics of Surgical
Interventions to Modify the Brain
Arthur Caplan
University of Pennsylvania
This talk will examine emerging technologies to modify the brain, analyze some
of the key ethical challenges in monitoring brain activity, altering brain chemistry
and using electrical stimulation to modify behavior. The ethics of research will
be reviewed in this fast changing area including the acceptability of sham surgery,
consent from vulnerable subjects, levels of acceptable risk, working with IRBs
and conflicts of interest.
Currently, the Emmanuel and Robert Hart Director of the Center for Bioethics and
the Sidney D. Caplan Professor of Bioethics at the University of Pennsylvania
in Philadelphia, Art Caplan is the author or editor of thirty books and over 550
papers in refereed journals. His most recent books are Smart Mice Not So Smart
People (Rowman Littlefield, 2006) and the Penn Guide to Bioethics (Springer,
2009). He has served on a number of national and international committees
including as the Chair, National Cancer Institute Biobanking Ethics Working
Group; the Chair of the Advisory Committee to the United Nations on Human
Cloning; the Chair of the Advisory Committee to the Department of Health and
Human Services on Blood Safety and Availability; a member of the Presidential
Advisory Committee on Gulf War Illnesses; the special advisory committee to
the International Olympic Committee on genetics and gene therapy; the ethics
committee of the American Society of Gene Therapy and the special advisory
panel to the National Institutes of Mental Health on human experimentation on
vulnerable subjects. Most recently he was the Co-Director of the Joint Council
of Europe/United Nations Study on Trafficking in Organs and Body Parts and is
an advisor to DARPA on synthetic biology. Dr. Caplan is a member of the board
of directors of The Franklin Institute, the National Center for Policy Research on
Women and Families, the Iron Disorders Foundation and the National Hemophilia
Foundation’s Ethics Committee. He is on the Board of Visitors of the Columbia
University School of Nursing. Dr. Caplan is the recipient of many awards
and honors including the McGovern Medal of the American Medical Writers
171
PL
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Issues in Ethics Plenary
Kona 5
Ports, Patches and Implants: The Ethics of Surgical
Interventions to Modify the Brain (continued)
Arthur Caplan
Association and the Franklin Award from the City of Philadelphia. He received
the Patricia Price Browne Prize in Biomedical Ethics for 2011. He was a person
of the Year-2001 from USA Today. He was described as one of the ten most
influential people in science by Discover magazine in 2008. PL
172
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Issues in Ethics
Kona 5
Animal Research in Neuropsychopharmacology: What are the
Critical Ethical Issues?
David Jentsch
University of California, Los Angeles
Because of the way in which life on our planet originated, we share a tremendous
amount in common (genetically, physiologically, biochemically, etc.) with nonhuman animals, particularly other mammals. Consequently, we believe that - by
studying non human animals, we believe that we can better understand our own
brains and minds – how they operate both in health and in disease. On the other
hand, there appear to differences between humans and even our closest relatives
in the animal world, with ample evidence of very substantial gaps in morallyrelevant cognitive abilities, like theory of mind. A conflict has arisen between
those in support of and against animal use in biomedical research, and the
disagreement focuses on these two issues. Many scientists believe that biological
similarity makes animals useful models for investigating the building blocks
of our bodies, while the differences in cognition and socio-emotional function
justify their involuntary participation. Perhaps ironically, opponents of that
work often make precisely the opposite arguments, namely that animals are too
biologically different from us to be useful models and that their complex humanlike minds make their involvement in research heinous. In the resulting conflict –
which has escalated from name calling to criminally violent direct action against
researchers, key ethical issues are lost. In this presentation, I will address the
question of differences in moral agency in humans and animals and suggest that
embracing the concept that animal welfare is a responsibility of humans, rather
than a right of animals, and is key to successfully navigating the social milieu on
this complex issue.
J. David Jentsch received his Bachelor’s degree in behavioral biology from
The Johns Hopkins University (1992) and his PhD in neurobiology from Yale
University (1999). His graduate work, conducted under the supervision of Professor
Robert Roth, focused on characterizing the biochemical changes in prefrontal
cortical regions associated with prolonged experience with psychotomimetic and
stimulant drugs of abuse. After conducting post-doctoral training periods at the
173
PL
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Issues in Ethics
Kona 5
Animal Research in Neuropsychopharmacology: What are the
Critical Ethical Issues? (continued)
David Jentsch
University of Pittsburgh and Yale University, Dr. Jentsch was appointed as
an Assistant Professor at UCLA in 2001, where he is now a Professor in the
Departments of Psychology and Psychiatry; he is also Associate Director
for Research of the Brain Research Institute. His research focuses on genetic
and neurochemical mechanisms that influence cognition, impulse control and
decision-making in laboratory animals. In reaction to escalating extremism
amongst animal rights activists, which culminated in the firebombing of his car
in 2009, Dr. Jentsch formed the group Pro-test for Science. He and his colleagues
play a prominent role in scientific advocacy by coordinating the response of the
scientific community to attacks against researchers. In line with these efforts, Dr. Jentsch is a member of Board of Directors of the biomedical research advocacy
group: Americans for Medical Progress.
PL
174
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Issues in Ethics Plenary
Kona 5
The Process of Informed Consent: The Perspectives of a Clinical
Investigator on the Past and Future
Nina Schooler
State University of New York Downstate Medical Center
The belief that patients who will be the subjects of research should have a say in
the matter is a relatively recent concept. It has required biomedical researchers
to re-define their relationships with the people who will be the subjects of
research. As is suggested by the title of this symposium, that re-definition has
been challenging and has changed over time. The goal has been to insure that
research subjects, now often called research participants, understand the nature
of the research they are being asked to take part in, including risks and benefits,
give their consent freely and are able to exercise the right to withdraw that
consent should they change their minds. Rather than considering the social and
institutional structures that have developed to implement protection procedures,
e.g. Institutional Review Boards and their support system, I will focus on the
interaction between the research investigator and the person who is the potential
subject of research. I will first describe the implementation of procedures to
obtain informed consent and how this implementation has changed over time. I
will then examine the implications of the exclusion of individuals who do not
consent to participate on the results of clinical studies. This will be followed
by discussion of novel strategies to improve understanding of the risks and
benefits of potential research participation. The presentation will conclude with
recommendations for improvements to the process of informing people that leads
to enhanced understanding. The hope is that enhanced understanding can lead to
increased rates of participation and thus to clinical research that is more valid and
generalizable to the patent populations whose lives we hope to improve.
Dr. Schooler is Professor of Psychiatry and Behavioral Sciences at State University
of New York Downstate Medical Center, New York, NY; Adjunct Professor of
Psychiatry at the Georgetown University School of Medicine, Washington, DC;
Senior Research Psychologist in the Veterans Affairs VISN 5 Mental Illness
Research, Education, and Clinical Center; and Senior Research Scientist at The
Zucker Hillside Hospital in New York. In these capacities and settings, she conducts
175
PL
ACNP 50th Annual Meeting • Final Program
1:30 p.m. – 3:00 p.m.
Issues in Ethics Plenary
Kona 5
The Process of Informed Consent: The Perspectives of a Clinical
Investigator on the Past and Future (continued)
Nina Schooler
research on the treatment of schizophrenia and its long-term course. Prof. Schooler
received her Ph.D. in Social Psychology from Columbia University in New
York, and later served in leadership positions at the National Institute of Mental
Health, Bethesda, Maryland, where she led a series of significant multicenter
clinical trials of medication and psychosocial treatments for schizophrenia. She
subsequently joined the Department of Psychiatry at the University of Pittsburgh,
in Pennsylvania, where she directed the Psychosis Research Program and was
Professor of Psychiatry and Psychology. She then served as Director of Psychiatry
Research at the Zucker Hillside Hospital, conducting research in treatment of
first-episode psychosis and negative symptoms in schizophrenia until moving to
her present positions. PL
176
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session 1
Kona 4
Epigenetic Modifications in Development,
Aging and Mental Illness
Chair: Barbara Lipska
Co-Chair: Joel Kleinman
3:00 p.m.
Epigenetics and Gene Expression in the Human Brain
Mark Cookson
3:30 p.m.
DNA Methylation Changes in Development and Schizophrenia
Barbara Lipska
4:00 p.m.
Identifying Differentially Methylated Regions in Suicide Completers through Sequence Enrichment using MBD Protein and Next Generation Sequencing
Gustavo Turecki
4:30 p.m.
Maturation of Prefrontal Cortex in Health Disease: A Tail of Epigenomes in Transition
Schahram Akbarian
5:00 p.m.
Discussant: Joel Kleinman
PA
177
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session 2
Kona 5
Molecular Mechanisms Informing PTSD Risk,
Treatment and Prophylazis
Chair: Rachel Yehuda
Co-Chair: Eric Vermetten
3:00 p.m.
Molecular Mediators of Stress differentiate Resilience and Risk for PTSD in a Highly Traumatized Population
Kerry Ressler
3:30 p.m.
Cytosine Methylation and Expression of GR Related Genes in Association with PTSD Treatment Response
Rachel Yehuda
4:00 p.m.
Prospective Research in Military Cohorts: The Course of Stress-
Related Biological Parameters in Response to Exposure to a War Zone
Eric Vermetten
4:30 p.m.
High Dose Hydrocortisone Immediately after Trauma may alter the Trajectory of Posttraumatic Stress Disorder: Translational Interplay between Clinical and Animal Studies
Joseph Zohar
PA 5:00 p.m.
Discussant: Charles Marmar
178
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session 3
Kona 1-3
New Directions in Understanding the Neurocircuitry of Choice,
Value, and Decision-Making
Chair: Suzanne Haber
Co-Chair: Steven Grant
3:00 p.m.
Contrasting Reward Signals in Orbitofrontal Cortex and Anterior Cingulate Cortex
Jon Wallis
3:30 p.m.
The Neural Computation and Comparison of Values in Simple Choice
Antonio Rangel
4:00 p.m.
Human Ventral Striatal Neurons during a Gambling Task
Emad Eskandar
4:30 p.m.
Money, Value, and Motivation in Cocaine Addiction: Unique Roles of the vmPFC, ACC, Striatum and Midbrain
Rita Goldstein
5:00 p.m.
Discussant: Ben Greenberg
PA
179
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session 4
Kohala 3
A Convergence in Autism and Schizophrenia Genetics:
The Conundrum of Shared Risks and Divergent Outcomes
Chair: Matthew State
Co-Chair: Thomas Lehner
PA
3:00 p.m.
Rare CNVs Reveal Genetic Overlap between Autism, Schizophrenia and Bipolar Disorder
Jonathan Sebat
3:30 p.m.
Large-Scale Follow up of Candidate Variants from Sequencing Schizophrenia, Epilepsy and Autism Genomes
David Goldstein
4:00 p.m.
Findings from Number Variation and Whole Exome Sequencing in Autism Spectrum Disorders and the Overlap with Loci Implicated in Schizophrenia
Matthew State
4:30 p.m.
Connecting Genotype-Phenotype in Neurodevelopmental Disorders
Pat Levitt
5:00 p.m.
Discussant: Robert Ring
180
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session 5
Kohala 4
Neurodevelopmental Pathology of Cortical Interneurons in
Schizophrenia: Is it the Journey or the Destination that Matters?
Chair: Cynthia Weickert
3:00 p.m.
Factors determining Migratory Dynamics and Homing of Interneurons
Seong-Seng Tan
3:30 p.m.
Postnatal Interneuron Development: Setting the Cellular Stage for Schizophrenia Samantha Fung
4:00 p.m.
The Maturation of Neural Synchrony during Human Brain Development
Peter Uhlhaas
4:30 p.m.
Excitatory Projection Neuron Subtypes control the Distribution of Local Inhibitory Interneurons in the Cerebral Cortex
Paola Arlotta
5:00 p.m.
Discussant: Patricio O’Donnell
PA
181
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session 6
Monarchy
Will We have Drugs or Not? Addressing the Crisis in
Neuropsychiatric Drug Discovery
Chair: Eric Nestler
Co-Chair: David Michelson
3:00 p.m.
Better Novel CNS Target Validation for Drug Development is Feasible
William Potter
3:30 p.m.
Who will develop the Next Generation of Medications for Mental Illness? The NIMH Perspective
Thomas Insel
4:00 p.m.
Taking Science Personally: A Non-Profit Research Foundation’s Approach to Accelerating Therapeutic Development
Sohini Chowdury
4:30 p.m.
Of Lazarus and Zombies: Looking for Life after Death in Discontinued Compounds
David Michelson
5:00 p.m.
Discussant: Steven Hyman
PA
182
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session 7
Queen’s 5-6
Toward A Neuroimmune-Medicated Subtype of
Autism Spectrum Disorders
Chair: Christopher McDougle
3:00 p.m.
Clinical Evidence for an Immune-Mediated Form of Autism
Christopher McDougle
3:30 p.m.
Modeling an Autism Risk Factor in Mice leads to Permanent Changes in the Immune System
Elaine Hsiao
4:00 p.m.
Gene Expression Signatures in Autism Spectrum Disorders
Louis Kunkel
4:30 p.m.
Evidence for an Autoimmune Form of Autism
David Amaral
5:00 p.m.
Discussant: Susan Swedo
PA
183
ACNP 50th Annual Meeting • Final Program
Notes
184
Wednesday Morning At A Glance
7:00 am – 8:30 am
Women’s Task Force Meeting
Water’s Edge Boardroom
Panel Sessions
8:30 am – 11:00 am
Role of Phagocytes in Synaptic Plasticity and
Remodeling of Tissues in the Nervous System
8:30 am – 11:00 am
Neuroactive Cytokines: Critical Therapeutic
Targets for Depression and Treatment Resistant
Depression?
8:30 am – 11:00 am
Novel Approaches to Therapeutic Development
in Alzheimer’s Disease
8:30 am – 11:00 am
Translational Approaches to Understanding
Negative Symptoms
8:30 am – 11:00 am
The Development of Novel Pain Therapeutics:
New Strategies to Overcome Drug Discovery
Barriers
Kohala 3
Kona 1-3
Monarchy
Kona 4
Queen’s 5-6
8:30 am – 11:00 am
Novel Functions of Prefrontal Cortex Regions
in Motivated Behavior: Implication for Psychiatric
Disorders
8:30 am – 11:00 am
Progress in Understanding the Role of GABA and
GABAA Receptor Biology in Psychiatric Disease
11:15 am – 12:30 pm
ACNP Business Meeting
(ACNP Fellows, Members & Associate Members Only)
Kona 5
Kohala 4
Monarchy
Wednesday
PA-Panel MP-Mini Panel PL-Plenary SG-Study Group
Wednesday Afternoon At A Glance
12:30 pm – 2:00 pm
12:30 pm – 2:00 pm
12:30 pm – 2:00 pm
12:30 pm – 2:00 pm
SOBP Program Committee Meeting
Queen’s 4
Travel Awardee Luncheon
(by invitation only)
Water’s Edge Ballroom
SIRS Industry Task Force Meeting
Donatonio’s
ACNP Special Session: “Ask the Experts” Career
Development Program
Queen’s 5-6
Mini Panel Sessions
3:00 pm – 4:15 pm
4:15 pm – 5:30 pm
Downstream Effects of Visual and Auditory
Perceptual Impairment in Schizophrenia
GABA, Glutamate and Neural Synchrony
in Schizophrenia
Panel Sessions
3:00 pm – 5:30 pm
From Genome to Marco-Connectome: Integrating
High-Dimensional Genetic, Imaging and Behavioral
Data with Application to Large-Scale Studies of
Alzheimer’s
3:00 pm – 5:30 pm
Novel Synaptic Targets in Depression Emerging from
Clinical, Biochemical, and Circuit Based Approaches
3:00 pm – 5:30 pm
The Autism Sequencing Consortium (ASC):
Unraveling the Genetic and Functional Architecture
Of Autism Spectrum Disorders
3:00 pm – 5:30 pm
Neural Mechanisms of Environmental Risk for
Psychiatric Disorders
3:00 pm – 5:30 pm
Gimme Another Hit of Chocolate. Is Food Addictive?
3:00 pm – 5:30 pm
Drug of Abuse during Adolescence: A Development
Period of Vulnerability or Resilience?
Wednesday
5:30 pm – 7:30 pm
Poster Sessions III with Reception
7:30 pm – 9:00 pm
ACNP Special Session: An Oral History
of Neuropsychopharmacology
Kona 5
Kona 5
Kohala 4
Monarchy
Kona 1-3
Kona 4
Queen’s 5-6
Kohala 3
Kohala 1-2,
King’s & Grand Promenade
Monarchy
PA-Panel MP-Mini Panel PL-Plenary SG-Study Group
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kohala 3
Role of Phagocytes in Synaptic Plasticity and Remodeling of
Tissues in the Nervous System
Chair: Lei Yu
Co-Chair: Jonathan Pollock
8:30 a.m. Pruning CNS Synapses: Role of Microglia and the Complement
Cascade
Beth Stevens
9:00 a.m.
Engulfment and Elimination of Synpases by Astrocytes
Ben Barres
9:30 a.m.
In Vivo Studies of Microglial Function in Synaptic Plasticity
Wenbiao Gan
10:00 a.m.
Gene Targeting into the 21st Century: Mouse Models of Human
Disease from Cancer to Neuropsychiatric Disorders
Mario Capecchi
10:30 a.m.
Discussant: Jonathan Pollock
PA
185
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kona 1-3
Neuroactive Cytokines: Critical Therapeutic Targets for
Depression and Treatment Resistant Depression?
Chair: Husseini Manji
Co-Chair: Andrew Miller
8:30 a.m.
Cytokines-Neurochemiccals Interaction in Depression:
Biochemical, Genetic and Structural Aspects
Aye-Mu Myint
9:00 a.m.
Novel IL-1b Targets for Blockade of the Anti-Neurogenic and
Behavioral Actions of Stress
Ronald Duman
9:30 a.m.
The Role of TNFalpha in Synaptic Scaling
Robert Malenka
10:00 a.m.
Inflammation and Treatment Resistance in Major Depression: A Perfect Storm
Andrew Miller
10:30 a.m.
Discussant: Thomas Insel
PA
186
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Monarchy
Novel Approaches to Therapeutic Development
in Alzheimer Disease
Chair: Ralph Nixon
Co-Chair: Mary Sano
8:30 a.m.
Histone Acetyltransferase (HAT) Activators as Chromatin Remodelers in the Treatment of Alzheimer’s Disease
Ottavio Arancio
9:00 a.m.
Proinflammatory Cytokine Overproduction: A Contributor
to CNS Pathophysiology that is a Viable Target for Disease
Progression Modification
Linda van Eldik
9:30 a.m.
Apolipoprotein-E: An Obvious Target for Alzheimers Disease
Michael Vitek
10:00 a.m.
Targeting Neuronal Protein Indigestion as a Therapeutic
Approach for Alzheimer’s Disease
Ralph Nixon
10:30 a.m.
Discussant: Mary Sano
PA
187
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kona 4
Translational Approaches to Understanding Negative Symptoms
Chair: Stephen Marder
8:30 a.m. 9:00 a.m.
Facilitating Novel Treatment Development and Neurobiological
Research for Negative Symptoms: Findings from the
Collaboration to Advance Negative Symptom Assessment in
Schizophrenia (CANSAS)
William Horan
Neural Substrates of Emotion Processing and Expressivity
Deficits in Schizophrenia
Raquel Gur
9:30 a.m.
Emotion Experience in Schizophrenia: Timing Matters
Ann Kring
10:00 a.m.
Preclinical Studies Investigating the Neurobiological and Genetic
Underpinnings of Motivated Behavior
Jared Young
10:30 a.m.
Discussant: William Carpenter
PA
188
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Queen’s 5-6
The Development of Novel Pain Therapeutics: New Strategies to
Overcome Drug Discovery Barriers
Chair: Robert Lenox
Co-Chair: Frank Porreca
8:30 a.m. 9:00 a.m.
9:30 a.m.
10:00 a.m.
Identifying Mechanisms underlying Affective Components of
Pain and Pain Relief in Rodents to Promote Discovery of New
Therapies
Frank Porreca
Imaging Opioid Effects on the Brain – From Preclinical to
Postclinical
David Borsook
Neuroimaging as a Tool to Predict Analgesic Efficacy in Chronic
Pain Patients and Determine the Significance of Expectation in
Clinical Trial Design
Irene Tracey
Overcoming Scientific and Structural Barriers to Discovery of
Therapies for Pain
Chas Bountra
10:30 a.m.
Discussant: Robert Lenox
PA
189
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kona 5
Novel Functions of Prefrontal Cortex Regions in Motivated
Behavior: Implication for Psychiatric Disorders
Chair: Peter Kalivas
8:30 a.m. 9:00 a.m.
9:30 a.m.
10:00 a.m.
Effects of Cocaine use on the Role of Orbitofrontal Cortex in
Learning in Response to Violations in Reward Expectation
Geoffrey Schoenbaum
When the Prefrontal OFF Switch is Broken: How does
Extinction Occur?
Jamie Peters
Context-Induced Relapse to Heroin Seeking is Controlled by
Selectively Activated Neurons in Ventral but not Dorsal Medial
Prefrontal Cortex
Yavin Shaham
Cross-Cortical Phase Synchrony between the Medial Prefrontal
Cortex and Anterior Cingulate Cortex during Stimulus
Expectancy
Bita Moghaddam
10:30 a.m.
Discussant: Peter Kalivas
PA
190
ACNP 50th Annual Meeting • Final Program
8:30 a.m. – 11:00 a.m.
Panel Session
Kohala 4
Progress in Understanding the Role of GABA and GABAA
Receptor Biology in Psychiatric Disease
Chair: Nicholas Brandon
8:30 a.m. 9:00 a.m.
9:30 a.m.
GABA Signaling, Genetic Variation, Neurodevelopment, and the
Molecular Pathology of Schizophrenia
Thomas Hyde
Circuit-Specific Alterations in Mediators of Cortical GABA
Neurotransmission in Schizophrenia
David Lewis
Modifying GABAAR Clustering in the Prefrontal Cortex of
Mice induces Behavioral Deficits Reminiscent of Schizophrenia
Stephen Moss
10:00 a.m.
GABAergic Regulation of the HPA Axis in Depression
Donald Rainnie
10:30 a.m.
Discussant: Daniel Weinberger
PA
191
ACNP 50th Annual Meeting • Final Program
12:30 p.m. – 2:00 p.m.
Special Session
Queen’s 5-6
ACNP Special Session: “Ask the Experts”
Career Development Program
Chair: Marlene Freeman
Moderators: Linda Carpenter
Paul Holtzheimer
Panelists:
Pierre Blier
Linda Brady
Alan Breier
Wayne Drevets
John Krystal
Eric Nestler
Philip Ninan
David Rubinow
Carol Tamminga
Michael Thase
Mentorship-style advice and information will be provided to facilitate the
advancement of Associate Members, including tips on how to negotiate for faculty
positions and compare opportunities across institutions; suggestions on how to
progress from a K award to a R01 award; perspectives on obtaining foundation
grants and grants from industry sponsors; information about how to become a
more active member in ACNP, along with thoughts on achieving a manageable
balance of time and energy participating in organizations like ACNP as well
as other duties; reflections on hitting the right personal balance for attending
to demands of career versus the needs of partners/spouses and children. Data
from a variety of perspectives (basic and clinical scientists; men and women;
PhDs and MDs; individuals funded 100% for scientific work, and individuals
in administrative positions; individuals who have worked in different types of
institutional settings).
Representatives of the new Member Advisory Task Force Committee (Linda
PA Carpenter and Paul Holtzheimer) will act as moderators for this lively and
interactive session. A panel of “Experts” comprised of ACNP leaders and
successful senior scientists will be gathered to address critical topics of interest
to their junior colleagues in the College. Since the path to career success and
personal fulfillment may vary considerably from one individual to the next (based
on different backgrounds, circumstances, interests, skills and career goals), each
panelist will be asked to address questions or topics, posed by the moderators
192
ACNP 50th Annual Meeting • Final Program
12:30 p.m. – 2:00 p.m.
Special Session
Queen’s 5-6
ACNP Special Session: “Ask the Experts”
Career Development Program (continued)
during this program, from their own unique professional perspective. In addition
to describing their best and most timely mentoring pearls, panelists may be asked
to share lessons learned from their own (good or bad) past experiences with
career development challenges. Moderators will facilitate a discussion of why/
how some strategies offered by the panel may be particularly useful for some,
but not all, of our Associate Members, given certain professional and personal
considerations. Moderators will also facilitate the identification of career
development advice that achieves consensus endorsement by many or all of the
panel members. Associate members may send suggestions for names of expert
panel members, and for specific questions/topics for consideration, in advance
of the annual meeting. Invited panel members will be given a list of topics/
questions that reflect the needs/requests of the junior membership in advance
of the program, so they can prepare their thoughts. However, the format of this
program is informal, as panelists will not be asked to deliver slide presentations. PA
193
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 4:15 p.m.
Mini Panel Session
Kona 5
Downstream Effects of Visual and Auditory Perceptual
Impairment in Schizophrenia
Chair: Michael Green
3:00 p.m.
3:25 p.m.
3:50 p.m.
To Find the Stream Follow the Waves: Neurophysiological
Mechanisms of Downstream Dysfunction
Daniel Javitt
Effects of Visual Perceptual Organization Impairment of Later
Cognitive Processing in Schizophrenia
Steven Silverstein
Downstream Ripples of Impaired Perceptual Processing in
Schizophrenia
Michael Green
4:15 p.m. – 5:30 p.m.
Mini Panel Session
Kona 5
GABA, Glutamate and Neural Synchrony in Schizophrenia
Chair: Lawrence Kegeles
Co-Chair: Steven Siegel
MP
4:15 p.m.
Increasing Signal to Noise Ratio through Modulation of GABAB receptors
Steven Siegel
4:40 p.m.
Glutamatergic Dysfunction in Schizophrenia: A Chemical Shift Imaging and Single Voxel H-MRS Study
Juan Bustillo
5:05 p.m.
GABA and Glutamate-Glutamine in Schizophrenia Measured with Proton Magnetic Resonance Spectroscopy
Lawrence Kegeles
194
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session
Kohala 4
From Genome to Macro-Connectome: Integrating HighDimensional Genetic, Imaging and Behavioral Data, with
Application to Large-Scale Studies of Alzheimer’s Disease,
Schizophrenia and Substance Abuse
Chair: Vince Calhoun
Co-Chair: Godfrey Pearlson
3:00 p.m.
The Statistical Challenges of High Dimensional Neuroimaging and Genetic Data Analyses
Jean-Baptiste Poline
3:30 p.m.
New Findings in Schizophrenia via Robust Identification of Linked Genetics Factors and Functional Brain Regions within a Multivariate Framework
Vince Calhoun
4:00 p.m.
A Large Scale Multivariate Parallel ICA Method reveals Novel Imaging Genetic Relationships for Alzheimer’s Disease in the ADNI Cohort
Godfrey Pearlson
4:30 p.m.
Substance Use Disorders: Linking Genes, BOLD Response, and Clinical Phenotypes
Kent Hutchison
5:00 p.m.
Discussant: David Glahn
PA
195
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session
Monarchy
Novel Synaptic Targets in Depression Emerging from Clinical,
Biochemical, and Circuit Based Approaches
Chair: Lisa Monteggia
Co-Chair: Lois Winsky
3:00 p.m.
Is Synaptic Plasticity Involved in the Mechanism Underlying the
Rapid Antidepressant Effects of N-Methyl-D-Aspartate Receptor
Antagonists?
Carlos Zarate
3:30 p.m.
4:00 p.m.
NMDA Receptor Blockade at Rest Triggers Rapid Behavioural
Antidepressant Responses
Lisa Monteggia
Fast Optical Probing of Mechanisms underlying DepressionRelated Behaviors
Melissa Warden
4:30 p.m.
Synaptic Mechanisms in Models of Depression
Roberto Malinow
5:00 p.m.
Discussant: Charles Zorumsk
PA
196
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session
Kona 1-3
The Autism Sequencing Consortium (ASC): Unraveling the
Genetic and Functional Architecture of Autism Spectrum
Disorders
Chair: Thomas Lehner
Co-Chair: Matthew State
3:00 p.m.
Applying Biological Pathways to Next-Generation Sequence Data in Autism Spectrum Disorders
Joseph Buxbaum
3:30 p.m.
The Genetic Architecture of Autism Spectrum- and Related- Neurodevelopmental Disorders Revealed through High-
Resolution Genome Analysis
Stephen Scherer
4:00 p.m.
Examples of Recessive and Oligogenic Disease
Richard Gibbs
4:30 p.m.
Transcriptome and Genome Analysis of ASD
Daniel Geschwind
5:00 p.m.
Discussant: Thomas Insel
PA
197
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session
Kona 4
Neural Mechanisms of Environmental Risk for Psychiatric
Disorders
Chair: Anders Meyer-Lindenberg
Co-Chair: Charles Nemeroff
3:00 p.m.
Risk, Resilience and Gene-Environment Interplay in Primates
Stephen Suomi
3:30 p.m.
Neurobiology of Gene-Environment Interactions in Mediating Child Abuse Associated Risk for Mood and Anxiety Disorders
Charles Nemeroff
4:00 p.m.
How does Cannabis Increase Risk of Schizophrenia?
Robin Murray
4:30 p.m.
Neural Mechanisms for Environmental Risk Related to Urbanicity and Migration
Andreas Meyer-Lindenberg
5:00 p.m.
Discussant: Daniel Weinberger
PA
198
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session
Queen’s 5-6
Gimme Another Hit of Chocolate. Is Food Addictive?
Chair: Walter Kaye
Co-Chair: Guido Frank
3:00 p.m.
Neurobiology of Compulsive Eating: Role for Striatal Dopamine D2 Receptors
Paul Kenny
3:30 p.m.
Overeating of Sugars and Fats: Links to Addiction and Obesity
Nicole Avena
4:00 p.m.
Excessive Over- and Under- Eating Differentially Determine Brain Reward Learning in Humans
Guido Frank
4:30 p.m.
Is Food Restriction in Anorexia Nervosa caused by Reduced Reward and/or Increased Inhibition?
Walter Kaye
5:00 p.m.
Discussant: Kent Berridge
PA
199
ACNP 50th Annual Meeting • Final Program
3:00 p.m. – 5:30 p.m.
Panel Session
Kohala 3
Drug of Abuse during Adolescence: A Developmental Period of
Vulnerability or Resilience?
Chair: Susan Anderson
Co-Chair: Patricio O’Donnell
3:00 p.m.
The Relationship between Substance Use and Brain Development in Human Adolescents: Insights from Neuroimaging
Adriana Galvan
3:30 p.m.
Adolescence is a Period of High Risk for Addiction: The Role of Prefrontal Dopamine System and Cocaine Cues in Rats
Susan Anderson
4:00 p.m.
Mechanisms of Adolescent (in) Vulnerability
Kyle Frantz
4:30 p.m.
Unique Effects of Nicotine on Adolescent Limbic System Function
Frances Leslie
5:00 p.m.
Discussant: Patricio O’Donnell
PA
200
ACNP 50th Annual Meeting • Final Program
7:30 p.m. – 9:00 p.m.
Special Session
Monarchy
An Oral History of Neuropsychopharmacology
Chair: Samuel Gershon
Co-Chair: Martin M. Katz
7:30 p.m.
Volume 1: Starting Up (ed. Edward Shorter)
Volume 2: Neurophysiology (ed. Max Fink)
Edward Shorter
7:40 p.m.
Volume 3: Neuropharmacology, The Neurotransmitter Era
Fridolin Sulser (ed.)
7:50 p.m.
Volume 4: Psychopharmacology (ed. Jerome Levine)
Donald Klein
8:00 p.m.
Volume 5: Neuropsychopharmacology (ed. Samuel Gershon)
David Janowsky
8:10 p.m.
Volume 6: Addiction
Herbert D. Kleber (ed.)
8:20 p.m.
Volume 7: Special Areas (ed. Barry Blackwell)
Volume 8: Diverse Topics (ed. Carl Salzman)
Volume 9: Update (ed. Barry Blackwell)
Carl Salzman
8:30 p.m.
Volume 10: History of the ACNP
Martin M. Katz (ed.), John Davis
8:45 p.m.
Discussant
Thomas A. Ban (Series ed.)
201
PA
ACNP 50th Annual Meeting • Final Program
Notes
202
7:00 am – 8:00 am
ACNP/AsCNP/CINP/ECNP/JSNP Meeting
Queen’s 4
Mini Panel Sessions
8:00 am – 9:15 am
The Use of Intraoperative Techniques to Assess
the Physiology of the Anterior Cingulate Cortex
Kona 5
9:15 am – 10:30 am
Genes, Fear and Anxiety: From Mice to Humans
Kona 5
Panel Sessions
8:00 am – 10:30 am
Glutamate Targets for CNS Therapy: Insights
Obtained from a Potential Dynamic Duo
8:00 am – 10:30 am
Beyond Geonome – Wide Association Studies:
New Approaches to Risk of Psychiatric Illness
8:00 am – 10:30 am
Rapid Acting Antidepressants increase
Synaptogenesis
8:00 am – 10:30 am
The Putative Role of ER Stress in Neuropsychiatric
Illness
8:00 am – 10:30 am
Sex Difference in Brain and Behavior: Emerging
Genetic and Cellular Mechanisms
Kohala 3
8:00 am – 10:30 am
Serotonin Signaling during Development:
Unexpected Sources, Large Neuron Heterogeneity,
Limited System Plasticity and Big Impact on
Physiology and Behavior
Kohala 4
9:00 am – 12:00 pm
ACNP Council Meeting
Monarchy
Kona 1-3
Kona 4
Queen’s 5-6
Queen’s 4
PA-Panel MP-Mini Panel PL-Plenary SG-Study Group
Thursday
Thursday Morning At A Glance
Thursday
Thursday Afternoon At A Glance
Panel Sessions
12:00 pm – 2:30 pm
APOE and Alzheimer’s Disease:
Neurosusceptibility, Neuroprotection and New
Treatments
12:00 pm – 2:30 pm
From Transcription to Oscillations: How Sick
Interneurons create a Schizophrenia-Like
Phenotype
Monarchy
Kona 1-3
12:00 pm – 2:30 pm
Contribution of Genetic Epidemiology to
Identifying Genetic and Environmental Risk
Factors for Neurologic and Psychiatric Disorders
Kona 4
12:00 pm – 2:30 pm
Is Love Epigenetic? Transformative Effects of
Social Experience and of Oxytocin
Kona 5
12:00 pm – 2:30 pm
Enhancing Cognitive Performance: Molecular,
Pharmacological, and Experiential Strategies
Queen’s 5-6
12:00 pm – 2:30 pm
Functional Connectivity in Neural Systems as a
Developmental Abnormality in Creating Risk for
Bipolar Disorder
Kohala 2
12:00 pm – 2:30 pm
Optogenetic Dissection of Cortico-Limbic Circuit
Function and Dysfunction
Kohala 3
12:00 pm – 2:30 pm
Translating Pharmacogenetics into Clinical Utility:
Optimizing the Phenotype Kohala 4
PA-Panel MP-Mini Panel PL-Plenary SG-Study Group
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 9:15 a.m.
Mini Panel Session
Kona 5
The Use of Intraoperative Techniques to Assess the Physiology of
the Anterior Cingulate Cortex
Chair: Darin Dougherty
8:00 a.m. Boundaries of Anterior Cingulate Cortex and Midcingulate Concept
Brent Vogt
8:25 a.m.
Intraoperative Physiologic Evidence of Anterior Cingulate Cortex Modulation of Autonomic Arousal and Neuroimaging Correlations
Andre Gentil
8:50 a.m.
Human Anterior Dorsal Cingulate Neuronal Activity during a Stropp Interference Task
Emad Eskandar
MP
203
ACNP 50th Annual Meeting • Final Program
9:15 a.m. – 10:30 a.m.
Mini Panel Session
Kona 5
Genes, Fear and Anxiety: From Mice to Humans
Chair: John Neumaier
Co-Chair: Larry Zweifel
9:15 a.m. Genetic Factors driving Corticolimbic Mediation of Fear in Mouse Models
Andrew Holmes
9:40 a.m.
Genetic Dissection of the Role of Phase Dopamine in Fear Processing and Generalized Anxiety
Larry Zweifel
10:05 a.m.
Neural Correlates of Impaired Fear Inhibition and Extinction in PTSD
Tanja Jovanovic
MP
204
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 10:30 a.m.
Panel Session
Monarchy
Glutamate Targets for CNS Therapy: Insights Obtained from a
Potential Dynamic Duo
Chair: Dean Wong
Co-Chair: Rikki Waterhouse
8:00 a.m. Glycine Transporter-1 and Metabotropic Glutamate Receptor mGluR5 as Potential Therapeutic Targets for Schizophrenia
Kenji Hashimoto
8:30 a.m.
Modulation of Glutamatergic and Dopaminergic Transmission by RG1678, a Novel and Potent Glycine Reuptake Inhibitor
Daniela Alberati
9:00 a.m.
Imaging Biomarkers for Glutamate: Focus on mGluR5glyT
Dean Wong
9:30 a.m.
Pharmacological Strategies for NMDAR Enhancement
Daniel Javitt
10:00 a.m.
Discussant: Darryle Schoepp
PA
205
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 10:30 a.m.
Panel Session
Kona 1-3
Beyond Genome-Wide Association Studies: New Approaches to
Risk of Psychiatric Illness
Chair: Robert Freedman
8:00 a.m. De Novo Copy Number Variants Confer Risk for Bipolar Disorder, Schizophrenia and Autism
Jonathan Sebat
8:30 a.m.
Genomic Studies of Rare and Common Variation in Schizophrenia and Bipolar Disorder
Shaun Purcell
9:00 a.m.
Brain eQTLs and Function-based GWAS of Bipolar Disorder identified Novel Disease Risk Gene
Chunyu Liu
9:30 a.m.
Biologic and Epidemic Approaches to Risk of Psychiatric Illness
Elliot Gershon
10:00 a.m.
Discussant: Joel Kleinman
PA
206
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 10:30 a.m.
Panel Session
Kona 4
Rapid Acting Antidepressants increase Synaptogenesis
Chair: Ronald Duman
Co-Chair: Wayne Drevets
8:00 a.m. Scopolamine Produces a Rapid Antidepressant Response: Comparison with Ketamine
Wayne Drevets
8:30 a.m.
Rapid-Acting Antidepressants Require mTOR Signaling and Synaptic Protein Synthesis
Ronad Duman
9:00 a.m.
Synaptogenesis and Rapidly Acting Antidepressants: Comparison
between Ketamine and Scopolomine
George Aghajanian
9:30 a.m.
Aging alters Stress-Induced Structural Plasticity and Recovery in Medial Prefrontal Cortex
John Morrison
10:00 a.m.
Discussant: Husseini Manji
PA
207
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 10:30 a.m.
Panel Session
Queen’s 5-6
The Putative Role of ER Stress in Neuropsychiatric Illness
Chair: David Bredt
Co-Chair: Guang Chen
8:00 a.m. Impact of Endoplasmic Reticulum (ER) Signaling on Neuroplasticity and Neuronal Survival
Michael Jackson
8:30 a.m.
A Possible Role of XBP1 in Neural Pasticity
Takaoki Kasahara
9:00 a.m.
Oxidative Damage to Biomolecules as a Potential Therapeutic Target for Bipolar Disorder
L. Trevor Young
9:30 a.m.
Roles of ER Stress Modluators, Bcl-2 and BI-1, in Stress Coping and Action of Antidepressant
Guang Chen
10:00 a.m.
Discussant: De-Maw Chuang
PA
208
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 10:30 a.m.
Panel Session
Kohala 3
Sex Differences in Brain and Behavior: Emerging Genetic and
Cellular Mechanisms
Chair: Rita Valentino
Co-Chair: C. Neill Epperson
8:00 a.m. Parent-of-Origin Effects in the Male and Female Mouse Brain
Christopher Gregg
8:30 a.m.
Sex Chromosome Genes and Hormones Interact to Mediate Behavior
Emilie Rissman
9:00 a.m.
Sex Differences in Stress Responses: From Molecules to Mood
Debra Bangasser
9:30 a.m.
Sex-Specific Signaling Mechanisms in Schizophrenia
Eugenia Gurevich
10:00 a.m.
Discussant: C. Epperson
PA
209
ACNP 50th Annual Meeting • Final Program
8:00 a.m. – 10:30 a.m.
Panel Session
Kohala 4
Serotonin Signaling during Development: Unexpected Sources,
Large Neuron Heterogeneity, Limited System Plasticity and Big
Impact on Physiology and Behavior
Chair: Sheryl Beck
Co-Chair: Mark Ansorge
8:00 a.m. Developmental and Physiological Properties of Raphe Neuron Subpopulations Sheryl Beck
8:30 a.m.
The Placenta, Serotonin and Developmental Programming
Pat Levitt
9:00 a.m.
Redefining Brain Serotonergic Neurons by Genetic Lineage and Selective in Vivo Silencing
Russell Ray
9:30 a.m.
Serotonin Signaling during Development – Impact on Raphe Function, Limbic Circuitry and Behavior
Mark Ansorge
10:00 a.m.
Discussant: Jay Gingrich
PA
210
ACNP 50th Annual Meeting • Final Program
12:00 p.m. – 2:30 p.m.
Panel Session
Monarchy
APOE and Alzheimer’s Disease: Neurosusceptibility,
Neuroprotection and New Treatments
Chair: Terry Goldberg
Co-Chair: Steven Paul
12:00 p.m.
Brain Imaging, Genomics, and the Prevention of Alzheimer’s Disease
Eric Reiman
12:30 p.m.
APOE2 and Neuroprotective Responses: Molecular, Biomarker, and Cognitive Findings
Terry Goldberg
1:00 p.m.
Apolipoprotein E4: A Causative Factor and Therapeutic Target in Neuropathology, including Alzheimer’s Disease
Robert Mahley
1:30 p.m.
ApoE and the Molecular Pathogenesis of Alzheimer’s Disease: Therapeutic Implications
Steven Paul
2:00 p.m.
Discussant: Floyd Bloom
PA
211
ACNP 50th Annual Meeting • Final Program
12:00 p.m. – 2:30 p.m.
Panel Session
Kona 1-3
From Transcription to Oscillations: How Sick Interneurons
create a Schizophrenia-Like Phenotype
Chair: James Meador-Woodruff
Co-Chair: Rita Cowell
12:00 p.m.
A Critical Role for PGC-1a in the Transcriptional Control of Parvalbumin-Positive Interneuron Function
Rita Cowell
12:30 p.m.
What’s Wrong with Cortical Disinhibition? Exploring the Role of GABAergic Interneuron Dysfunction in Distinct Neuropsychiatric Disorder-Like Phenotypes
Kazu Nakazawa
1:00 p.m.
Recurrent Excitation-Inhibition in Local Cortical Circuits: Synaptic Properties Relevant for Gamma Oscillations
Guillermo Gonzalez-Burgos
1:30 p.m.
Cell Type Selective Reduction in NMDAR1 leads to Cognitive and Negative Symptom-like Deficits
Steven Siegel
2:00 p.m.
Discussant: David Lewis
PA
212
ACNP 50th Annual Meeting • Final Program
12:00 p.m. – 2:30 p.m.
Panel Session
Kona 4
Contribution of Genetic Epidemiology to Identifying Genetic
and Environmental Risk Factors for Neurologic and Psychiatric
Disorders
Chair: Kathleen Merikangas
Co-Chair: Emmanuel Mignot
12:00 p.m. Sources of Heterogeneity of Migraine: Longitudinal Stability and
Comorbidity with Mood Disorders
Kathleen Merikangas
12:30 p.m.
Genetic Studies of Schizophrenia in Sweden: Population-Based Samples, Discordant Mono-Zygotic Twins and Co-Morbidity with Bipolar Disorder and Infantile Autism
Christina Hultman
1:00 p.m.
Genetic Heritability, Shared Environmental Factors, and Sex Differences among Twin Pairs with Autism
Neil Risch
1:30 p.m.
Interaction between Genetic Susceptibility and Infections in the Etiology of Narcolepsy
Emmanuel Mignot
2:00 p.m.
Discussant: Robert Freedman
PA
213
ACNP 50th Annual Meeting • Final Program
12:00 p.m. – 2:30 p.m.
Panel Session
Kona 5
Is Love Epigenetic? Transformative Effects of Social
Experiences and of Oxytocin
Chair: James Harris
Co-Chair: James Leckman
12:00 p.m.
Social Monogamy as a Model for Love: Does Oxytocin Explain the Protective Effects of Love?
Sue Carter
12:30 p.m.
The Epigenetics of Social Behavior and the Oxytocin Receptor
Jessica Connelly
1:00 p.m.
Epigenetic and Transgenerational Transmission of Individual Differences in Maternal Behaviour: Role of Estrogen Receptor Alpha, BDNF and Oxytocin – Dopamine Interactions in Regulation of Maternal Mood
Michael Merzenich
1:30 p.m.
The Cross Generation Transmission of Social Affiliation in Humans: Oxytocin, Brain, and Interactive Synchrony
Ruth Feldman
2:00 p.m.
Discussant: Stephen Porges
PA
214
ACNP 50th Annual Meeting • Final Program
12:00 p.m. – 2:30 p.m.
Panel Session
Queen’s 5-6
Enhancing Cognitive Performance: Molecular, Pharmacological
and Experimental Strategies
Chair: Robert Bilder
12:00 p.m.
Cognitive Enhancement in Animal Models of Neuropsychiatric Disorder
Trevor Robbins
12:30 p.m.
The Role of Insulin-Like Growth Factors and Insulin in Memory Enhancement
Cristina Alberini
1:00 p.m.
Brain Plasticity Perspective about the Origin & Treatment of Psychiatric Illness
Michael Merzenich
1:30 p.m.
Cognitive Enhancing Drugs and Society
Barbara Sahakian
2:00 p.m.
Discussant: Robert Bilder
PA
215
ACNP 50th Annual Meeting • Final Program
12:00 p.m. – 2:30 p.m.
Panel Session
Kohala 2
Functional Connectivity in Neural Systems as a Developmental
Abnormality in Creating Risk for Bipolar Disorder
Chair: Kiki Chang
12:00 p.m.
Anterior Limbic Abnormalities in Youth with Bipolar Parent
Melissa DelBello
12:30 p.m.
Abnormal Structural and Functional Integrity of Emotion Regulation Neural Circuitry differentiate Healthy Adolescents at High Risk for Mood Disorders from Healthy Low-Risk Adolescents
Mary Phillips
1:00 p.m.
Brian Activation Predicts Clinical Change following Family Focused Therapy in Youth at High-Risk for Bipolar Disorder
Amy Garrett
1:30 p.m.
Functional Connectivity Abnormalities in Youth at High-Risk for Bipolar Disorder
Kiki Chang
2:00 p.m.
Discussant: Ellen Leibenluft
PA
216
ACNP 50th Annual Meeting • Final Program
12:00 p.m. – 2:30 p.m.
Panel Session
Kohala 3
Optogenetic Dissection of Cortico-Limbic Circuit Function and
Dysfunction
Chair: Lorna Role
12:00 p.m.
Optogenetic Probe of the Role of Cholinergic Neurons in Cocaine Conditioning
Karl Deisseroth
12:30 p.m.
Optogenetic Tuning of Cholinergic Inputs to Basolateral Amygdala
Lorna Role
1:00 p.m.
Optogenetic Dissection of Development and Function of Newborn Neurons in the Adult Brain
Shaoyu Ge
1:30 p.m.
Optogenetic Manipulation of Cortical Activity alters Behavioral Flexibility in Making and Breaking Habits
Ann Graybiel
2:00 p.m.
Discussant: Eric Nestler
PA
217
ACNP 50th Annual Meeting • Final Program
12:00 p.m. – 2:30 p.m.
Panel Session
Kohala 4
Translating Pharmacogenetics into Clinical Utility:
Optimizing the Phenotype
Chair: Thomas Schulze
Co-Chair: Anil Malhotra
12:00 p.m.
Common and Rare Variation in the POMC Pathway Contributes to Antipsychotic Drug-Induced Weight Gain
Anil Malhotra
12:30 p.m.
Genetic Variation in CYP450s Impacts Clearance of Antipsychotics
Kristin Bigos
1:00 p.m.
Rare Outcomes and Rare Alleles in Treatment-Resistant Depression
Gonzalo Laje
1:30 p.m.
The Consortium on Lithium Genetics (ConLiGen): Phenotypic Characterization and Genome-Wide Association Study of Lithium Response
Thomas Schulze
2:00 p.m.
Discussant: Donald Goff
PA
218
ACNP 50th Annual Meeting • Final Program
Notes
219
ACNP 50th Annual Meeting • Final Program
Notes
220
Posters
Posters
Notes
ACNP 50th Annual Meeting • Final Program
Poster Session I – Monday
1.
Psychostimulant Treatment of Age-related Cognitive Decline: Attention
Enhancing Effects of Methylphenidate are Reduced in Senescent Rats
Barry D. Waterhouse, Richard Chu, Jed S. Shumsky, Shevon E. Nicholson
2.
Using Translatable Human Biomarkers to Assess Clinical Relevance of
Mouse Models of Obsessive Compulsive Disorder
Susanne E. Ahmari, Victoria B. Risbrough, Lauren Leotti, Cara
Malapani, Edward E. Smith, Mark A. Geyer, Rene Hen, H. Blair
Simpson
3.
Repeated Psychological Trauma causes Enduring Corticotropin-Releasing
Factor-Dependent Sensitization of Basolateral Amygdala Noradrenergic
Systems: A Substrate for PTSD-like Startle and Sensorimotor Gating
Abnormalities?
Vaishali P. Bakshi, Abha K. Rajbhandari
4.
Rapid Tryptophan Depletion Moja-De decreases Serotonergic Function
and influences Anxiety-Like Behavior in a Strain-Selective Way in Mice
Caroline Sarah Biskup, Andrew Arrant, Amanda Day, Cynthia Kuhn,
Florian Daniel Zepf
5.
MicroRNA Regulation of Fear Extinction Memory
Timothy W. Bredy
6.
Antagonism of Kappa-Opioid Receptors reduces Stress-Induced Effects
Ashlee Van’t Veer, F. Ivy Carroll, William A. Carlezon
221
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
7.
Epigenetic Differences in the Developing Hippocampus and Amygdala in
a Novel Rat Model of Anxiety and Depression
Sarah M. Clinton, Rebecca K. Simmons, Danielle N. Simpson,
Stanley J. Watson, Huda Akil
8.
Fear Conditioning in Rat Pups and Fear Reactions Acquired from
Exposure to Fearful Mothers involve Similar Brain Structures
Jacek Debiec, Regina Sullivan
9.
Essential Role for Orbitofrontal 5-HT1B Receptors in OCD-Like Behavior
and SRI Response in Mice
Nancy A. Shanahan, Lady P. Velez, Virginia L. Masten, Stephanie C. Dulawa
10. Exposure to Traumatic Stress in Rats Differentially Affects AlcoholRelated Behaviors and Brain ERK Phosphorylation
Nicholas W. Gilpin, Scott Edwards, George F. Koob
11. Norepinephrine Transporter A457P Knockin Mice display Anxiety-Like
Behavior and Tachycardia
Jana K. Shirey-Rice, Rebecca Klar, Sarah N. Redmon, Jessica J. Krueger, Nathan M. Wallace, Martin Appasamy, Charlene Finney,
Suzanna Lonce, Andre Diedrich, David H. Robertson, Maureen K. Hahn
12. Altered Noradrenergic Activity in an Animal Model of Post-Traumatic
Stress Disorder
Sophie A. George, Dayan Knox, Andre L. Curtis, Rita J. Valentino,
Israel Liberzon
222
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
13. Analysis of Norepinephrine Dynamics within the Bed Nucleus of the Stria
Terminalis in Rat Models of Substance Abuse and Post-Traumatic Stress
Disorder
Zoe A. McElligott, Paul L. Walsh, R. Mark Wightman
14. Reelin mediates the Neurosteroid-Induced Long-Lasting Improvement of
Aggression and Anxiety-Like Behavior in a Mouse Model of PTSD
Mauricio S. Nin, Luis A. Martinez, Marianela Nelson, Graziano
Pinna
15. Differential Role of ΔFosB in the Prefrontal Cortex in CCK Sensitivity
and Vulnerability to Stress
Vincent Vialou, Deveroux Ferguson, Eric J. Nestler
16. Manipulating Brain pH alters Fear Memory Consolidation via AcidSensing Ion Channel-1a
John Wemmie
17. Role of Serotonin 2A Receptor Signaling during Development in
Modulating Adult Affective Behavior
Elena Y. Demireva, Qinghui Yu, Jeff M. Muller, Mark Ansorge, Jay
A. Gingrich
18. The Role of Cortical Norepinephrine in the Development of Executive
Function
Kara L. Agster, Andrew T. Bates, Rachel E. Cain, Lori A. Newman,
Barry D. Waterhouse, Jill A. McGaughy
223
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
19. Open Board
20. Elevated Glutamate Levels in the Striatum, Nucleus Accumbens Core and
Prefrontal Cortex of the Spontaneously Hypertensive Rat Model of ADHD
Paul E.A. Glaser, Erin M. Miller, Greg A. Gerhardt
21. Haploinsufficiency of the Intellectual Disability and Autism Susceptibility
Gene, SynGAP1, causes Premature Excitatory Synapse Development and
Neonatal Hippocampal Dysfunction
Gavin Rumbaugh, James Chelliah, Max Aceti
22. Neurogenesis is not Required for Lithium’s Effect in the Forced Swim
Test suggesting that Lithium’s Antidepressant-Like Effect is not Mediated
via GSK-3 Inhibition
Galila Agam, Nirit Kara, Sapna Narayan, Haim Einat, Vidita Vaidya,
R.H. Belmaker
23. HDAC6 Regulates GR Signaling in Serotonin Pathways with Critical
Impact on Stress Resilience
Julie Espallergues, Sarah L. Teegarden, Sheryl G. Beck, Olivier
Berton
24. Linking Depression to Inflammation through Dysregulated Glycogen
Synthase Kinase-3 (GSK3)
Eleonore Beurel, Richard S. Jope
224
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
25. Native Immune System Regulation of the Brain Serotonin Transporter
Chong-Bin Zhu, Kathryn Lindler, Nicole Baganz, William Hewlett,
Randy Blakely
26. Fewer Mature Neurons in the Anterior Dentate Gyrus in Major
Depression: Rescue by Antidepressants
Maura Boldrini, Laika R. Simeon, Tanya H. Butt, Mihran J. Bakalian, Kelly M. Burke, Mark D. Underwood, Gorazd B. Rosoklija, Andrew J. Dwork, René Hen, J. John Mann, Victoria
Arango
27. The Autism-associated Integrin Beta 3 Gene (ITGB3) modulates the
Behavioral and Neurochemical Response to Chronic Stress in Mice
Seth A. Varney, Christa F. Gaskill, Tammy Jessen, Ana Carneiro
28. Stimulation of the Innate Immune System is accompanied by Modulation
of Protein Expression and Phosphorylation State of Synaptic Markers in
the Hippocampus
Janet A. Clark, Ankita Narayan, Maryam Nizami, Brian J. Platt
29. Calcium Sensing Proteins in Depressive Disorder
Yogesh Dwivedi, Hui Zhang, Ghanshyam N. Pandey
30. Activity-Dependent Expression of sFRP3 regulates Adult Hippocampal
Neurogenesis and Antidepressant Actions
Mi-Hyeon Jang, Yasuji Kitabatake, Junjie U. Guo, Ju-Young Kim,
Kurt A. Sailor, Guo-li Ming, Hongjun Song
225
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
31. The FGF14:Nav Channel Complex is a New Target of the Akt/GSK3
Signaling Pathway
Fernanda Laezza, Alexander Shavkunov, Tetyana Buzhdygan,
Miroslav Nenov
32. Optical Activation of Nucleus Accumbens Neurons modulates Depressionand Anxiety-Like Behaviors
Samir Zaman, Pamela J. Kennedy, David M. Dietz, Eric J. Nestler,
Mary Kay Lobo
33. Gene Expression from Lymphoblastoid Cell Lines and Brain Tissue in
Bipolar Disorder: Convergent and Divergent Patterns
Haiming Chen, Nulang Wang, Margit Burmeister, Christopher Ross,
Melvin McInnis
34. Circulating Abeta40 Negatively influences Plasma BDNF Levels
Nunzio Pomara, Davide Bruno, Anilkumar Pillai, Jay J. Nierenberg,
Stephen D. Ginsberg, Pankaj D. Mehta, Henrik Zetterberg, Kaj
Blennow, Peter F. Buckley
35. Incomplete Coverage of Blood Vessels in Orbitofrontal Cortex by
Astrocytic End-feet in Major Depressive Disorder
Grazyna Rajkowska, Jonathan C. Hughes, Samuel S. Newton, Craig
A. Stockmeier, Jose J. Miguel-Hidalgo, Dorota Maciag
36. Epigenetic Modifications in Postmortem Frontal Cortex from Bipolar
Disorder and Alzheimer’s Disease Patients
Jagadeesh S. Rao, Sam Klein, Vasken Kaleshian, Edmund A. Reese,
Stanley I. Rapoport
226
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
37. Optogenetic Control of Mesolimbic Dopamine Neural Activity
Recapitulates the Anxiety-Related Phenotype of the Clock-∆19 Mouse
Model of Mania
Michelle M. Sidor, Sade Spencer, Kay Tye, Melissa Warden, Kafui
Dzirasa, Karl Deisseroth, Colleen A. McClung
38. Peripheral Biomarkers in New Onset of Major Depressive Disorder in
Midlife Women: The Harvard Study of Moods and Cycles
Guilherme A. Behr, Benicio N. Frey, Lee S. Cohen, Jose C. F. Moreira, Michael W. Otto, Bernard L. Harlow, Claudio N. Soares
39. Influence of Major Depressive Disorder on the Volume and Number of
Neurons, Glia, and Perivascular Cells in the Basolateral Amygdala: A
Postmortem Stereological Study
Marisa J. Rubinow, Gouri Mahajan, James C. Overholser, George J. Jurjus, Lesa Dieter, Nicole Herbst, Jose J. Miguel-Hidalgo, Warren
May, Grazyna Rajkowska, Craig A. Stockmeier
40. Selective Optogenetic Stimulation of Parvalbumin-Positive Basal
Forebrain Neurons Reliably entrains Cortical Gamma Oscillations and
Promotes Wakefulness
Tae Kim, James T. McKenna, James M. McNally, Karl Deisseroth,
Robert E. Strecker, Ritchie E. Brown, Radhika Basheer, Robert W. McCarley
41. The Power of Expectation Bias
Janet B.W. Williams, Danielle Popp, Kenneth A. Kobak, Michael J. Detke
227
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
42. A Pilot Study Evaluating the Cognitive Effects of Rimonabant in People
with Schizophrenia
Robert W. Buchanan, Douglas L. Boggs, Robert R. Conley, David
A. Gorelick, Robert P. McMahon, James M. Gold, James Waltz,
Marilyn A. Huestis, Deanna L. Kelly
43. Movement Disorder Trajectories and Treatment Outcomes in a 1-Year
Study of Patients with Schizophrenia
Lei Chen, Haya Ascher-Svanum, Anthony Lawson, Virginia Stauffer,
Allen Nyhuis, Virginia Haynes, Kory Schuh, Bruce J. Kinon
44. Does Acute Oxytocin Administration enhance Social Cognition in
Individuals with Schizophrenia?
Michael C. Davis, Junghee Lee, Michael F. Green, Stephen R. Marder
45. An Intervention to Test the Alpha7 Nicotinic Receptor Model in
Schizophrenia: CDP-choline, a Cholinergic Agonist, and Galantamine, a
Positive Allosteric Modulator
Stephen I. Deutsch, Nina R. Schooler, Barbara L. Schwartz, Clayton
H. Brown, Stephanie M. Rosse, Richard B. Rosse
46. Cardiovascular Effects of Folate Supplementation in Schizophrenia: An
Interim Analysis
Vicki L. Ellingrod, Tyler B. Grove, Stephan F. Taylor
47. Moderate Dose Varenicline Treatment on Neurobiological and Cognitive
Biomarkers in Schizophrenia Smokers and Non-smokers
Elliot Hong
228
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
48. The Alpha7 Neuronal Nicotinic Receptor (NNR) Modulator TC-5619 had
Beneficial Effects and was generally Well Tolerated in a Phase 2 Trial in
Cognitive Dysfunction in Schizophrenia (CDS)
David Hosford, Geoffrey Dunbar, Jeffrey Lieberman, Anthony
Segreti
49. Daytime Sleepiness as a Mediator of Treatment Outcome in a Placeboand Quetiapine XR-Controlled Trial of Lurasidone in Patients with
Schizophrenia
Robert Silva, Josephine Cucchiaro, Andrie Pikalov, Jane Xu, Cynthia
Siu, Antony Loebel, Amir Kalali
50. Clinical Trials of Potential Cognitive-Enhancing Drugs in Schizophrenia:
What Have We Learned So Far?
Richard Keefe, Robert Buchanan, Stephen Marder, Nina Schooler,
Ashish Dugar, Milana Zivkov, Michelle Stewart
51. Adjunctive Lisdexamfetamine Dimesylate Treatment of Predominant
Negative Symptoms of Schizophrenia in Clinically Stable Adults
Maintained on Atypical Antipsychotic Agents: a 14-Week Trial
Robert Lasser, Bryan Dirks, Henry Nasrallah, Courtney Kirsch,
Joseph Gao, Mary Ann Knesevich, Jean-Pierre Lindenmayer
52. Relapse Prevention with Lurasidone vs. Quetiapine XR in Chronic
Schizophrenia: Results of a 12-Month, Double-Blind Study
Antony Loebel, Josephine Cucchiaro, Jane Xu, Kaushik Sarma,
Andrei Pikalov, Amir Kalali
53. Paternal Age and Treatment Response in Adolescents with Schizophrenia
Dolores Malaspina, Opler Mark, Srihari Gopal , Isaac Nuamah ,
Adam Savitz , Jaskaran Singh , David Hough
229
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
54. Correlation between Antipsychotic Efficacy and Weight Gain with
Iloperidone in Short and Long-term Trials in Schizophrenia
Henry A. Nasrallah, Marla Hochfeld, Xiangyi Meng, Richard Wu,
Adam Winseck, Saeeduddin Ahmed
55. Examining Methods for Computing “Clinical Response” in Placebo
Controlled Trials of Antipsychotics in the NEWMEDS Repository
Jonathan Rabinowitz, Nomi Werbeloff, François Menard, Judith
Jaeger, Bruce Kinon, Virginia Stauffer, Francine S. Mandel, Shitij
Kapur
56. Time Course of Dropout Rates in Schizophrenia Trials Conducted from
1966 to 2010: A Systematic Review and Meta-analysis
Ofer Agid, Cynthia Siu, Robert B. Zipursky, Gary Remington
57. Should Patients with Long Durations of Untreated Psychosis be included
in Studies of First Episode Schizophrenia?
Delbert Robinson, Suzanne Sunday
58. PROACTIVE: Initial Results of an RCT Comparing Long-Acting
Injectable Risperidone to 2nd Generation Oral Antipsychotics
Nina R. Schooler, Peter F. Buckley, Jim Mintz, Donald C. Goff,
Alexander Kopelowicz, John Lauriello, Theo Manschreck, Alan J. Mendelowitz, Del D. Miller, Daniel R. Wilson, Juan Bustillo, Joanne
B. Severe, John M. Kane
59. The Effects of Prolonged Administration of an Alpha7 Nicotinic
Cholinergic Agonist Intervention on Neurocognitive Function in
Schizophrenia
Barbara L. Schwartz, Stephen I. Deutsch, Nina R. Schooler,
Stephanie M. Rosse, Clayton H. Brown, Richard B. Rosse
230
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
60. A 6-week Randomized, Double-Blind, Placebo-Controlled, Comparator
Referenced, Multicenter Trial of Vabicaserin in Subjects with Acute
Exacerbation of Schizophrenia
Joan H.Q. Shen, Yonggang Zhao, Sharon Rosenzweig-Lipson,
Danielle Popp, Janet B.W. Williams, Earl Giller, Michael J Detke,
John Kane
61. Early Reduction in PANSS-T Score predicts Later Response to Iloperidone
Therapy: Results from a Pooled Analysis
Stephen M. Stahl, Saeeduddin Ahmed, Xiangyi Meng, Lewis E. Warrington
62. Adjunctive Treatment with the Selective Glycine Uptake Inhibitor Org
25935 in persistent Negative Symptoms of Schizophrenia: Results from
the GIANT Trial
Armin Szegedi, Wim Jansen, Craig Karson, Jacques Schipper, Joep
Schoemaker
63. Neural Correlates of Emotional Response Inhibition in ObsessiveCompulsive Disorder: Emotional Go/no-go Task Development
Heather A. Berlin, Kurt Schulz, Jin Fan, Wayne Goodman
64. Alteration in Neural Response to Emotional Conflict and Conflict
Resolution among Behaviorally Inhibited Adults
Johanna M. Jarcho, Nathan A. Fox, Daniel S. Pine, Ellen Leibenluft,
Tomer, Shechner, Monique Ernst
65. Amygdala-Frontal Circuit Function during Threat Perception, Negative
Affect Regulation, and Rest Across Anxiety Disorders and Major
Depression
K. Luan Phan, Mike Angstadt, Christine Rabinak, Katherine Prater,
Heide Klumpp, Avinash Hosanagar, Kortni Meyers, Scott A. Langenecker
231
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
66. Cortisol Response to Awakening in OEF/OIF Combat Veterans with and
without PTSD: Relationship to PTSD Symptoms, Treatment Response,
and Neural Correlates of Emotional Processing
Sheila Rauch, Anthony King, John Greco, Erin Smith, Barbara
Rothbaum, Israel Liberzon
67. A2 Noradrenergic Neurons induce Fear and Control the Sensitivity to
Anxiety-Related Behavioral Responses
Clara M. Tourino, Luis de Lecea
68. Attention Bias Variability and Relations to Symptoms in PTSD
Gang Wu, Brian Iacoviello, Rany Abend, Daniel Pine, Adriana
Feder, James Murrough, Christopher Bailey, Alexander Neumeister,
Yair Bar-Haim, Dennis Charney
69. Neural Response to Peer Evaluation in Adolescents At Risk for Social
Anxiety Disorder due to Childhood Behavioral Inhibition
Amanda E. Guyer, Eric E. Nelson, Yair Bar-Haim, Koraly PerezEdgar, Monique Ernst, Daniel S. Pine, Nathan A. Fox
70. Functional Connectivity of Response Control in ADHD and Pediatric
Bipolar Disorder with and without ADHD
Alessandra M. Passarotti, James Ellis, Jacklynn Fitzgerald, Jon
O’Neal, Ezra Wegbreit, Mike C. Stevens, Mani N. Pavuluri
71. Fronto-Opercular Control Circuits mediate the Effect of Methylphenidate
on Reaction Time Variability
Chandra S. Sripada, Daniel Kessler, Robert Welsh, Israel Liberzon
232
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
72. Cognitive and Clinical Outcomes associated with Cannabis Use in Patients
with Psychotic Disorders
Raphael Braga, Katherine Burdick, Pamela DeRosse, Anil Malhotra
73. Executive Function in Alcohol-Dependent Adults With and Without
Comorbid Bipolar Disorder: Performance on the Color-Word Interference,
Alcohol, and Emotion Stroop Task
Bryan Tolliver, Delisa Brown, James Prisciandaro, Kathleen Brady
74. From Brain to Behavior - Neural Correlates of Real-World Functioning
Difficulties in Patients with Bipolar Disorder: An fMRI Study
Thilo Deckersbach, Amy T. Peters, Amanda M. Duffy, Andrew D. Peckham, Darin Dougherty, Andrew A. Nierenberg, Scott L. Rauch
75. Neural Response to Monetary Reward in Depression: A Meta-Analysis
Thomas M. Olino, Erika E. Forbes
76. The 2-Year Course of Cognitive Function and IADLs in Older Adults with
Bipolar Disorder
Ariel Gildengers, Meryl A. Butters, Denise Chisholm, Stewart J. Anderson, Amy Begley, Margo B. Holm, Joan C. Rogers, Charles F. Reynolds, Benoit H. Mulsant
77. Impaired Fixation to Eyes in Children with Bipolar Disorder or Severe
Mood Dysregulation
Pilyoung Kim, Chris Baker, Joseph Arizpe, Brooke Rosen, Varun
Razdan, Sarah Jenkins, Daniel Pine, Ellen Leibenluft
233
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
78. Social Cognitive Performance Profile of Bipolar Disorder and
Schizophrenia
Junghee Lee, Jonathan K. Wynn, Lori Altshuler, David Glahn,
William P. Horan, Michael F. Green
79. Developing Translational Neurocognitive measures Specific and Sensitive
to Electroconvulsive Therapy
Shawn M. McClintock, Sarah H. Lisanby, Ira Bernstein, Munro
Cullum, Matthieu Chansard, Brittany Staub, Herbert Terrace,
Mustafa M. Husain
80. Effects of Sertraline on Neurocognition in Outpatients with Major
Depressive Disorder
James L. Reilly, Rebekka Lencer, Michael E. Thase, John A. Sweeney
81. Maternal Brain Responses to Baby-Stimuli are Modulated by
Psychopathological Risk
James E. Swain, S. Shaun Ho, Katherine L. Rosenblum, Eric
Finegood, Patricia Richardson, Leyla B. Akce, Sheila M. Marcus, K. Luan Phan, Maria Muzik
82. The Behavioral and Functional Anatomical Correlates of Autobiographical
Memory Deficits associated with Depression Extend to Individuals at
High Risk for Developing Depression
Kymberly D. Young, Patrick S.F. Bellgowan, Jerzy Bodurka, Wayne
C. Drevets
83. Selective Serotonin Reuptake Inhibitors (SSRI) in Pregnancy: Current
Knowledge of Effects on the Offspring
Regina Casper
234
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
84. Change in Glucose and Lipid Metabolism using Stable Isotope Tracing
during Euglycemic Clamp Conditions during Initial Antipsychotic
Treatment for Disruptive Behavior in Youth
John W. Newcomer, Ginger E. Nicol, Michael D. Yingling, Julia A. Schweiger, Karen S. Flavin, Martha J. Hessler
85. Relationship of Change in Adiposity to Psychiatric Symptom Change
during Randomized Initial Antipsychotic Treatment in Pediatric Disruptive
Behavior Disorders
Ginger E. Nicol, Michael D. Yingling, Karen S. Flavin, Julia A. Schweiger, Martha J. Hessler, John W. Newcomer
86. Pharmacogenetics of Glutamate System Genes and SSRI-Associated
Sexual Dysfunction
Jeffrey R. Bishop, Sharon S. Chae, Shitalben Patel, Jessica Moline,
Vicki L. Ellingrod
87. Deep Brain Stimulation Research for Treatment-Resistant Depression:
Empirical Investigation of Ethical Concerns
Paul Christopher, Carl Fisher, Paul Appelbaum, Paul Holtzheimer,
Helen Mayberg, Sarah Lisanby, Laura Dunn
88. Neuron-Specific Deletion of Histone Methyltransferase Mll1 is associated
with Behavioral Deficits and Altered Histone Methylation at Neuronal
Gene Promoters
Mira Jakovcevski, Wenje Mao, Iris Cheung, Caroline Connor, Juerg
Straubhaar, Schahram Akbarian
89. Emotional Modulation of Response Inhibition in Unaffected Siblings of
Patients with Bipolar I Disorder
Katherine Burdick, Jesse Brand, Nisali Gunawardane, Anil Malhotra,
Terry Goldberg
235
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
90. Whole Exome Sequencing in First Degree Cousin Pairs with Early Age-atOnset Bipolar Disorder
David T. Chen, Nirmala Akula, Layla Kassem, Francis J. McMahon
91. Association of Genetic Variants with Baseline Pain in Patients with Major
Depressive Disorder
John Houston, Wei Zou, Virginie Aris, Bonnie Fijal, Smriti Iyengar,
Alexandra Heinloth, James Martinez
92. Whole Genome Sequencing identifies a Coding Sequence Variant in the
NTRK1 Gene Segregating with Both Bipolar Disorder and Medullary
Cystic Kidney Disease in an Unusual Family
John Kelsoe, Tatyana Shekhtman, Szabolcs Szelinger, David Craig
93. Differential Effects of BDNF and 5-HTT Polymorphisms on InterferonRelated Depression
Francis Lotrich, Robert Ferrell, Denise Sorisio, Salwa Albusaysi,
David Brent
94. Identification of Mitochondrial Somatic Mutations in Subjects with Mood
Disorders and Schizophrenia
Firoza Mamdani, Pedro A. Sequeira, Brandi Rollins, William E. Bunney, Fabio Macciardi, Marquis P. Vawter
95. CRHR1 Genetic Variation potentiates Stress-Induced Anhedonia: A
128-Channel Event-Related Potential Study
Diego A. Pizzagalli, Ryan Bogdan, Roy H. Perlis, Diane L. Santesso,
Jesen Fagerness
236
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
96. COMT Gene Influences on Novelty Seeking across Species: A Preliminary
Study
Arpi Minassian, Victoria Risbrough, Xianjin Zhou, William Perry,
John R. Kelsoe, Mark A. Geyer
97. More Severe Longitudinal Bipolar Illness Course associated with Early
Life Stress in Brain-Derived Neurotrophic Factor Met Allele Carriers but
not Non-Met Allele Carriers
Shefali Srivastava, Joachim Hallmayer, Po W Wang, Shelley J. Hill,
Sheri L. Johnson, Terence A. Ketter
98. Decreased Tryptophan Hydroxylase 1 mRNA Expression in PMS/PMDD
Meir Steiner, Rachel Snaidero, Sonali Lokuge, Jane A. Foster, Marg
Coote, Brianne Nicholls, Claudio N. Soares, Benicio N. Frey
99. Relationship between DISC1 SNPs and Brain Volumes in First Episode
Schizophrenia
Nancy Andreasen, Beng-Choon Ho, Eric Epping, Brett Weiss,
Marsha Wilcox, Steven Ziebell, Thomas Wassink
100. The Psychosis Spectrum in a Young Community Sample
Monica E. Calkins, Jan Richard, Kathleen R. Merikangas, Marcy
Burstein, Leanne Heaton, Haijun Qui, Frank D. Mentch, L. Hermannsson, John Connolly, Debra J. Abrams, Rosetta Chiavacci,
P.M. Sleiman, James Loughead, Ruben C. Gur, Hakon Hakonarson,
Raquel E. Gur
237
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
101. A Map Function Predictive of Risk of Psychosis Obtained by Concurrent
Genome Wide Association to Dopaminergic Deficits
Gabriel A. de Erausquin, Igor Zwir, María Calvó, Néstor V. Florenzano, Gabriela Gonzalez Aleman, Maribel Martínez, Eduardo
Padilla, Mason C. Breed, Bruno A. Benítez, Helen Belálcazar,
Gonzalo Guerrero, Beatriz Molina Rangeon, Manuel Sedó, Horacio
A. Conesa, Javier I. Escobar, C. Robert Cloninger
102. Analysis of CpG SNPs in Serotonin System Genes: Analysis of Suicide
Attempt in Schizophrenia
Vincenzo De Luca, Clement Zai, John Strauss, James L. Kennedy
103. Heritability Analyses of Endophenotypic Measures for Schizophrenia from
the Consortium on the Genetics of Schizophrenia (COGS)
Tiffany A. Greenwood, Robert Freedman, Michael F. Green, Raquel
E. Gur, Keith H. Nuechterlein, Ann Olincy, Allen D. Radant,
Nicholas J. Schork, Larry J. Seidman, Larry J. Siever, Neal R. Swerdlow, Debby W. Tsuang, David L. Braff
104. Identification of Functional Variants in Schizophrenia Patients by Next
Generation Sequencing
Colin A. Hodgkinson, David St. Clair, Polina Iarikova, Qiaoping
Yuan, Basel Baghal, Isioam Mordi, David Goldman
105. Further Investigations into the Serotonin Transporter Gene in
Schizophrenia: A Family-Based Association Study
Bhaskar Kolachana, Qiang Chen, Fengyu Zhang, Daniel Weinberger
106. Genomewide Association Study implicates NDST3 in Schizophrenia and
Bipolar Disorder
Todd Lencz, Saurav Guha, Pamela DeRosse, Anil Malhotra, Chunyu
Liu, Ariel Darvasi
238
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
107. Functional Polymorphisms of the MC4R and NPY Genes are associated
with Antipsychotic-Induced Body Weight Gain
Daniel J. Mueller, Nabilah I. Chowdhury, Arun K. Tiwari, Eva J. Brand, Jeffrey A. Lieberman, Herbert Y. Meltzer, James L. Kennedy
108. Association between a Genetic Marker in HTR2A and Response to
the mGlu2/3 Agonist LY2140023 Monohydrate in the Treatment of
Schizophrenia
Laura K. Nisenbaum, Fangyi Zhao, AnnCatherine M. Downing,
Leanne M. Munsie, Peining Chen, Bonnie A. Fijal, Michelle R. Smith, Brian A. Millen, David H. Adams, Bruce J. Kinon
109. Validation of Candidate Endophenotypes for use in Genomic and Clinical
Outcome Studies of Schizophrenia
Gregory A. Light, Neal R. Swerdlow, Anthony J. Rissling, Allen D. Radant, Catherine A. Sugar, Joyce Sprock, Marlena Pela, Mark A. Geyer, David L. Braff
110. Structure of Endophenotypes in Schizophrenia: Factor Analysis of
15 Putative Endophenotypes from the Consortium on the Genetics of
Schizophrenia (COGS)
Larry J. Seidman, Gerhard Hellemann, Robert Freedman, Raquel E. Gur, Ruben C. Gur, Gregory A. Light, Keith H. Nuechterlein, Ann
Olincy, Allen D. Radant, Larry J. Siever, Jeremy M. Silverman, Neal
R. Swerdlow, Debby W. Tsuang, Ming T. Tsuang, David L. Braff,
Michael F. Green
111. X-linked GABA Receptor Genes show Altered Expression in the Anterior
Cingulate Cortex in Schizophrenia
Monsheel Sodhi, John A. Bostrom, Daniel J. Mount, Vahram
Haroutunian, James H. Meador-Woodruff
239
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
112. NMDAR2B Genotype influences Prefrontal mRNA Levels and Working
Memory in Schizophrenia and Controls
Cynthia S. Weickert, Samantha Fung, Katherine Allen, Loretta
Moore, Mico Chan, Xu-Feng Huang, Kelly Newell, Alice Rothwell,
Thomas W. Weickert
113. Genome-Wide Analysis of Antipsychotic Drug Response in Schizophrenia
Fengyu Zhang, Kristin Bigos, Daniel Weinberger
113. Reduced Anterior Cingulate Glutamate in Euthymic, but not Depressed,
Patients with Obsessive Compulsive Disorder
Christopher Pittenger, Eileen Billingslea, Lihong Jiang, Suzanne
Waylink, Andrew J. Kobets, Brian Pittman, Michael H. Bloch,
Gerard Sanacora, Graeme F. Mason
114. Effects of Rapid Tryptophan Depletion on Reactive Aggression in Adults
with Attention-Deficit/Hyperactivity Disorder (ADHD)
Marco Zimmermann, Marco Grabemann, Christian Mette, Mona
Abdel-Hamid, Jennifer Ueckermann, Markus Kraemer, Jens
Wiltfang, Bernhard Kis, Florian Daniel Zepf
115. CRP is Associated with Physical Well-Being, but not with Early Life
Stress or Depression Symptoms, in Healthy Adults
Linda Carpenter, Cyrena Gawuga, Audrey Tyrka, Lawrence Price
117. Proinflammatory Cytokines in Teenage Suicide Brain
Ghanshyam N. Pandey, Hooriyah S. Rizavi, Xinguo Ren, Yogesh
Dwivedi
240
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
118. Vipri Gene Expression is Increased in the Post-Mortem Hippocampus of
Individuals with Major Depressive Disorder
Cortney A. Turner, William E. Bunney, Alan F. Schatzberg, Jack D. Barchas, Richard M. Myers, Edward G. Jones, Huda Akil, Stanley J. Watson Jr.
119. Cerebrospinal Fluid Substance P-Like Immunoreactivity: Correlates with
Aggression in Personality Disordered Subjects
Emil F. Coccaro, Michael J. Owens, Becky Kinkead, Charles B. Nemeroff
120. Hippocampal NAA and Volume: Potential Response Biomarkers to
Glutamate Based Drugs
Chadi G. Abdallah, Jeremy D. Coplan, Andrea Jackowski, Xiangling
Mao, Dikoma C. Shungu, Sanjay J. Mathew
121. Predictors and Consequences of Cognitive Therapy for PTSD: Neural
Responses to Emotional Anticipation
Robin L. Aupperle, Carolyn B. Allard, Erin M. Grimes, Alan N. Simmons, Taru Flagan, Michelle Behrooznia, Shadha H. Cissell,
Steven R. Thorp, Sonya B. Norman, Martin P. Paulus, Murray B. Stein
122. Neural Dysfunction when Appraising Threat during Extinction Recall:
Effects of Anxiety and Development
Jennifer C. Britton, Maxine A. Norcross, Kristin L. Szuhany, Shmuel
Lissek, Christian Grillon, Daniel S. Pine
123. Shared Pathophysiology in the Pediatric Anxiety Disorders: ConflictRelated Hyperactivation of the Anterior Cingulate Cortex
Kate D. Fitzgerald, Yanni Liu, Robert C. Welsh, Emily R. Stern,
Gregory L. Hanna, Chris S. Monk, K. Luan Phan, Stephan F. Taylor
241
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
124. Structural Brain Differences in PTSD are Associated with Impaired Fear
Inhibition
David Gutman, Kerry J. Ressler, Negar Fani, Tim Ely, Ebony
M. Glover, Seth D. Norrholm , Orion Kiefer, Ivo Dinov, Alex
Zamanyan, Arthur Toga, Tanja Jovanovic
125. Neural and Behavioral Correlates of Peritraumatic Dissociation in an
Acutely Traumatized Sample
Ruth A. Lanius, Judith K. Daniels , Nicholas J. Coupland , Kathleen
M. Hegadoren , Brian H. Rowe , Richard W.J. Neufeld
126. Reduced Amygdala Serotonin Transporter Binding in Posttraumatic Stress
Disorder revealed by [11C]AFM and Positron Emission Tomography
James W. Murrough, Yiyun Huang, Jian Hu, Shannan Henry, Wendol
Williams, Jean-Dominique Gallezot, Christopher R. Bailey, John H. Krystal, Richard E. Carson, Alexander Neumeister
127. fMRI Study of Fear Acquisition and Extinction in Posttraumatic Stress
Disorder
Yuval Neria, Scott Schafer, Margarita V. Bravova, Mohammed R. Milad, Martin Lindquist, Maria J. Malaga, Mariana J. Neria, Myrna
M. Weissman, John C. Markowitz, Tor D. Wager, Gregory Sullivan
128. Towards the Neural Basis of Attentional Training Effects in Social Anxiety
Charles T. Taylor, Robin L. Aupperle, Taru Flagan, Sarah G. Sullivan, Alan N. Simmons, Nader Amir, Martin P. Paulus, Murray
B. Stein
129. Cognitive Behavioral Therapy in Obsessive-Compulsive Disorder
Changes Connectivity between Anterior Insula and Default Mode Network
Emily R. Stern, James L. Abelson, Joseph A. Himle, Jamey J. Lister,
Kate D. Fitzgerald, Robert C. Welsh, Stephan F. Taylor
242
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
130. Increased Risk for Affective Disorders Programmed In Utero? High
Prenatal Maternal Cortisol and Size of the Amygdala in the 6-9 Year-old
Offspring
Claudia Buss, Elysia P. Davis, Jens C. Pruessner, L. Tugan Muftuler,
Kevin Head, Anton Hasso, Curt A. Sandman
131. Dynamic Causal Modeling of Incentive Anticipation in Healthy Adults
and Adolescents
Youngsun T. Cho, Amanda E. Guyer, Stephen J. Fromm, Allison
Detloff, Daniel S. Pine, Julie L. Fudge, Monique Ernst
132. Reorganization of Somatosensory Cortex in Adult Women with Histories
of Childhood Abuse
Christine M. Heim, Helen S. Mayberg, Tanja Mletzko, Charles B. Nemeroff, Jens C. Pruessner
133. Social Subordination Stress and Serotonin Transporter (5HTT)
Polymorphisms Affect the Development of Brain White Matter Tracts in
Juvenile Female Macaques: Behavioral and Neuroendocrine Correlations
Brittany R. Howell, Jodi Godfrey, Vasiliki Michopoulos, Xiaodong
Zhang, Govind Nair, Xiaoping Hu, Mark Wilson, Mar Sanchez
134. Healthy Pediatric White Matter Development: A Diffusion Tensor Imaging
Study and Meta-Analysis
Bart D. Peters, Philip R. Szeszko, Joaquim Radua, Pamela DeRosse,
Jian-Ping Zhang, Toshikazu Ikuta, Patricia Gruner, Anil K. Malhotra
135. Amphetamine Effects on MATRICS Performance in Healthy Adults:
Prelude to a Genetic Analysis
Neal R. Swerdlow, Jo A. Talledo, Justin Kei, Susrutha Thanam,
Sarah N. Lamb, Gregory A. Light
243
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
136. Does Birthweight Directly impact Later Neurocognitive Outcomes? A
Strict Test using Longitudinal Neuroimaging in Monozygotic Twins
Armin Raznahan, Liv Clasen, Francois Lalonde, Jonathan
Blumenthal, Jay Giedd
137. Effects of Early Life Stress on Infant Macaque Brain Development: A
Longitudinal Study of Structural and Functional Connectivity Changes
using Diffusion Tensor Imaging and Resting State fMRI
Mar Sanchez, Yundi Shi, Xiadong Zhang, Govind Nair, David
Grayson, Xiaoping Hu, Phil Fisher, Damien Fair, Martin Styner,
Brittany R. Howell
138. Impact of In-Scanner Head Motion on Multiple Measures of Functional
Connectivity: Relevance for Studies of Neurodevelopment in Youth
Theodore D. Satterthwaite, Daniel H. Wolf, James Loughead, Kosha
Ruparel, Mark A. Elliott, Hakon Hakonarson, Ruben C. Gur, Raquel
E. Gur
139. Effect of BDNF Val66Met and Childhood Trauma on Brain Volume,
Resting State Functional Connectivity and Alexithymia in Healthy
Volunteers
Betty Jo Salmeron, Mary R. Lee, Courtney L. Gallen, Pradeep
Kurup, Thomas J. Ross, Colin A. Hodgkinson, David Goldman,
Mary-Anne Enoch, Hong Gu, Elliot A. Stein
140. Brain Network Properties in Autism, ADHD, and Typically Developing
Children: Similarities and Differences
Adriana Di Martino, F. Xavier Castellanos, Michael P. Milham
244
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
141. Preliminary Findings from a Developmental Meta-Analysis of Neural
Correlates of Autism Spectrum Disorders
Daniel P. Dickstein, Matthew F. Pescosolido, Brooke L. Reidy,
Thania Galvan, Eric M. Morrow
142. Prenatal Exposure to Cigarettes or Alcohol affects the Volume of
Cerebellum in Attention-Deficit/Hyperactivity Disorder
Patrick de Zeeuw, Fenny Zwart, Regina Schrama, Herman van
Engeland, Sarah Durston
143. Functional Magnetic Resonance Imaging of Social Neural Processing in
Rats exposed Prenatally to Valproic Acid
Ada C. Felix-Ortiz, Marcelo Febo
144. Disturbed Microstructural Integrity of the Frontostriatal Tracts and
Cognitive Dysfunction in Children with Attention Deficit Hyperactivity
Disorder
Susan S.-F. Gau, Kathleen R. Merikangas, Wen-Yih I. Tseng
145. Corpus Callosum and Anterior Commissural Aberrations in Aggressive
Bipolar Youth
Kirti Saxena, Leanne Tamm, Annie Walley, Alex Simmons, Nancy
Rollins, Graham Emslie, Xin Fan, Jair Soares, Hao Huang
146. The Impact of Methylphenidate Treatment on Cortical Activation
during Facial Emotion Processing in Children with Dysphoric Mood
Dysregulation Disorder
Leslie Hulvershorn, Tom Hummer, Yang Wang, Annemarie Loth,
Amit Anand
245
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
147. Decreased Connectivity of the Medial Prefrontal Region With-in the
Default Mode Network in Youths with ADHD is Associated with Reduced
Attention
Melissa Lopez-Larson, Jeffery Anderson, Michael Ferguson,
Deborah Yurgelun-Todd
148. Assessing Amyloid Load in Non-demented Young Adults with Down’s
Syndrome
Julie C. Price, Bradley T. Christian, William E. Klunk, Ann D. Cohen, Sigan L. Hartley, Marsha M. Seltzer, Sterling C. Johnson,
Dhanabalan Murali, Peter D. Bulova, Rameshwari Tumuluru, Brian
L. Lopresti, Chester A. Mathis, Benjamin L. Handen
149. Altered Functional Connectivity in Boys and Girls with Attention Deficit
Hyperactivity Disorder
Dardo Tomasi, Nora Volkow
150. Reduced Insular Volume in Attention Deficit Hyperactivity Disorder
Deborah Yurgelun-Todd, Erin McGlade, Melissa Lopez-Larson
151. Diffusion Imaging in Individuals with Partial Deletions of the Williams
Syndrome Critical Region
Stefano Marenco, Dharshan Chandramohan, Shane Kippenhan,
Lindsay Walker, Katheine Roe, Aarthi Padmanabhan, Philip Kohn,
Carolyn Mervis, Ariel Pani, Colleen Morris, Daniel Weinberger,
Carlo Pierpaoli, Karen Berman
152. Functional and Anatomical Connectivity underlying Vulnerability to
Auditory Hallucinations in Schizophrenia Spectrum Disorders
Ann K. Shinn, Atilla Gönenç, Randy Buckner, Dost Öngür
246
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
153. Subcortical D3/D2 Receptor Binding in Cocaine Dependent Humans
David Matuskey, Jean-Dominique Gallezot, Keunpoong Lim, MinqQiang Zheng, Shu-Fei Lin, Richard E. Carson, Robert Malison,
Yu-Shin Ding
154. Inhibition, Body Mass Index, and Eating Behavior across the Life Span
Lawrence Maayan, Allison S. Larr, Melissa Benedict, Alexis
Moreno, Laura Panek, Daniel Javitt
155. Acute Cortisol Elevations Bias Memory Formation in a Negative
Direction only in Depressed Individuals with a History of Early Loss
Heather C. Abercrombie, Allison L. Jahn, Roxanne M. Hoks
156. Detection of Depression in a Clinical Population with Comorbid Pain
Using a Multi-Analyte Biomarker Approach
John A. Bilello, Linda M. Thurmond, Katie M. Smith, Bo Pi, Perry
F. Renshaw
157. Lack of Stress Reactivity to an Extended Continuous Performance Task in
Symptomatic Outpatients with Bipolar Disorder: A Pilot Study
David E. Fleck, Matthew S. Smith, Michelle J. Durling, Thomas
D. Geracioti, Nosakhare N. Ekhator, Erik B. Nelson, Melissa P. DelBello, Caleb M. Adler, Stephen M. Strakowski
158. Low CSF Oxytocin reflects High Intent in Suicide Attempters
Jussi Jokinen, Peter Nordström, Kerstin Uvnäs Moberg, Marie
Åsberg
247
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
159. Modulation of Adult Hippocampal Neurogenesis through HPA Axis
Activity Determines the Divergent Effects of Distress and Eustress on
Affective Disorders
Michael L. Lehmann, Rebecca A. Brachman, Keri Martinowich,
Robert Schloesser, Miles Herkenham
160. Supraphysiological Levothyroxine Treatment improves Mood in
Association with Limbic Metabolism of Bipolar Depressed Patients: A
Randomized, Placebo-Controlled Study
Michael Bauer, Steven Berman, Thomas Stamm, Mazda Adli,
Maximilian Pilhatsch, Edythe D. London, Peter C. Whybrow, Florian
Schlagenhauf
161. Avoidance, Safety Behaviour, and Reassurance Seeking in Generalized
Anxiety Disorder
Katja Beesdo-Baum, Elsa Jenjahn, Michael Hoefler, Ulrike Lueken,
Eni S. Becker, Juergen Hoyer
162. Pilot Study of Mindfulness-based Exposure Therapy for PTSD in OEF/
OIF Veterans: Preliminary Clinical Outcomes and Pre-post fMRI
Neuroimaging
Anthony King, Nicholas Giardino, Sheila Rauch, Sripada Rebecca,
Jiun-Yiing Hu, Israel Liberzon
163. Clinical Survey of the Mother-Infant Mental Health Clinic: Psychiatric
Characteristics of Consecutive 109 Patients
Keiko Yoshida, Yukako Fujinaga, Hiroshi Yamashita, Shigenobu
Kanba
248
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
164. EEG Predictors of Response to Brain Computer Interface Treatment in
ADHD
Tih Shih Lee, Stephanie Teng, Haihong Zhang, Yudong Zhao, Choon
Guan Lim, Cuntai Guan, Daniel Fung, Yin Bun Cheung, K. Ranga
Krishnan
165. GABAA and TLR4 Innate Immunity Receptors of the Ventral Tegmental
Area Regulate Nicotine Sensitization in Alcoholic Rats
Kaitlin T. Warnock, Juan Liu, Timothy Kelly, Laure Aurelian, Harry
L. June
166. Real-time fMRI Neurofeedback targeting Inhibitory Control Brain
Activation decreases Emotional Reactivity to Smoking Cues in Cigarette
Smokers
Luke E. Stoeckel, X.J. Chai, Oliver Hinds, Alice Coakley, Alex
Tighe, Susan Whitfield-Gabrieli, John Gabrieli, A. Eden Evins
167. Preliminary Results Showing that Treatment Seeking Cigarette Smokers
can use Realtime fMRI Feedback of Regional Brain Activity to Reduce
Cue Induced Craving and Regional Blood Flow
Mark S. George, Xingbao Li, Karen Hartwell, Jeffrey J. Borckardt,
James J. Prisciandaro, Michael E. Saladin, Paul Morgan, Kevin
Johnson, Todd LeMatty, Kathleen Brady
168. Cortical Excitability in Current Cocaine Users: A Transcranial Magnetic
Stimulation Study Investigating Glutamatergic and GABAergic Processes
Colleen A. Hanlon, Sarah E. Fredrich, Mark S. George
169. Deep Brain Stimulation in Treatment Resistant Alcohol Addiction Longterm Results of the Magdeburg Pilot Study
Ulf J. Mueller, Bernhard Bogerts
249
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
170. Plasma Levels of Vasopressin are Altered in Response to Opioid Receptor
Modulation: Mass Spectrometric Quantification
Brian Reed, Brian T. Chait, Mary Jeanne Kreek
171. Experienced Oioid Abusers’ Attempts to Prepare a Crush-Resistant
Oxymorphone Extended-Release Formulation for Intranasal or
Intravenous Use
Suzanne K. Vosburg, Jermaine D. Jones, Jeanne M. Manubay, Judy
B. Ashworth, Irma H. Benedek, Sandra D. Comer
172. Risperidone Long-acting Injections - How to Optimize the Treatment?
Eva Ceskova, Jan Pecenak, Ivan Tuma, Pavel Mohr, Martin Anders
173. Gradual and Overlapping Antipsychotic Switch Strategies are associated
with Less All-Cause Discontinuation in Schizophrenia: Results from a
Meta-analysis of Different Switch Strategies
Christoph U. Correll, Vishesh Agarwal, John M. Kane
174. Long-Term Safety and Effectiveness of Lurasidone in Schizophrenia:
Results of a 22-Month, Open-Label Extension Study
Josephine Cucchiaro, Jay Hsu, Masaaki Ogasa, Robert Silva, Doreen
Simonelli, Andrei Pikalov, Antony Loebel, Christoph U. Correll
175. Noradrenargic Involvement in Basic Information processing Deficits in
Schizophrenia: The Effects of Clonidine on Sensorimotor Gating and
Mismatch Negativity
Bob Oranje, Birte Glenthoj
250
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
176. Mid- and Long-Term Efficacy & Effectiveness of Antipsychotic
Medications for Schizophrenia: A Data-Driven, Personalized Clinical
Approach
Ira Glick, John Davis, Christoph Correll
177. Cognitive Performance in Patients with Schizophrenia Treated with
Lurasidone: Results from a Placebo- and Active-Controlled Acute Phase
Study followed by a 6 Month Double-Blind Extension
Philip Harvey, Josephine Cucchiaro, Andrei Pikalov, Antony Loebel,
Cynthia Siu
178. EVP-6124, An Alpha-7 Nicotinic Partial Agonist, Produces Positive
Effects on Cognition, Clinical Function, and Negative Symptoms in
Patients with Chronic Schizophrenia on Stable Antipsychotic Therapy
Herbert Y. Meltzer, Maria Gawryl, Susan Ward, Nancy Dgetluck,
Chaya Bhuvaneswaran, Gerhard Koenig, Michael G. Palfreyman,
Dana C. Hilt
179. Profound Time-related Remission of Psychosis and Improvement in
Cognition and Grey Matter Volume in Treatment Resistant Schizoprenia/
Psychotic Spectrum Disorder during Treatment with High Dose
Risperidone CONSTA: A Case Report
Herbert Y. Meltzer, Minyoung Sim, Turner N. Jernigan, Wade M. Allen, Christopher Cannistraci, Adam Anderson
180. Antidepressant and Anxiolytic Effects of Quetiapine Strongly Correlate to
Neuropeptide Y Increase and Corticotropin-releasing Hormone Decrease
in CSF from Schizophrenic Patients
Aleksander A. Mathé, Pierre Baumann, Georg Nikisch
251
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
181. Safety of Lurasidone in Short-Term Schizophrenia Trials: A
Comprehensive Database Analysis
Andrei Pikalov, Robert Silva, Josephine Cucchiaro, Jay Hsu, Jane
Xu, Antony Loebel
182. D2 Receptor Occupancy measured with 18F-Fallypride following
Lurasidone Treatment in Schizophrenia Patients
Steven G. Potkin, David B. Keator, Adrian Preda, Dana Nguyen,
Antony Loebel, Jogeshwar Mukherjee, Josephine Cucchiaro,
Marilyn Keslerwest, Nikunj Shah
183. Effects of Metformin on Weight, Glucose, and Cognition in Chronic
Psychotic Patients
Robert C. Smith, Saurabh Dwivedi, Davis John
184. The Effect of Switching from Olanzapine, Quetiapine,or Risperidone
to Aripiprazole on Risk of Cardiovascular Disease: Results from the
Comparison of Antipsychotics for Metabolic Problems (CAMP) Study
T. Scott Stroup, Robert M. Hamer, Neepa Ray, Susan M. Esscok,
Jeffrey A. Lieberman
185. Schizophrenia and Co-morbid Tobacco Addiction: The Role of
Impulsivity, Decision-Making and Executive Function Deficits
Victoria C. Wing, Caroline E. Wass, Rachel Rabin, Tony P. George
186. Efficacy and Safety of Second- vs First-Generation Antipsychotics in First
Episode Schizophrenia: A Systematic Review and Meta-analysis
Jianping Zhang, Juan Gallego, Delbert Robinson, Anil Malhotra,
John Kane, Christoph Correll
252
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
187. Investigation of Sex-Dependent Effects of Marijuana in Heavy Marijuana
Smokers
Ziva D. Cooper, Margaret Haney
188. Short-Term Modafinil Administration improves Working Memory and
Sustained Attention in Cocaine-Dependent Volunteers
Ari D. Kalechstein, James J. Mahoney, Ryan S. Bennett, Ravi S. Shah, Jin H. Yoon, Lee C. Chang, Richard De La Garza
189. Effects of Subjective and Objective Responses to Alcohol Challenge in
Indo and Afro Trinidadians
Karelia Montane Jaime, Samuel Shafe, Cindy L. Ehlers
190. Naltrexone accentuates Ethanol-Induced Feelings of Intoxication and the
fMRI BOLD Response to Negative Facial Expressions in the Insula of
Treatment Seeking Alcoholics
David T. George, Melanie Schwandt, Lishu Zhang, Dan Rio, Reza
Momenam, Daniel Hommer, Vijay Ramchandani, Markus Heilig
191. Nabilone decreases Marijuana Withdrawal and Relapse in the Human
Laboratory
Margaret Haney, Gillinder Bedi, Ziva Cooper, Suzanne Vosburg,
Sandra Comer, Richard Foltin
192. Anhedonia predicts Diminished Sociocognitive Reward Processing in
Abstinent Cigarette Smokers
Adam M. Leventhal, Marcus Munafò, Jennifer W. Tidey, Steve
Sussman, John R. Monterosso, Ping Sun, Christopher W. Kahler
253
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
193. A Single Administration of Low Dose Varenicline Saturates α4β2*
Nicotinic Acetylcholine Receptors in the Human Brain
Shahrdad Lotfipour, Mark Mandelkern, Miguel Alvarez-Estrada,
Arthur L. Brody
194. The ACE Inhibitor Perindopril may Attenuate Psychostimulant
Effects produced by Methamphetamine in Non-Treatment-Seeking,
Methamphetamine-Dependent Volunteers
Thomas F. Newton, Richard De La Garza, Yasmine Omar, Colin N. Haile, Daryl Shorter, Rollin Hawkins, Chandra Nerumalla
195. Pharmacogenetics of Naltrexone in Asian Americans: A Randomized
Placebo-Controlled Laboratory Study
Lara Ray, Spencer Bujarski, Karen Miotto
196. Transdermal Alcohol Measurement for the Assessment of Naturalistic
Alcohol Drinking
Robert M. Swift, Andrea Grenga, Junghyun Kim, Linda Tempelman,
Michael Moeller
197. The Effect of Receptor Reserve on Allosteric Enhancement of Efficacy at
Gq-coupled Muscarinic Receptors
John Ellis, Edward Stahl, Gwendolynne Elmslie
198. Alpha2-Adrenoceptor Blockade contributes to the Asenapine-Induced
Elevation of Prefrontal Cortical Catecholamine Outflow
Monica M. Marcus, Olivia Frånberg , Torgny H. Svensson
254
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
199. The Novel ALPHA7 Receptor Partial Agonist, BMS-902483,
demonstrates Robust Efficacy across Models of Cognitive and Sensory
Deficits in Schizophrenia
Amy Easton, Regina Lidge, Kelli Jones, Yu-Wen Li, Rick Pieschl,
Sivarao Digavalli, Ping Chen, Zubin Bhagwagar, Jim Cook, Dalton
King, Christiana Iwuagwu, John Macor, Robert Zaczek, Richard
Olson, Linda J. Bristow
200. IN VITRO Characterization of BMS-902483, a Potent, Partial Agonist
at the ALPHA7 Nicotinic Acetylcholine Receptor for the Treatment of
Cognitive Deficits in Schizophrenia and Alzheimer’s Disease
Linda J. Bristow, Nicholas Lodge, Adam Hendricson, Ryan
Westphal, Yu-Wen Li, Rex Denton, Debra Post-Munson, Lizbeth
Gallagher, Thaddeus Molski, Rick Pieschl, Jim Cook, Dalton King,
Christiana Iwuagwu, Richard Olson, John Macor, Robert Zacek
201. Blockade of Pramipexole effects on Prepulse Inhibition and Accumbens
c-Fos Expression by U99194
Wei-li Chang, Michelle R. Breier, Richard L. Saint Marie, Alex
Yang, Samantha R. Hines, Neal R. Swerdlow
202. A Deficit of Serotonin 2A Receptors mediates the Resistance of Egr3Deficient Mice to Sedation by Clozapine
Alison Williams, Wendy Ingram, Sarah Levine, Jack Resnik, Scott
Janowski, Christy Kamel, James Lish, Diana Elizalde, Alexey
Kozlenkov, Javier González-Maeso, Amelia Gallitano
203. EVP-6124, an Alpha-7 Nicotinic Receptor Partial Agonist, enhances
Cognition and Efflux of Dopamine, Acetylcholine, and Glutamate in Rat
Cortex at Low Brain Concentrations
Mei Huang, Herbert Meltzer, Anna Felix, John Panos, Nick van
Goethem, Sonya Bertrand, Dorothy Flood, Chaya Bhuvaneswaran,
Maria Gawryl, Jos Prickaerts, Daniel Bertrand, Dana Hilt, Gerhard
König
255
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
204. Discovery of the First β-Arrestin-Biased Dopamine D2 Ligands for
probing Signaling Pathways Essential for Antipsychotic Efficacy
John Allen, Julianne Yost, Vincent Setola, Xin Chen, Maria Sassano,
Meng Chen, Niels Jensen, Sean Peterson, Prem Yadav, Xi-ping
Huang, Stephen Frye, William Wetsel, Marc Caron, Bryan Roth, Jian
Jin
205. Opioidergic Mechanism of Body Weight Gain in Olanzapine-Treated Rats
Daniel B. Kurbanov, Paul J. Currie, Igor Elman
206. Differential Effects of AMPA Receptor Potentiators and Glycine Reuptake
Inhibitors on Antipsychotic Efficacy and Prefrontal Glutamatergic
Transmission
Torgny H. Svensson, Monica M. Marcus, Anna Malmerfelt,
Mohammed Shahid, Kent Jardemark
207. Behavioral Analysis of Antipsychotic Efficacy in Beta-Arrestin2Knockout Mice with Biased Dopamine D2Receptor Ligands
William C. Wetsel, John A. Allen, Bryan L. Roth, Jian Jin, Marc G. Caron
208. The Triple Monoamine Reuptake Inhibitor, AMR-2, improves Attentional
Set Shifting in a Rat Neurodevelopmental Model of Schizophrenia
Linda J. Bristow, Amy Easton, Matthew A. Seager, Richard Olson,
Shuang Liu, Kelli Jones, Melissa LaPaglia, Karen Heman, Yu-Wen
Li, John Macor, Bruce J. Sargent, John Houston, Bruce F. Molino,
Robert Zaczek
209. Increased Glutamate Tone in the Nucleus Accumbens mediates Excessive
Ethanol Drinking in Dependent Mice
Howard C. Becker, William C. Griffin, Vorani Ramachandra, Patrick
J. Mulholland
256
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
210. Buspirone reduces the Reinforcing Effects of Cocaine and Cocaine +
Nicotine Polydrug Combinations in Rhesus Monkeys
Nancy K. Mello, Jack Bergman, Peter A. Fivel, Stephen J. Kohut
211. Methamphetamine Self-Administration attenuates the Persistent
Dopaminergic Deficits caused by a Subsequent Methamphetamine
Treatment
Lisa M. McFadden, Gregory C. Hadlock, Paula L. Vieira-Brock,
Glen R. Hanson, Annette E. Fleckenstein
212. Src Tyrosine Kinase-Mediated Activation of NMDA Receptor Function in
the Dorsal Hippocampus is Necessary for Drug Context-induced Cocaineseeking Behavior in Rats
Xiaohu Xie, Amy Arguello, Audrey Wells, Heather Lasseter, Rita
Fuchs
213. Effects of Neurotensin (NT) Systems on Maintenance, Extinction and
Reinstatement of Methamphetamine (METH) Self-Administration (SA)
Amanda Hoonakker, Mario Alburges, Glen Hanson
214. Ceftriaxone increases Glutamate Transport and Basal Glutamate Levels in
the Nucleus Accumbens Core of Cocaine Self-Administering Animals
Lori A. Knackstedt, Kate J. Reissner
215. Progressive Behavioral Supersensitivity to Nicotine during Early
Withdrawal from Chronic Cocaine Administration and Prevention of
Cocaine Sensitization using Mecamylamine
Steven T. Szabo, J. Corey. Fowler, Kamal Bhatia, Tong H. Lee
257
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
216. Preclinical Evidence that Alcohol Intake can be Suppressed by an H3
Receptor Antagonist
Timothy Lovenberg, Pascal Bonaventure, Leah Aluisio, Ian Fraser,
Marc R. Azar, Molly Brennan, George F. Koob
217. Evidence for Significant White Matter Alterations following Chronic
Cocaine Exposure in a Nonhuman Primate Model of Drug SelfAdministration
Linda Porrino, Hilary Smith, Si-Wei Wang, Thomas Beveridge,
Michael Nader
218. Self-Administration of Compounds with Functional Selectivity at GammaAminobutyric Acid Type A Receptor Subtypes in Midazolam- but not
Cocaine-Experienced Monkeys
James K. Rowlett, Nina M. Shinday, Bradford D. Fischer, Donna M. Platt, David S. Reynolds, Gerard R. Dawson, John R. Atack
219. Deficits in Ventral Prefrontal Cortex Group1 Metabotropic Glutamate
Receptor Function Mediate Resistance to Extinction during Protracted
Withdrawal from an Extensive History of Cocaine Self-Administration
Osnat Ben-Shahar, Arianne D. Sacramento, Bailey W. Miller, Sierra
M. Webb, Melissa Wroten, Amanda L. Caruana, Evan Gordon, Kyle
L. Ploense, Jennifer Ditzhazy, Tod E. Kippin, Karen K. Szumlinski
220. The Novel Neuropeptide S - Receptor Antagonist, NCGC00185684,
decreases Alcohol Self-Administration in Rats by Suppressing Motivation
for Alcohol
Michelle Zook, Lauren Bell, Melanie L. Schwandt, Annika Thorsell,
Samarjit Patnaik, Juan Marugan, Roberto Ciccocioppo, Markus
Heilig
258
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
221. Orexin mediates Yohimbine Actions in BNST and Impaired Extinction of
Cocaine Place Preference through a Norepinephrine-Independent Process
Kelly L. Conrad, Adeola R. Davis, Yuval Silberman, Douglas J. Sheffler, Angela D. Shields, Sam A. Saleh, Namita Sen, Heinrich
J.G. Matthies, Jonathan A. Javitch, Craig W. Lindsley, Danny G. Winder
222. Effects of Ethanol on Endocannabinoid Modulation of Up-States in
Prefrontal Cortex
John J. Woodward, Matthew Pava
223. Inverted-U Relationship between Cortical Oscillations and Dopamine:
EEG and Computational Studies
Raymond Y. Cho, Christopher Walker, Kubra Komek, G. Bard
Ermentrout, RyAnna Verbiest
224. Does the NMDA-Receptor Antagonist, Ketamine, Mimic the Pattern of
EEG Gamma Oscillation Abnormalities Observed in Schizophrenia? A
Test of the NMDA-Receptor Hypofunction Model
Daniel H. Mathalon, Brian J. Roach, Judith Jaeger, Handan GunduzBruce, John H. Krystal, Judith M. Ford
225. Homeostasis and Quantitative Sleep EEG in Alcohol Dependent Adults
Roseanne Armitage, Robert Hoffmann, J. Todd Arnedt, Deirdre
Conroy, Kirk Brower
226. The Effect of Cocaine in Higher Order Local Circuitry as Revel by
Optogenetics and Pharmacological Methods-Orchestrate and Disorganize
Tamas Tompa, Antonieta Lavin
259
ACNP 50th Annual Meeting • Final Program
Poster Session I—Monday
227. Activation of Ventral Tegmental Area GABAergic Neurons disrupts
Reward Consumption
Ruud van Zessen, Garret Stuber
260
ACNP 50th Annual Meeting • Final Program
Poster Session II – Tuesday
1.
A Zebrafish Model for the Functional Analysis of Genes in Autism
Ellen J. Hoffman, Antonio Giraldez, Matthew State
2.
Histidine Decarboxylase Deficiency produces Tourette Syndrome
Phenomenology and Dopamine Dysregulation in Humans and Mice
Lissandra C. Baldan Ramsey, Kyle Williams, Jean-Dominique
Gallezot, Michael Crowley, George Anderson, Bennett L. Leventhal,
Hiroshi Ohtsu, John H. Krystal, Linda Mayes, Ivan de Araujo, YuShin Ding, Matthew W. State, Christopher Pittenger
3.
Poor and Unstable Sustained Attentional Performance in Sign-Trackers:
An Animal Model of Poor Top-Down Cognitive Control of Attention
Giovanna Paolone, Christopher C. Angelakos, Paul J. Meyer, Terry
E. Robinson, Martin Sarter
4.
Further Support for a Brain-Selective Activity of 17β-Estradiol Prodrug
(DHED) in the Mouse
Michal Arad, Laszlo Prokai, Katalin Tatrai-Prokai, Istvan
Merchenthaler, Todd D. Gould
5.
Depressive-Like Effects of Blocking Astrocytic Glutamate Uptake in the
Prefrontal Cortex
Catherine John, Karen Smith, Ashlee Van’t Veer, Bruce Cohen, Dost
Öngür, Anita Bechtholt-Gompf
6.
Estrogen increases Stress Resilience and Hippocampal Synaptic
Physiology in the Learned Helplessness Rodent Model of Major
Depression
Teruko M. Bredemann, Lori L. McMahon
261
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
7.
Susceptibility and Resilience to Chronic Social Defeat-Induced Anhedonia
in Rats: Effects of Chronic Fluoxetine Treatment
Andre Der-Avakian, Athina Markou
8.
Paternal Transmission of Stressed-Induced Pathologies
David M. Dietz, Quincey LaPlant , Vincent Vialou, Emily L. Watts,
Georgia E. Hodes, Scott J. Russo, Jian Feng, Ronald S. Oosting, Eric
J. Nestler
9.
A Functional Role for Interleukin 6 in Susceptibility to Depression
Georgia E. Hodes, Viktoria Steizhammer, Sam A. Golden, Daniel
J. Christoffel, Jane Magida, Wolfgang Kluge, Carol A. Tamminga,
Subroto Ghose, Erik H.F. Wong, Chi-Ming Lee, Sabine Bahn, Scott
J. Russo
10. Behavioral Stress-Induced Activation of FoxO3a in the Cerebral Cortex of
Mice and the Underlying Signaling Mechanisms
Wenjun Zhou, Ligong Chen, Xiaohua Li
11. Neurocircutiry in the Learned Helplessness Model of Depression Revealed
by Whole Brain c-fos Expression
Martine M. Mirrione, Nora Ruth, Bo Li, Fritz A. Henn
12. Stress Exposure produces a Switch from Appetitive to Aversive Signaling
by Corticotropin-Releasing Factor in the Nucleus Accumbens
Julia Lemos, Matthew Wanat, Jeffrey Smith, Beverly Reyes,
Elisabeth Van Bockstaele, Charles Chavkin, Paul Phillips
262
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
13. Neuropeptide Y System Gene Expression in the Non-human Primate
Amygdala is associated with Anxious Temperament
Patrick H. Roseboom, Steven A. Nanda, Andrew S. Fox, Jonathan A. Oler, Steve E. Shelton, Ned H. Kalin
14. Effect of Chronic Unpredictable Stress on Cortical GABAergic Neurons
Ashley Lepack, Mounira Banasr, Ronald Duman, Gerard Sanacora
15. A Role for Dopamine Transmission in the Acute Rescue of a Depression
Phenotype induced by Chronic Mild Stress
Kay Tye, Julie Mirzabekov, Sung-yon Kim, Melissa Warden, Ilana
Witten, Karl Deisseroth
16. Can a Model with Predictive Validity for Clinical Efficacy in TreatmentResistant Depression be derived from the Differential Behavioral Effects
of Drugs across Mouse Strains?
Jeffrey M. Witkin, Kurt Rasmussen, David Bleakman, Karen Baker,
Nicholas J. Brandon, Zoe A. Hughs, Michelle P. Kelly
17. The Serotonin Syndrome in Monoamine Oxidase (MAO) AB Knockout
Mice: Enhanced Behavioral Responses induced by 5-hydroxy-Ltryptophan (5-HTP) and Tramadol
Meredith A. Fox, Micaella G. Panessiti, Teresa J. Tolliver, Kevin
Chen, Jean C. Shih, Dennis L. Murphy
18. Behavior-Based Neuroactive Drug Discovery in the Zebrafish
David Kokel, Randall Peterson
263
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
19. Roles of GSK-3 and HDAC Inhibition in Beneficial Effects of Combined
Lithium and Valproate Treatment in Transgenic Mouse Models of
Huntington’s Disease
Chi-Tso Chiu, De-Maw Chuang
20. Hippocampal Neuroplasticity is Altered in an Animal Model of NMDA
Receptor Hypofunction, the Serine Racemase Knockout Mouse
Darrick T. Balu, Amy Truong, John P. Corradi, Angela M. Cacace,
Joseph T. Coyle
21. Evidence that Mutation in Neuregulin 1, a Schizophrenia Susceptibility
Gene, alters Glucose Tolerance in Animals
Nancy M. Bivens, Jay A. Gingrich
22. Early Developmental Elevations of Kynurenic Acid, an Endogenous
Negative Modulator of Alpha7 Nicotinic Receptors: A Novel Animal
Model of the Cognitive deficits in Schizophrenia
Ana Pocivavsek, Kathleen S. Alexander, David Bortz, Hui-Qiu Wu,
Robert Schwarcz, John P. Bruno
23. Inhibition of COMT Reverses the Novel Object Recognition Deficit in
COMT-Val Transgenic Mice
Gregory V. Carr, Francesco Papaleo, Daniel R. Weinberger
24. Decreased Synaptogenesis, increased Excitability of Cortical Neurons
and Impaired Working Memory in Transgenic Mice expressing the
Schizophrenia Associated KCNH2 3.1 Isoform
Jingshan Chen, Peixiong Yuan, Qingjun Tian, Feng Yang, Grace
Zhang, Jiemin Jia, Yun Wang, Jing Du, Paul Glineburg, Gregory
Carr, Francesco Papaleo, James Pickel, Zheng Li, Daniel Weinberger
264
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
25. Development of a Discrete Trials Task to Assess Serotonergic Modulation
of Interval Timing in Mice
Adam L. Halberstadt, Jared W. Young, Mark A. Geyer
26. Enhanced Cue-Induced Relapse to Cocaine Seeking in the Neonatal
Ventral Hippocampal Lesion (NVHL) Rat Model of Schizophrenia
Rose-Marie Karlsson, Daniel Kircher, Yavin Shaham, Patricio
O’Donnell
27. Vesicular Monoamine Transporter 1 (VMAT1) Null-Mutant Mice
show Neurodegenerative Changes in Hippocampus - Implications for
Schizophrenia and Bipolar Disorder beyond the Monoamine Hypothesis
Pushpinder K. Multani, Marcel Estévez, Rachel Hodge, Glenn A. Doyle, Falk W. Lohoff
28. Effects of Genetic Reduction of Activity-Dependent BDNF on Cortical
Slow-Wave Activity
Keri Martinowich, Robert Schloesser, Nicholas Hardy, Dennisse
Jimenez, Bai Lu, Daniel Weinberger
29. Further Validation of a New Animal Model of Schizophrenia: Whole
Transcriptome Sequencing by RNA-Seq reveals Expression Changes of
Genes in mPFC Relevant to Schizophrenia
Charles D. Nichols, David A. Martin, Danuta Marona-Lewica, David
E. Nichols
30. Validation of the Cesarean Section Birth Model of Schizophrenia in Rat at
the Molecular Level
Gabriela Novak, Theresa Fan, Susan George
265
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
31. Rat Strain Sensitivity to Startle-Enhancing Effects of D1 Stimulation vs. Gating-Disruptive Effects of D2 Stimulation Neal R. Swerdlow, Steve T.C. Pham, Daniel Keolasy, Samantha R. Hines
32. DISC1 Partners with Serine Racemase to Modulate D-serine Production
by Astrocytes Mikhail Pletnikov, Ting Martin Ma, Jun Nomura, Bagrat Abazyan,
Sofya Abazyan, Akira Sawa, Solomon H. Snyder
33. Initial Behavioral and Neurochemical Characterization of Perinatal
Ketamine Administration in Mice
Susan B. Powell, Luis F. Garcia, Jared W. Young, M. Margarita
Behrens
34. Behavioural Evaluation of Positive AMPA Receptor Modulators CX1739
and CX1837 in Rodent Models of Sustained Attention and Non-Spatial
Working Memory
Mohammed Shoaib, Mark Varney
35. Dopamine Transporter Knockdown Mice exhibit Poorer Within-Session
Risk Learning in a Mouse Iowa Gambling Task consistent with Bipolar
Mania Patients
Jared W. Young, Jordy van Enkhuizen, Mark A. Geyer
36. Progression of Drug Cue-Induced Phasic Dopamine Release from Limbic
to Sensorimotor Striatum mediates Action Selection of Drug-Taking
Behavior in a Rodent Model of Drug Addiction
Ingo Willuhn, Barry J. Everitt, Paul E.M. Phillips
266
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
37. DREADDed Decision-Making: Revealing a Role for the ‘Direct’ Pathway
in Reward Preference
Susan Ferguson, Paul Phillips, Bryan Roth, John Neumaier
38. Melatonin Receptor Deletion abrogates Methamphetamine-Induced
Locomotor Sensitization and Reward in C3H/HeN Mice
Margarita L. Dubocovich, Shannon J. Clough, Anthony J. Hutchinson, Iwona Stepien, Randall L. Hudson
39. Circuit-Specific Spine Modifications with Fear Memory
Jeff D. Sanders, Dilip Jeste, Mark Mayford
40. Human Hair Follicle Derived Induced Pluripotent Stem Cells (iPSC) and
Their Differentiation into Dopaminergic Neurons as a Model to Study
Neurdevelopmental Abnormalities in Schizophrenia
Dorit Ben-Shachar, Odil Robicsek, Isabelle Petit, Nava Salman,
Rachel Karry, Daniel Aberdam
41. GABAA and GABAB Receptor Subunits display Altered Expression in
Cerebella of Subjects with Schizophrenia, Bipolar Disorder, and Major
Depression
S. Hossein Fatemi, Timothy D. Folsom
42. Chandelier Cell Inputs to Pyramidal Neurons in Schizophrenia and
Development
Kenneth N. Fish, Gil D. Hoftman, Robert A. Sweet, David A. Lewis
267
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
43. The Role for NDEL1 in nNOS Signaling for Schizophrenia: Implications
for Cortical Development and Prefrontal Cortex-Mediated Cognitive
Behaviors
Atsushi Saito, Yu Taniguchi, Sandra P. Zoubovsky, Sun-Hong Kim,
Balakrishnan Selvakuma, Vladimir M. Pogorelov, Peter Yoon,
Solomon H. Snyder, Akira Sawa, Mikhail V. Pletnikov, Atsushi
Kamiya
44. Identification of Cellular Signatures for Schizophrenia and Bipolar
Disorder in iPSC-Derived Neuronal Cells
Rakesh Karmacharya, Steven D Sheridan, Kraig Theriault,
Sabine Bavamian, Jennifer Wang, Elizabeth G.J. O’Brien, Sigrun
Gustafsdottir, Katherine L Madden, Donna McPhie, Roy H. Perlis,
Dost Ongur, Alykhan Shamji, Anne E. Carpenter, Bruce M. Cohen,
Stuart L. Schreiber, Stephen J. Haggarty
45. DARPP-32 Transcripts are Upregulated in the Prefrontal Cortex of Major
Psychiatric Disorders and Associated with Genetic Variants
Yasuto Kunii, Thomas M. Hyde, Tianzhang Ye, Chao Li, Daniel R. Weinberger, Joel E. Kleinman, Barbara K. Lipska
46. Ultrastructural Features of the Anterior Cingulate Cortex in Postmortem
Brain from Subjects with Schizophrenia and Controls
Rosalinda Roberts, Joy Roche, Adrienne Lahti
47. Mitochondrial Variants and Subjects with Psychatric Disorders
Marquis P. Vawter, Brandi Rollins, Firoza Mamdani, Fabio
Macciardi, William E. Bunney, Pedro A. Sequeira
48. Prenatal Stress decreases Expression of Transcription Factors in
GABAergic Neuron Progenitors and GABAergic Progenitor Migration
Hanna E. Stevens, Flora M. Vaccarino
268
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
49. The Phosphodiesterase Isoform 4a5 (PDE4a5) is the Critical Mediator of
Hippocampus-Dependent Cognitive Impairments induced by Sleep Loss
Robbert Havekes, Jennifer H.K. Choi, Vibeke M. Bruinenberg,
George S. Baillie, Alan J. Park, Jonathan P. Day, Leonardo A. Guercio, Edward Linton, Hannah Schoch, Sara J. Aton, Peter
Meerlo, Miles D. Houslay, Ted Abel
50. Managing Functional and Cognitive Decline in Patients with Mild-toModerate Alzheimer’s Disease: A 48-week, Randomized, Double-blind
Evaluation of 13.3 mg/24 h (15 cm2) versus 9.5 mg/24 h (10 cm2)
Rivastigmine Patch
Jeffrey Cummings, Lutz Frölich, Sandra E. Black, Serge Bakchine,
Giuseppe Bellelli, José L. Molinuevo, Reto W. Kressig, Pamela
Downs, Angelika Caputo, Sibel Tekin, Christine Strohmaier
51. Efficacy and Tolerability of 5 mg Lu AA21004 in an 8-Week European
Trial of Adults with Generalized Anxiety Disorder
Leszek Bidzan, Atul R. Mahableshwarkar, Paula Jacobsen, Mingjin
Yan, David V. Sheehan
52. Cognitive Behavioral Therapy augments the Efficacy of Paroxetine in
Partial Responders with Social Anxiety Disorder
Carlos Blanco, Richard Heimberg, Thomas Rodebaugh, Franklin
Schneier, Debra Ledley, Keng-Han Lin, Brigette Erwin, Michael
Liebowitz
53. Efficacy and Tolerability of 5 mg Lu AA21004 in an 8-Week US Trial of
Adults with Generalized Anxiety Disorder
Anthony J. Rothschild, Atul R. Mahableshwarkar, Paula Jacobsen,
Mingjin Yan, David V. Sheehan
269
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
54. Venlafaxine Treatment of Compulsive Hoarding
Sanjaya Saxena, Jennifer Sumner, Brandon Barrios
55. Alzheimer Disease Trials Simulations to Test New Research Criteria,
Biomarkers, and Other Proposed Methodological Improvements
Lon S. Schneider, Richard E. Kennedy, Gary R. Cutter
56. Evaluation of Mibampator for Agitation and Aggression in Alzheimer’s
Disease
Paula Trzepacz, Thomas Konechnik, Tammy Forrester, Brian Willis,
Catherine Shuler, Linda Tabas, Jeffrey Cummings, Constantine
Lyketsos
57. Medication and Parent Training in Children with Pervasive Developmental
Disorders and Serious Behavior Problems: Effectiveness and Tolerability
of Aripiprazole in Risperidone Nonresponders
Kimberly A. Stigler, Christopher J. McDougle, Laurence Scahill,
Michael G. Aman, L. Eugene Arnold, Cynthia Johnson, Ben Handen,
Ben Vitiello
58. A Placebo-Controlled Trial of Riluzole for Treatment of Childhood-Onset
Obsessive Compulsive Disorder
Lisa Joseph, Paul Grant, Susan Swedo
59. Selegiline Transdermal System (STS) in Patients with Recurrent Unipolar
Major Depression: A Post-Hoc Analysis of 2 Randomized, Double Blind
Studies
Donald Robinson, Kimberly B. Portland
270
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
60. A Translational Approach to Evaluate the Efficacy and Safety of the
Novel AMPA Receptor Positive Allosteric Modulator Org 26576 in Adult
Attention-Deficit/Hyperactivity Disorder
Pilar Cazorla, Rene Kroon, Mohammed Shahid, Frank Tarazi,
Jacques Schipper, Mark Stein, Armin Szegedi, Lenard Adler
61. Magnetic Seizure Therapy (MST): Introduction to a Promising Treatment
for Geriatric Depression in Development
Stefan B. Rowny
62. Some Urban Legends Of CNS Clinical Trial Methodology: Unsuccessful
Solutions to the Problem of Failed Trials
Janet B.W. Williams, Danielle Popp, Scott Reines, Michael J. Detke
63. Randomized Sham Controlled Double-blind Trial of Repetitive
Transcranial Magnetic Stimulation (rTMS) for Adults with Severe
Tourette Syndrome
Angeli Landeros-Weisenberge, Maria Motlagh, Antonio Mantovani,
James F. Leckman, Sarah H. Lisanby
64. A Multi-Center Investigation of Folate plus B12 Supplementation in
Schizophrenia
Joshua L. Roffman, Steve Lamberti, Eric Achtyes, Eric A. Macklin,
Gail Galendez, Lisa Raeke, Noah J. Silverstein, Dan Tuinstra,
Michele Hill, Donald C. Goff
65. MK-6096, a Dual Orexin Receptor Antagonist, improves Subjective
Measures of Sleep and Functioning in Adults with Primary Insomnia
Kathryn M. Connor, Xin Zhao, Erin Mahoney, Saheeda Jackson,
Jill Hutzelmann, Ellen Snyder, Duane Snavely, Jay Pearson, David
Michelson, Darryle Schoepp, Thomas Roth, W. Joseph Herring
271
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
66. Power Spectral Analysis of the Dual Orexin Receptor Antagonist
Suvorexant (MK-4305) in Patients with Primary Insomnia and in Healthy
Subjects
W. Joseph Herring, Junshiu Ma, Ellen Snyder, Vladimir Svetnik, Jill
Hutzelmann, Kenneth Liu, Christopher Lines, Thomas Roth, Darryle
Schoepp, David Michelson
67. Lowered CSF levels of Nociceptin in Females with Fibromyalgia
Syndrome
Lars Tanum, Morten Vinje, Gunnar Ordeberg, Fred Nyberg
68. Naltrexone plus Aripiprazole compared to Naltrexone Alone and Placebo
in the Treatment of Alcohol Dependence - A Double Blind Pilot Study Raymond Anton, Alicia Baros, Patricia Latham, Pat Randall, Scott
Stewart, Derick Vergne, Konstantin Voronin
69. Associations between aspects of Verbal Memory and Brain Structure in
Late Life Depression
Melissa Lamar, Rebecca Charlton, Aifeng Zhang, Anand Kumar
70. Clinical Assessment Methodology for Alzheimer’s Disease Prevention
Trials: A Global and Multi-Axial 2 Year Study of Pre-Mild Cognitive
Impairment (Pre-MCI) Subjective Cognitive Impairment (SCI)
Barry Reisberg, Ricardo Osorio, Asif Khan, Kamalika Roy, Carol
Torossian, Isabel Monteiro, Salman Anwar, Melanie B. Shulman,
Iryna Lobach
272
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
71. The Potent M1 Receptor Allosteric Agonist GSK1034702 improves
Episodic Memory in the Nicotine Abstinence Model of Cognitive
Dysfunction in Humans
Pradeep J. Nathan, Jeannette Watson, Jesper Lund, Gary L. Peters,
Chris M. Dodds, Philip Lawrence, Graham D. Bentley, Barry V. O’Neill, Jonanthan Robertson, Paul Maruff, Marc Laruelle, Edward
T. Bullmore
72. Maternal Pre-Pregnancy Body Mass Index is Associated with ADHD
Symptoms and Impaired Inhibitory Control in 6-9 Year Old Children
Sonja Entringer, Claudia Buss, Elysia P. Davis, Pathik D. Wadhwa,
Curt A. Sandman
73. Influence of COMT val158met on Resting State Functional Connectivity
over Adolescence
Aarthi Padmanabhan, Kai Hwang , David Montez, Beatriz Luna
74. Impulsivity in Bipolar Disorder: Relationships with Neurocognitive
Dysfunction and Substance Use History
Katherine Burdick, Robyn Powers, Serge Sevy, Anil Malhotra
75. Motivational Saliency Signal in Ventral Striatum is Modulated by Genetic
Variation in the ARC Gene Region
Caroline F. Zink, Sam A. Colalillo, David N. Blitzer, M. Ryan
Haynes, Kuan H. Wang, Daniel R. Weinberger
76. Sensory and Motor Contributions to Visuomotor Impairments in
Individuals with Autism Matthew Mosconi, Suman Mohanty, Lauren Schmitt, Edwin Cook,
David Vaillancourt, John Sweeney
273
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
77. Evidence of Neurodevelopment Differences in Prodromal and First
Episode Psychosis Subjects in the Prepulse Inhibition Paradigm
Kristin Cadenhead, Jean Addington, Tyrone Cannon, Barbara
Cornblatt, Thomas McGlashan, Diana Perkins, Larry Seidman, Ming
Tsuang, Elaine Walker, Scott Woods, Robert Heinssen
78. Superior Temporal Gyrus and Frontal 50 and 100 ms Auditory
Abnormalities in Schizophrenia
Yu-Han Chen, J. Christopher Edgar, Mingxiong Huang, Michael A. Hunter, Amanda Olson, Emerson M. Epstein , Jose M. Cañive
79. Contributions of Social Anhedonia and Social Anxiety to Impaired Social
Adjustment in the Putative Schizophrenia Prodrome
Victoria Cressman, Scott A. Schobel, Sara Steinfeld, Judy L. Thompson, Shelly Ben-David, Alex Crumbley, Scott A. Small, Holly
Moore, Cheryl M. Corcoran
80. Functional Activation during Probabilistic Reinforcement Learning in
Schizophrenia: Relationship to Anhedonia/Avolition
Erin C. Dowd, Deanna M. Barch
81. Toxoplasma gondii Exposure affects Neural Processing Speed as
Measured by Acoustic Startle Latency in Schizophrenia and Controls
Brad Pearce, Sydney Hubbard, Hilda N. Rivera, Patricia P. Wilkins,
Marylynn C. Fisch, Wendy Hasenkamp, Robin Gross, Nancy
Bliwise, Jeffrey L. Jones, Erica Duncan
82. The Effects of Cannabis Dependence on Cognitive Function in Males with
Schizophrenia
Rachel A. Rabin, Konstantine K. Zakzanis, Zafiris J. Daskalakis,
Tony P. George
274
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
83. White Matter Disruption in Adolescents with Childhood Maltreatment
History
Uma Rao, Hao Huang, Tejasvi Gundapuneedi
84. Impaired Auditory Object Formation in Schizophrenia as Revealed by
Theta-Gamma Oscillatory Entrainment Dynamics
David I. Leitman, Daniel H. Wolf, Jonathan McDaniel, Yin Li, Julie
Rosen, James Loughead, Raquel E. Gur, Ruben C. Gur, Bruce I. Turetsky
85. Sensory and Cognitively Mediated Event-Related Potentials Index
Symptoms in First Episode Schizophrenia
Elisabetta del Re, Margaret Niznikiewicz , Tracey Petryshen , Robert
W. McCarley
86. Impaired Context Processing as a Potential Marker of Risk in ClinicalHigh-Risk Youth
Tara Niendam, Natalie Hutchison, J. Daniel Ragland, Jong Yoon,
Marjorie Solomon, Michael Minzenberg, Cameron Carter
87. Metacognitive Assessmrent of Self-Awareness in Schizophrenia
Mujeeb Shad, Matcheri Keshavan, Perry Mihalakos, Jair Soares,
Carol Tamminga
88. Deficits of Inhibitory Behavioral Control in Schizophrenia and Bipolar
Disorder
John Sweeney, James Reilly, Richard Keefe, Scot Hill, Mark
Bushong, Carol Tamminga, Matcheri Keshavan, Godfrey Pearlson,
Gunvant Thaker
275
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
89. Effects of the Val(158)Met Catechol-O-Methyltransferase Gene
Polymorphism on Olfactory Processing in Schizophrenia
Vidyulata Kamath, Paul J. Moberg, Raquel E. Gur, Richard L. Doty,
Bruce I. Turetsky
90. Neuroscience-Informed Cognitive Training in Schizophrenia
“Normalizes” Brain-Behavior Associations in Auditory and Verbal
Working Memory Processes: Evidence from MEG and fMRI
Sophia Vinogradov, Karuna Subramaniam, Alex Herman, Josh
Woolley, Tracy Luks, Melissa Fisher, John Houde, Srikantan
Nagarajan
91. Gamma Oscillations to the Gestalt Perception of Coherent and Incoherent
Motion in Schizophrenia Thomas J. Whitford, Shahab Ghorashi, Ryan Mears, Kevin M. Spencer
92. Facial Processing in Bipolar Disorder and Schizophrenia: An EventRelated Potential Study
Jonathan K. Wynn, Kristopher I. Mathis, Carol Jahshan, Lori
Altshuler, David Glahn, Michael F. Green
93. Trajectory of Comorbidities in Obsessive-Compulsive Disorder
Euripedes C. Miguel, Maria A. de Mathis, Juliana B. Diniz, Ana G. Hounie, Victor Fossaluza, James Leckman, David Pauls, Roseli G. Shavitt, Maria C. Rosario
94. The Relationship of Adult Attention Deficit Hyperactivity Disorder to
Anxiety Disorders in a Clinical Sample Michael Van Ameringen, William Simpson, Beth Patterson
276
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
95. Paternal Age and Risk of Bipolar Disorder in Offspring
Alan S. Brown, Ian W. McKeague, YuanYuan Bao, Ling Shen,
Catherine A. Schaefer
96. Health Behaviors Contribute to Arterial Stiffness Later in the Course of
Bipolar Disorder
Simrit Sodhi, Jonathan Linder, William H. Coryell, Del D. Miller,
William G. Haynes, Jess G. Fiedorowicz
97. Maternal Iron Deficiency and Risk of Bipolar and Schizophrenia
Y.-Y. Ho, Y. Bao, I.W. McKeague, L. Shen, C.A. Schaefer, A.S. Brown
98. Sleep Apnea Risk and Clinical Correlates in Patients with Bipolar
Disorder
Isabella Soreca, Jessica C. Levenson, Meredith J. Lotz, Ellen Frank,
David J. Kupfer
99. Examining the Validity of Cyclothymic Disorder in a Youth Sample:
Results from Two Studies
Anna Van Meter, Eric Youngstrom, Jennifer Youngstrom, Norah
Feeny, Christine Demeter, Robert L. Findling
100. Altered Anxiety-Like Behavior in BDNF Val66Met Mice is Rescued with
Early-Life Fluoxetine
Iva Dincheva, Jianmin Yang, Tina Marinic, Kevin Bath, Helena
Frielingsdorf, Theresa Teslovic, James Kocsis, Barbara Hempstead,
Francis Lee
277
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
101. Convergent Functional Genomics of Anxiety Disorders: Translational
Identification of Genes, Biomarkers, Pathways and Mechanisms
Helen Le-Niculescu, Yokesh Balaraman, Sagar Patel, Mikias Ayalew,
Ronald Kuczenski, Anantha Shekhar, Nicholas Schork, Mark Geyer,
Alexander Niculescu
102. Maternal Behavior and DNA Methylation of Nr3c1 and Egr1 Genes Show
Sex Differences and are Altered by Sex Composition of the Litter
Therese A. Kosten, Wen Huang, David A. Nielsen
103. Effect of APOE Genotype on Brain Functional Connectivity during
Episodic Memory Encoding in Healthy Aging
Joseph C. Masdeu, Qiang Chen, Venkata Mattay, Philip Kohn, John
Muse, Bhaskar Kolachana, Lisa Nichols, Daniel R. Weinberger,
Karen F. Berman
104. CDH2 Gene Variants in Obsessive Compulsive Disorder and Tourette
Syndrome
Pablo R. Moya, Jens R. Wendland, Kiara C. Timpano, Liza M. Rubenstein, Zaker Rana, Ruby Fried, Marzena Galdzicka, Edward I. Ginns, Nicholas H. Dodman, Dennis L. Murphy
105. Variation in the Oxytocin Receptor Gene is Associated with Increased
Anxiety in Individuals with a History of Exposure to Early Life Stress
Amanda J. Myers, Leanne Williams, Justine M. Gatt, Erica
McAuley-Clark, Carol Dobson-Stone, Peter Schofield, Charles B. Nemeroff
278
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
106. Resting Dorsal Anterior Cingulate Cortex Metabolism mediates the Effect
of a FKBP5 Haplotype on Posttraumatic Stress Symptoms in Monozygotic
Twins Discordant for Combat Exposure
Roger K. Pitman, Joshua L. Roffman, Lisa M. Shin, Avital M. Fischer, Stephen A. Haddad, Richie Siburian, David G. Brohawn,
Rachel Yehuda, Jordan W. Smoller
107. Sex Differences in PTSD: Dissociation of the Roles for SRD5A2 and
PACAP Polymorphisms in a Highly Traumatized Civilian Population
Charles F. Gillespie, Lynn M. Almli, Bekh Bradley, Kristina B. Mercer, Elisabeth B. Binder, Karen N. Conneely, Joseph Cubells,
Alicia K. Smith, Kerry J. Ressler
108. The Impact of APOE2 Genotype on APOE mRNA Expression, APOE
Protein Level, and the Transcriptome in Human Post-Mortem Cortical
Tissue
Concepcion Goldberg, Thomas M. Hyde, Shufen Chen, Joel E. Kleinman, Peter Davies, Terry E. Goldberg
109. Epigenetic Modulation of Leukocyte Glucocorticoid Receptor in Healthy
Adults: Effects of Childhood Parenting Experiences
Audrey R. Tyrka, Lawrence H. Price, Carmen Marsit, Oakland C. Walters, Linda L. Carpenter
110. Genetic Variation in the Pituitary Adenylate Cyclase-Activating
Polypeptide 1 Receptor, Type I (PAC1) Gene is Associated with Increased
Anxiety and Depressive Symptoms in Adolescent Females with High
Levels of Childhood Stress
Suman Baddam, Rene Olvera, Carolina Livi, Douglas Williamson
279
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
111. Systems Biological Analyses implicate Glutamate Signaling
Abnormalities in Autism and in Intellectual Disability: Implications for
Psychopharmacology
Joseph D. Buxbaum, Avi Ma’ayan, Yan Kuo, Ruth Dannenfelser,
Catalina Betancur
112. Rare Copy Number Variants in Tourette Syndrome Disrupt Genes in
Histaminergic Pathways and Overlap with Autism
Thomas V. Fernandez, Stephan J. Sanders, A. Gulhan ErcanSencicek, Youeun Song, James F. Leckman, Robert A. King, Donald
L. Gilbert, Gary A. Heiman, Jay A. Tischfield, Pieter J. Hoekstra,
Matthew W. State
113. Whole Exome Sequencing of Autism Families Reveals De Novo
Mutations in Integrin-Related Genes
James S. Sutcliffe, Ben M. Neale, Aniko Sabo, Emily L. Crawford,
Nicholas G. Campbell, Ana Carneiro, Eric Boerwinkle, Joseph D. Buxbaum, Edwin H. Cook, Bernie Devlin, Richard A. Gibbs, Gerard
D. Schellenberg, Mark J. Daly
114. COMT Val158Met Variant and Functional Haplotypes Associated with
Childhood ADHD History in Women with Bulimia Nervosa
Allan S. Kaplan, Zeynep Yilmaz, Clement C. Zai, Robert D. Levitan,
James L. Kennedy
115. The Hypo-Functional 7-Repeat Allele of DRD4 predicts both Objective
and Reported Fat Intake in 4- to 6- Year Old Girls
Robert Levitan, Patrícia Silveira, André Portella, James Kennedy,
Hélène Gaudreau, Caroline Davis, Meir Steiner, Claudio Soares,
Stephen Matthews, Laurette Dubé, Michael Meaney
280
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
116. Exploring an Association between Genetic Variation in Lipid Metabolism
Genes and Mental Disorders and Suicide Attempts
Enrique Baca-Garcia, M. Mercedes Perez-Rodriguez, Maria A. Oquendo, Jorge Lopez-Castroman, Concepcion Vaquero-Lorenzo,
Pablo Fernandez-Navarro
117. Interaction between FKBP5 and Childhood Trauma Increases Risk for
Aggressive Behavior
Laura Bevilacqua, Vladimir Carli, Marco Sarchiapone, Danielle K. George, David Goldman, Alec Roy, Mary-Anne Enoch
118. Association of an Intronic Deletion in the Corticotropin-Releasing
Hormone Receptor Gene with Depression in African Americans
Charles B. Nemeroff, Manuel Ramirez-Restrepo, Anzhelika Engel,
Kristina Butze Mercer, Kerry Ressler, Amanda J. Myers
119. The Brain Epigenome: Mapping Brain Relevant Gene Regulatory
Elements using Next Generation Sequencing
Cathy Barr
120. Length of CAG Repeat in Huntingtin Below Disease Threshold Predicts
Volume of Specific Brain Regions
Peggy Nopoulos, Eric A. Epping, Tom Wassink, Bradley Schlaggar,
Joel Perlmutter
121. Daily Meditation in Distressed Dementia decreases NF-kappa B Signaling
and Increases Interferon Response Factor Transcription
Helen Lavretsky, Steve W. Cole, Jesusa M. G. Arevalo, Michael R. Irwin
281
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
122. Linkage Analyses of Twelve Endophenotypes for Schizophrenia from the
Consortium on the Genetics of Schizophrenia (COGS)
Tiffany A. Greenwood, Robert Freedman, Michael F. Green, Raquel
E. Gur, Keith H. Nuechterlein, Ann Olincy, Allen D. Radant,
Nicholas J. Schork, Larry J. Seidman, Larry J. Siever, Neal R. Swerdlow, Debby W. Tsuang, David L. Braff
123. Preliminary Evaluation of Resting-State Functional Connectivity in
Children with Bipolar Disorder or Attention Deficit Hyperactivity
Disorder
Daniel P. Dickstein, Brooke L. Reidy, Matthew F. Pescosolido,
Thania Galvan, Sharad Sikka, Maarten Mennes, Michael P. Milham
124. Serotonin Transporter Binding and Treatment Response to Fluoxetine in
Individuals Recovered from Anorexia Nervosa
Ursula F. Bailer, W. Gordon Frankle, Rajesh Narendran, Michael L. Himes, Chester A. Mathis, Walter H. Kaye
125. Food Motivation Circuitry Dysfunction during Hunger and Satiety: From
Active Anorexia Nervosa to Extreme Obesity
Laura Holsen, Elizabeth Lawson, Cary Savage, Laura Martin,
Amanda Bruce, Rebecca Lepping, Eunice Ko, Justine Blum, Nikos
Makris, Anne Klibanksi, William Brooks, Merlin Butler, Jennifer
Zarcone, Jill Goldstein
126. Differential Activation of Ventral and Dorsal Striatum by Motivational
Value and Motiavtion Salience
Daniel W. Hommer, Ashley Smith, Jodi Gilman, Reza Momenan
282
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
127. Functional and Structural Neuroimaging of Blast mTBI in Iraq and
Afghanistan Veterans
Elaine R. Peskind, Eric C. Petrie, Donna J. Cross, Kathleen
Pagulayan, David Hoff, Kim Hart, Matthew Tarobochia, Murray A. Raskind, Cynthia Mayer, Vasily Yarnykh, Satoshi Minoshima
128. Gonadal Steroid Hormones affect Hippocampal Activation during Spatial
Navigation in Women
Shau-Ming Wei, Philip D. Kohn, J. Shane Kippenhan, Erica B. Baller, Gabriela Alarcon, Peter J. Schmidt, Karen F. Berman
129. A Study of D2/D3 Dopamine Receptor Distribution in a Rare Form of
Tourette Syndrome using [11C]PHNO and Positron Emission Tomography
Jean-Dominique Gallezot, Wendol Williams, Keunpoong Lim,
Minq-Qiang Zheng, Erin Loring, Richard E. Carson, John Krystal,
Matthew State, Yu-Shin Ding
130. Free-Water Atlas Application to Neuro-Inflammation in Mild Traumatic
Brain Injury
Martha E. Shenton, Pasternak Ofer, Bouix Sylvain, Pelavin Paula,
Zafonte Ross, Kubicki Marek
131. Neural Correlates of Reappraisal Training to Reduce Negative Emotional
Reactivity in Borderline Personality Disorder
Harold W. Koenigsberg, Bryan Denny, Jin Fan, Xun Liu, Sarah Jo
Mayson, Stephanie Guerreri, Kevin N. Ochsner, Antonia S. New,
Marianne Goodman, Larry J. Siever
132. Distinct Patterns of Altered Default Mode Functional Connectivity in
Borderline and Schizotypal Personality Disorders
Daniel R. Rosell, Maarten Mennes, King-Wai Chu, Larry J. Siever,
Erin A. Hazlett
283
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
133. Relationship of Symptom Domains and Diagnostic Severity to PET Scan
Imaging in Borderline Personality Disorder
S. Charles Schulz, Jazmin Camchong, Ann Romine, Michael
Kuskowski, Jose V. Pardo, Kathryn Cullen, Kelvin O. Lim
134. Resting State Functional Connectivity in 22Q11.2 Deletion Syndrome, a
Recurrent Genetic Mutation Associated with Schizophrenia
Matthew Schreiner, Katherine Karlsgodt, Maria Jalbrzikowski,
Jennifer Ho, Carolyn Chow, Carrie Bearden
135. Preparing for Conflict: Increased Activation of the Dorsal Striatum in
Subjects at Ultra-High Risk for Psychosis
Tiziano Colibazzi, Guillermo Horga, Zhishun Wang, Pengwei Wang,
Kristin Klahr, Cheryl Corcoran, Bradley Peterson
136. Altered Language Network Activity in Young People at Genetic High-Risk
for Schizophrenia
Heidi W. Thermenos, Susan W. Gabrieli, Gul A. Jabbar, Richard
Juelich, John D.E. Gabrieli, Martha Shenton, Marek Kubicki, Alan
Francis, Duwors Robert, Matcheri S. Keshavan, Larry J. Seidman,
Lynn E. DeLisi
137. Alterations of Reward Anticipation in Initially Antipsychotic Naïve
Schizophrenia Patients Before and After Antipsychotic Monotherapy with
Amisulpride
Mette Nielsen, Egill Rostrup, Sanne Wulff, Henrik Lublin, Shitij
Kapur, Birte Glenthoj
138. Decreased Short-Distance fMRI Connectivity, Functional Pruning and
Network Randomization in Childhood-Onset Schizophrenia
Aaron F. Alexander-Bloch, Jay Giedd, Ed Bullmore, Nitin Gogtay
284
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
139. Associations between Cannabis Use and Longitudinal Structural Brain
Changes in Biological Relatives of Schizophrenia Patients
Beng-Choon Ho
140. A Functional MRI Study of the Nerocognitive Effect of Quetiapine
Compared to Haloperidol in Schizophrenia Hisham M. Ibrahim, C. Munro Cullum, Robert W. Greene, Carol A. Tamminga, Carol A. Tamminga
141. Antipsychotic Treatment Enhances the Cortical Substrate for Attention and
Behavioral Planning in First Episode Schizophrenia Sarah K. Keedy, James L. Reilly, Margret S.H. Harris, Tin Khine,
Sunil Shreshtha, Cherise Rosen, Robert Marvin, Peter J. Weiden,
John A. Sweeney
142. White Matter Geometry and Gender Effects in Adolescent-Onset
Schizophrenia
Marek Kubicki, Peter Savadjiev, Morgan E. Hough, Thomas
Whitford, Martha E. Shenton, Tim J. Crow, Anthony C. James
143. 18-F-Fallypride Binding Potential and Age in Patients with Schizophrenia
Monte Buchsbaum, Douglas Lehrer, Brad Christian, Jogeshwar
Mukherjee
144. Effects of a Novel H3 Antagonist on fMRI Activation during Working
Memory in Schizophrenia
Daniel Wolf, James Loughead, Kosha Ruparel, Janina Seubert, Mark
Elliott, Christian Kohler, Bruce Turetsky, Ruben Gur, Raquel Gur
285
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
145. Brain Circuits that Link Schizophrenia to High Risk of Cigarette Smoking
Lauren V. Moran, Hemaltha Sampath, Yihong Yang, Thomas J. Ross,
Gunvant K. Thaker, Elliot A. Stein, L. Elliot Hong
146. Water and Metabolite Transverse T2 Relaxation Time Abnormalities in the
White Matter in Schizophrenia
Fei Du, Alissa Cooper, Bruce Cohen, Perry Renshaw, Dost Ongur
147. Mechanisms underlying Hippocampal Dysfunction in Schizophrenia and
Related Psychotic Disorders
Scott A. Schobel, Nashid Chaudhury, Cheryl M. Corcoran, Martin
Styner, Beatriz Paniagua, Jeffrey A. Lieberman, Holly Moore, Scott
A. Small
148. Proton Spectroscopy Studies in Young Relatives at Genetic High Risk for
Schizophrenia: Evidence for Glutamatergic Alterations
Neeraj Tandon, Alan N. Francis, J.N. Stout, Kunal M. Sanghavi,
Vaibhav A. Diwadkar, Nicolas R. Bolo, Jeffrey A. Stanley, Matcheri
S. Keshavan
149. Dissociable Effects of NRG3 Genotype on Prefrontal Cortex Physiology
in Healthy Volunteers, Schizophrenia Patients, and Their Unaffected
Siblings
Heike Tost, Joseph H. Callicott, Radhakrishna Vakkalanka,
Katherine Gambale, Roberta Rasetti, Venkatta S. Mattay, Daniel R. Weinberger, Amanda J. Law
150. Greater Neuronal Response during Automatic Semantic Processing in
Schizophrenia
Jason R. Tregellas, Lisa B. Wilson, Shireen Shatti, Donald C. Rojas
286
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
151. Neuroinflammation in Temporal Cortex in Patients with Schizophrenia
Measured with (R)-[11C]PK11195 and PET
Bart N. van Berckel, Thalia van der Doef, Maqsood Yaqub, Matthijs
M. Bossong, Ronald Boellaard, Neeltje E. van Haren, Bert D. Windhorst, Adriaan A. Lammertsma, Wiepke Cahn, René S. Kahn
152. Subtyping Schizophrenia using a Multimodal Neuroimaging Approach
Aristotle Voineskos, George Foussias, Jason Lerch, Tarek Rajji,
Nancy Lobaugh, Gary Remington, James Kennedy, Bruce Pollock,
Benoit Mulsant
153. Neural Compensation to Maintain Cognitive Performance in
Schizophrenia
Derin Cobia, Mathew Schroeder, Katherine Blizinsky, John
Csernansky, Lei Wang
154. A Novel High Resolution fMRI Method to Measure Substantia Nigra
Function Reveals Up-Regulation of its Activity during Conditions
Requiring Greater Cognitive Control
Jong H. Yoon, Anthony Grandelis, Christian La, Steffan Soosman,
Cameron S. Carter, Michael J. Minzenberg
155. Brain Network Analysis and Global-Local Information Processing in Body
Dysmorphic Disorder
Jamie D. Feusner, Donatello Arienzo, Jesse Brown, Johnson
GadElkarim, Liang Zhan, Alex Leow
156. The 5-HT2A Receptor and Serotonin Transporter in Ecstasy Users: A PET
Study With [11C]MDL 100907 and [11C]DASB
Ragy R. Girgis, Nina B. Urban, Peter Talbot, Lawrence S. Kegeles,
Judy L. Thompson, Xiaoyan Xu, W. Gordon Frankle, Mark Slifstein,
Anissa Abi-Dargham, Marc Laruelle
287
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
157. Insula Activation during Inhibitory Control Mediates the Effect of
GABRA2 Genotype on Anxiety in a Family Sample Enriched for
Alcoholism
Mary M. Heitzeg, Sandra Villafuerte, Barbara J. Weiland, Preeti G. Samudra, Margit Burmeister, Robert A. Zucker, Jon-Kar Zubieta
158. Differential Effects of an NMDA Receptor Antagonist on Response
Inhibition-Related fMRI Activity in Individuals With and Without a
Family History of Alcoholism
Sharna Jamadar, Rachel Jiantonio, Shashwath Meda, Michael
Stevens, Elise DeVito, Marc Potenza, John Krystal, Godfrey
Pearlson
159. Association between CHRNA5 Genetic Variation at RSL6969968 and
Brain Reactivity to Smoking Images in Nicotine Dependent Women
Amy C. Janes, Jordan W. Smoller, Sean P. David, Blaise de B. Frederick, Stephen Haddad, Aditi Basu, Maurizio Fava, A. Eden
Evins, Marc J. Kaufman
160. Functional Connectivity Changes in Stimulant Abusers during Abstinence
Jazmin Camchong, Sheila Specker, Valerie Slaymaker, Angus
MacDonald, Bryon A. Mueller, Chris Bell, Ann L. Person, Kelvin O. Lim
161. Monoamine Oxidase a Binding during Acute Alcohol Withdrawal: A [11C]
Harmine PET Study
Brittany A. Matthews, Stephen J. Kish, Pablo M. Rusjan, Alan A. Wilson, Isabelle Boileau, Sylvain Houle, Jeffrey H. Meyer
288
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
162. Pharmacological MRI of D-amphetamine: Effects on a Go-No/go Task in
Cocaine Dependent Subjects
F. Gerard Moeller, Joel L. Steinberg, Scott D. Lane, Liangsuo Ma,
Thomas R. Kosten, Ponnada A. Narayana
163. Methylphenidate enhances Executive Function while Optimizing
Prefrontal Error-Related Processing in Both Health and Cocaine Addiction
Scott J. Moeller, Patricia A. Woicik, Dardo Tomasi, Nora D. Volkow,
Rita Z. Goldstein
164. Assessment of Brain Nicotine Accumulation during Cigarette Smoking: A
PET Study with 11C-Nicotine Cigarettes Alexey G. Mukhin, Yantao Zuo, Sudha Garg , Rachid Nazih, Behm
M. Frederique, Pradeep K. Garg , Jed E. Rose
165. History of Early Childhood Trauma is Positively Associated with Ventral
Striatal Dopamine Responses to Amphetamine Lynn M. Oswald, Gary S. Wand, Dean F. Wong, Hiroto Kuwabara,
James Brasic, Wichana Chamroonrat, Maria Guevara, Eram Zaidi
166. Regional Brain Structural Dysmorphology in Chronic Alcoholism, HIV
Infection, and their Comorbidity
Adolf Pfefferbaum, Margaret J. Rosenbloom, Torsten Rohlfing,
Edith V. Sullivan
167. Effects of Smoking Cessation on Reward Processing
Lena Rademacher, Katja N. Spreckelmeyer, Sören Krach, Susanne
Prinz, Ingo Vernaleken, Gerhard Gründer
289
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
168. Methamphetamine Induced Signal Change in Rats: An fMRI Study
Carmela M. Reichel, Xun Zhu, Saeid Taheri, Xingju Nie, Jane E. Joseph, Ronald E. See
169. Acute Baclofen’s Impact on Fronto-Limbic Connectivity: An Aid to
Modulate Negative Affect in Smokers?
Jesse J. Suh, Teresa R. Franklin, Ronald N. Ehrman, Kanchana
Jagannathan, Zachary D. Singer, Kathleen Marquez, Anita Hole,
Marina Goldman, Shing C. B. Lam, Ze Wang, Yin Li, Charles P. O’Brien, Anna Rose Childress
170. Elucidating Functional Relationships between Ventral Striatal Dopamine
Release and Prefrontal Cortical-Striatal Activity in Human Reward
Circuitry with fMRI and [11C]Raclopride PET: New Data
Michael J. Travis, Erika Forbes, N. Scott Mason, Jorge R. C. Almeida, Natalie D. Velasquez, Eric E. Rodriguez, Michael L. Himes, W. Gordon Frankle, Rajesh Narendran, Mary L. Phillips
171. Brain Dopamine Responses to the Expectation of Methylphenidate in
Non-Drug Abusing Subjects
Gene-Jack Wang, Nora Volkow, Tomasi Dardo, Frank Telang,
Christopher Wong, Jean Logan, Millard Jayne, Joanna Fowler
172. Low Striatal Dopamine Receptor Availability Linked to Weight Gain and
Caloric Intake during Abstinence from Chronic Methamphetamine Abuse
Todd Zorick, Mark A. Mandelkern, Timothy Fong, Dara G. Ghahremani, Chelsea Robertson, Brittany Sumerel, Edythe D. London
173. Objective Measurement of Postural Sway in Blast-Related Traumatic
Brain Injury
Brent G. Nelson, Scott R. Sponheim, Kelvin O. Lim
290
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
174. Copy Number Variation and Induced Pluripotent Stem Cells in
Schizophrenia: First Results from the Mount Sinai Conte Center LargeScale Fibroblast Studies identifies Known Recurrent CNVs
Joseph I. Friedman, Nagahide Takahashi, Guiqing Cai, Elodie
Drapeau, Laura Gouder, Andrea Matho, Jared Cooper, Emily
D’Antonio, Puneet Dabas, Marina Goldenberg, Gayle Myerson,
Vladan Novakovic, Kenneth L. Davis, Joseph D. Buxbaum
175. In vivo MicroRNA Detection and Quantitation in Cerebrospinal Fluid
Juan A. Gallego, Kierstyn Claycomb, Mahima Bhatt, Todd Lencz,
Anil K. Malhotra
176. Predictors of Inaccurate Self Assessment of Everyday Functioning in
Clinically Stable Outpatients with Schizophrenia
Samir Sabbag, Thomas Patterson, Robert Heaton, Philip Harvey
177. A Dimensional Approach to the Continuum between Bipolar Disorder
with Psychosis and Schizophrenia: The Schizo-Bipolar Scale
Matcheri Keshavan, David Morris, John Sweeney, Godfrey Pearlson,
Gunvant Thaker, Carol Tamminga, Larry Seidman, Shaun Eack
178. Ketamine for Treatment-Refractory Obsessive-Compulsive Disorder
Michael H. Bloch, James F. Leckman, John H. Krystal, Zubin H. Bhagwagar, Gerard Sanacora, Christopher Pittenger
179. Early Life Stress produces Immediate and Sustained Effects on
Corticolimbic Endocannabinoid Signaling Matthew N. Hill, Lisa Eiland, Tiffany T.Y. Lee, Cecilia J. Hillard,
Bruce S. McEwen
291
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
180. Double Blind Randomized Brief Treatment with Escitalopram associated
with Extinction Learning Facilitation in Healthy Subjects
Eric Bui, Scott Orr, Ryan J. Jacoby, Nicole LeBlanc, Mark Pollack,
Aparna Keshaviah, Naomi Simon
181. Assessing the Long-Term Safety and Tolerability of Desvenlafaxine in
Child and Adolescent Outpatients with Major Depressive Disorder
Robert L. Findling, James Groark, Deborah Chiles, Sara Gardiner,
Lingfeng Yang, Karen A. Tourian
182. Effects of Serotonin Augmentation on Test Meal Food Intake in Anorexia
Nervosa
Barbara E. Wolfe, Evgeniy E. Filin, Amy Silver-Ritter, David C. Jimerson
183. Treatment of Pathological Gambling with Tolcapone, a Catechol-Omethyl-transferase (COMT) Inhibitor
Jon E. Grant, Brian L. Odlaug, Suck Won Kim
184. The Relationship between Serum Lithium Concentrations and Clinical
Response in Bipolar Depression Caleb M. Adler, Tyler L. Thompson, Melissa P. DelBello, Thomas J. Blom, David E Fleck, Stephen M
185. Lack of Tyramine Pressor Response Effect with Oral CX157, a Reversible
MAOI
Daniel J. Burch, Mahnaz Asgharnejad, William A. Gerson
186. How Useful is Intravenous Ketamine for Depression in the ‘Real World’?
David Feifel, Kai MacDonald, Michael Messer, Luis Giuffra
292
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
187. Course of Improvement in Depressive Symptoms to a Single Intravenous
Infusion of Ketamine vs. Add on with Riluzole: Results from a FourWeek, Double-Blind, Placebo-Controlled Study
Jose A. Franco-Chaves, Lobna Ibrahim, Nancy DiazGranados,
Nancy Brutsche, Philip Kronstein, David A. Luckenbaugh, Husseini
K. Manji, Carlos A. Zarate Jr.
188. Changes in Suicide Risk and Depressive Symptoms with Low Dose
Lithium Combined with Citalopram Compared to Citalopram with
Placebo in a Group of Severely Ill Depressed Patients
Arif Khan, Shirin R.F. Khan, Joy Hobus, James Faucett, Vishaal
Mehra, Earl L. Giller, Richard L. Rudolph, Walter A. Brown
189. Interferon-Stimulated Genes: Antiviral Response and Psychiatric Side
Effects of IFN-alpha in HCV Patients
Gregory Oxenkrug, Paul Summergrad
190. Longstanding Cannabis Abuse is Associated with Decreased Likelihood of
Remission from Major Depression
Ihsan M. Salloum, Jack R. Cornelius , Antoine Douaihy , Ricardo
Caceda , Feng Miao, Kirisci Levent, Thase E. Michael
191. Comparative Efficacy and Tolerability of Quetiapine Monotherapy in
Major Depression Angelos Halaris, John Piletz, Edwin Meresh, Steven Kimmons,
James Sinacore
192. Brain Volume Changes in Adult Bipolar Depression Patients with Lithium
Treatment
Abhishek Lunagariya, Mark Nicoletti, Giovana Zunta Soares, John
Hatch, Jair Soares
293
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
193. Efficacy of Adjunctive OPC-34712 across Multiple Outcome Measures in
Major Depressive Disorder: A Phase II, Randomized, Placebo-controlled
Study
Michael Thase, Maurizio Fava, Mary Hobart, Aleksandar Skuban,
Peter Zhang, Robert D. McQuade, William H. Carson, Raymond
Sanchez, Robert A. Forbes
194. N-methyl-D-aspartate Mechanism of Depression and a Glycine
Transporter-I Inhibitor, N-Methylglycine (Sarcosine) for the Treatment of
Depression
Guochuan Emil Tsai, I-Hua Wei, Chih-Chia Huang, Kuo-Hao
Huang, Hsien-Yuan Lane
195. The Effects of Highly Active Antiretroviral Therapy (HAART) on
Neurocognitive Functioning of Patients with HIV
Tanya Alim, Mansoor Malik, Ziad Safadi, Anne Marie Duc, Maria
Hipolito
196. Metabotropic Glutamate Receptor 4 (mGluR4) Activation reverses Acute
Motor Deficits and Protects against Neurodegeneration in Animal Models
of Parkinson’s Disease Danielle Graham, Gael Hedou, Audrey Gray, Jeff Pimentel, Dongzi
Yu, Nicholas Clark, Hui Tian, Mark Shearman, Christoph Wiessner,
Tammy Dellovade
197. Effect of 5HT2c Receptor Blockade in a Novel Social Approach Test of
Anxiety-Like Behavior during Withdrawal from Chronic Intermittent
Ethanol
Catherine A. Marcinkiewcz, Alberto J. Lopez, Nora M. McCall,
Alexa M. Waters, Sheryl S. Moy, Thomas L. Kash
294
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
198. The Neurosteroid Pregnenolone prevents Predator Stress-Induced
Decreases in Open Arm Time in the Elevated Plus Maze Model for
Anxiety-Like Behaviors: Potential Relevance to Novel Secondary
Prevention Strategies in PTSD?
Shawn K. Acheson, Jason D. Kilts, Joshua T. Rogers, Lawrence J. Shampine, Christine E. Marx
199. Adolescent Rats are Insensitive to Fluoxetine: Role of Terminals and
Receptors
Cynthia Kuhn, Andrew Arrant
200. Identification of a New Therapeutic Intervention that Ameliorates
Behavioral Deficits in a Mouse Model of Fragile X Syndrome and Autism
Spectrum Disorder
Marjelo A. Mines, Christopher J. Yuskaitis, Margaret K. King,
Eleonore Beurel, Richard S. Jope
201. The Intrinsic Rewarding Properties of Pramipexole in Rats: Comparison
with L-DOPA, Effects on Risk-taking and Associative Learning, and
Alterations Following Dorsal Striatal Dopamine Deafferentation
T. Celeste Napier, Stephanie Tedford, Gregory Ruber, Sandra
Rokosik
202. Noradrenergic Function and Impulsivity in ASPD and Healthy Controls
Alan C. Swann, Marijn Lijffijt, Scott D. Lane, Joel L. Steinberg, F. Gerard Moeller
203. Dopamine D4 receptors Influence “Near-Miss”-like Errors on a Rodent
Slot Machine Task
Paul J. Cocker, Robert D. Rogers, Bernard Le Foll, Catharine A. Winstanley
295
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
204. Allosteric Modulators, Atypical and Constrained Agonists of the TRPV1
Ion Channel
Michael J. Iadarola, Krisztian Kaszas, Hong-Tao Ma, Jason M. Keller, Theresa R. Lii, Steven A. Titus, Melanie Bryant, Marc Ferrer,
Erika Englund, Juan Marugan, Owen B. Mcmanus, Noel Southall
205. Pharmacological and Behavioral Characterization of the Norepinephrine
and Dopamine Reuptake Inhibitor EB-1020: A Novel Pharmacotherapy for
ADHD
Frank I. Tarazi, Yong Kee Choi, Krystyna Gołembiowska,
Magdalena Kowalska, Frank P. Bymaster
206. Individual Differences in Inherent Impulsivity Track with CocaineSeeking Behavior and Serotonin (5-HT) 2C (5-HT2C) Receptor mRNA
Editing in the Corticoaccumbens Circuit
Noelle C. Anastasio, Robert G. Fox, Sonja J. Stutz, F. Gerard
Moeller, Ronald B. Emeson, Kathryn A. Cunningham
207. Glycosylation of Neuroactive Peptides to Enhance CNS Bioavailability
Edward Bilsky, Torsten Falk, Scott Sherman, Jim Cormier, Lindsay
St. Louis, Denise Giuvelis, Robin Polt, Glenn Stevenson
208. 1H-MRS Analysis of Neurochemical Profiles after Acute Ethanol in Adult
Mice Lacking Adenylyl Cyclase Isoforms 1 and 8
Matthew P. Galloway, Farhad Ghoddoussi, Nadeem Sawaf, H. Michael Marsh, Alana C. Conti
209. Synergistic Reduction of Cocaine Actions in Mice Treated with Cocaine
Hydrolase and Anti-Cocaine Antibodies or Vaccine
Yang Gao, Xiaoyun Shen, Frank Orson, Berma Kinsey, Tom Kosten,
Marilyn E. Carroll, Stephen Brimijoin
296
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
210. Effect of Adolescent vs. Adult Delta-9-THC Exposure on Neurocognitive
Function in Rats
Loren H. Parsons
211. Sex Differences in Fear Conditioning in Posttraumatic Stress Disorder
Sabra Inslicht, Thomas Metzler, Mohammed Milad, Suzanne
Pineles, Scott Orr, Charles Marmar, Thomas Neylan
212. Influence of Selective Serotonin Reuptake Inhibitors on Fear Extinction
Kelimer Lebron-Milad, Alina Tsareva, Roger K Pitman, Mohammed
R. Milad
213. Sleep in PTSD vs. Controls - Evidence for Possible Gender Dimorphism
Anne Richards, Thomas J. Metzler, Maryann Lenoci, Leslie Ruoff,
Thomas C. Neylan
214. Fetal Programming of Sex Differences in Stress Response Circuitry,
Endocrine and ANS Deficits in Adulthood: Implications for Understanding
Sex Differences in Comorbidity of Depression and CVD Risk
Jill M. Goldstein, Laura M. Holsen, Brandon Abbs, Sara
Cherkerzian, Anne Klibanski, Mady Hornig, Richard Sloan, Stephen
Buka
215. Sleep Abnormalities in Women with Bipolar Disorder may be Mediated
through Abnormal Response to Light Exposure and Bedtime Cortisol
Erika F.H. Saunders, Julio Fernandez-Mendoza, Masoud Kamali,
Scott Langenecker, Edward Bixler, Alexandros Vgontzas, Melvin
McInnis, Alan Gelenberg
297
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
216. Impulsivity as a Consequence or a Predictor of Cocaine use in Male and
Female Smoked Cocaine Users
Stephanie Collins Reed, Richard W. Foltin, Eric Rubin, Suzette M. Evans
217. Social and Environmental Enrichment alter the Effects of Marijuana on
Cocaine Reward in Male and Female Adolescent Rats
Sari Izenwasser, Amy K. Starosciak, Amanda Tristan, Shervin
Liddie, Jannifer Matos
218. The Role of Orexin in Adverse Menopause-Associated “Hot Flash” and
Anxiety Symptoms
Philip Johnson, Lauren Federici, Stephanie Fitz, Todd Skaar, Janet
Carpenter, Anantha Shekhar
219. The Impact of Placebo on IL-18 and its Relation to Analgesic Expectation
and Central µ-Opioid Receptor Activation
Alan Prossin, Steven Zalcman, Alisa Koch, Phillip Campbell, JonKar Zubieta
220. A Frontoparietal Cortical Network in Humans that is Preferentially
Responsive to Approaching Faces
Daphne J. Holt, Garth Coombs, Brittany S. Cassidy, Xiaomin Yue,
Scott L. Rauch, Shahin Nasr, Roger B.H. Tootell
221. Midbrain Dopamine Synthesis modulates Sustained BOLD and Transient
Gamma Oscillatory Neural Response to Facial Emotional Dynamics
Mbemba Jabbi, Tiffany Nash, Philip kohn, Angela Ianni, Dani
Rubinstein, Tom Holroyd, Frederick Carver, Jonathan Shane
Kippenhan, Richard Coppola, Karen Faith Berman
298
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
222. Dual Receptor Antagonism: How Monoamines May Modulate Brain
Activity
Jacob Geday, Albert Gjedde
223. Cognitive Deficits produced by Impaired Glutamate Release from
Astrocytes: Potential Implications of Diminished System xc- Activity to
Addiction and Schizophrenia
Victoria Lutgen, Aric Madayag, Peter Dietrich, Krista Qualmann,
David A. Baker
224. Cholinergic Pathways and Cognition in Patients with Schizophrenia
Tarek K. Rajji, Tiffany W. Chow, Aristotle N. Voineskos, Kira A. Links, Dielle Miranda, David C. Mamo, Zahinoor Ismail, Bruce G. Pollock, Benoit H. Mulsant
225. Effective Connectivity of AKT1-Mediated Dopaminergic Working
Memory Networks and its Relationship to the Pharmacogenetics of
Cognition in Schizophrenia
Hao Yang Tan, Anthony G. Chen, Bhaskar Kolachana, Jose A. Apud,
Venkatta S. Mattay, Joseph H. Callicott, Qiang Chen, Daniel R. Weinberger
226. Somatosensory Timing Deficits in Schizophrenia
Peter Teale, Bryce Pasko, Dan Collins, Don C. Rojas, Martin Reite
227. Anterior Cingulate Cortex Controls Stopping during Self-Limiting
Behavior
Benjamin Y. Hayden, John M. Pearson, Michael L. Platt
299
ACNP 50th Annual Meeting • Final Program
Poster Session II—Tuesday
228. The Glial Modulator Propentofylline impairs Reinstatement in a Rat
Model of Cocaine Abuse
Kathryn J. Reissner, Peter W. Kalivas
229. Dopamine Release in the Nucleus Accumbens Shell is Modulated by
Conditioned Odor Cues that have been Associated with Access to or
the Absence of Alcohol: A Bidirectional Mechanism Underlying the
Anticipation/Craving of Alcohol Reward
Gerald A. Deehan, Zheng-Ming Ding, Sheketha R. Hauser, William
A. Truitt, William J. McBride, Zachary A. Rodd
300
ACNP 50th Annual Meeting • Final Program
Poster Session III – Wednesday
1.
BK Channel β1 Subunit modulates Ethanol Drinking and Behavioral
Adaptations to Chronic Ethanol Exposure
Candice Contet, David Le, Amanda J. Roberts, Steven N. Treistman,
George F. Koob
2.
Long-Term, 96-hour Methamphetamine Self-Administration in Rats: A
Preclinical Model of Human Methamphetamine Addiction
Nicholas E. Goeders, Glenn F. Guerin, Elyse M. Cornett
3.
Essential Role for Fragile-X Mental Retardation Protein (FMRP) in
Normal Behavioral and Neuronal Morphological Adaptations following
Repeated Cocaine Administration
Laura N. Smith, Makoto Taniguchi, Karen C. Dietz, Miles R. Fontenot, Benjamin C. Zirlin, Kimberly M. Huber, Christopher W. Cowan
4.
A Preclinical Examination of Behavioral and Neurotransmitter Specific
Roles for the Ventral Tegmental Area in Reinforcer-Seeking and Drinking
Cristine L. Czachowski
5.
Low Sensitivity or Level of Response to Ketamine Predicts High Alcohol
Intake in Adolescent Rhesus Monkeys
James D. Higley, Andrea Sorenson, Bobbi S. Padro, John P. Capitanio, Lindell Stephen, Barr S. Christina, Stephen J. Suomi
6.
Altered Levels of Nicotinic Acetylcholine and Metabotropic Glutamate
Receptors Associated with Cue-Induced Nicotine-Seeking Behavior in a
Rat Model of Drug Relapse
Xiu Liu, Beata Karolewicz, Courtney Jernigan
301
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
7.
Gene X Early Environment Interactions determine Prefrontal Cortex DNA
Methylation Status and Drug-Seeking in Adult Mice
Tod E. Kippin, Kevin J. Dudley, Joannalee C. Campbell, Kyle L. Ploense, Xiang Li, Wei Wei, Dennis K. Gascoigne, John S. Mattick,
Karen K. Szumlinski, Tim W. Bredy
8.
Novelty Seeking mediates Early Life Stress Effects on Psychostimulant
Place Preference Conditioning in Monkeys
David M. Lyons, Christine L. Buckmaster, Alan F. Schatzberg
9.
Roles for Beta-2 Adrenergic Receptor Activation in the BNST and
CRF-R1 Receptor Activation in the VTA in Stress-Induced Reinstatement
of Cocaine Seeking Following Long-Access Self-Administration in Rats
John R. Mantsch, Jordan M. Blacktop, Yazmin Figueroa-Guzman,
Oliver Vranjkovic
10. Relapse to Cocaine-seeking Normalizes Elevated Phospho-CREB and
Phospho-Ser845-GluR1 in the Prefrontal Cortex and Phospho-Ser9Synapsin in Nucleus Accumbens after One Week of Abstinence
Wei-Lun Sun, Jacqueline F. McGinty
11. Female Vulnerability to Episodic and Continuous Social Stress: Dopamine
and Cocaine Self-Administration
Akiko Shimamoto, Elizabeth N. Holly, Joseph F. DeBold, Klaus A. Miczek
12. Brain-Regional Neuroadaptations in SK Channels following Chronic
Alcohol Exposure and Withdrawal
Patrick J. Mulholland, William C. Griffin III, Howard C. Becker
302
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
13. Double-Dissociation in the Control over the Acquisition and Performance
of Cocaine Seeking by the Dorsomedial and Dorsolateral Striatum
Jennifer E. Murray, David Belin, Barry J. Everitt
14. A Mutation in the Circadian Gene CLOCK increases the Vulnerability for
Cocaine Addiction in Mice
Angela Ozburn, Erin Larson, David Self, Colleen McClung
15. Protracted Withdrawal from Cocaine Self-Administration Flips the Switch
on 5-HT1B Receptor Modulation of Cocaine Reinforcement
Nathan S. Pentkowski, Timothy H.C. Cheung, William A. Toy,
Matthew Adams, John F. Neumaier, Janet L. Neisewander
16. Alpha-2 Adrenergic Agonists and CRF Antagonist Block the Negative
Emotional Signs of Naloxone-Precipitated Morphine Withdrawal in Rats
Susan B. Powell, Clay Archer, Colleen Lonergan, Navarre GutierrezReid, Zhongqi Zhang, Eric P. Zorrilla, George F. Koob, Gerhard
Schulteis
17. Electrical Stimulation of the Granular Insular Cortex attenuates NicotineTaking and -Seeking Behavior
Abhiram Pushparaj, Clement Hamani, José N. Nobrega, Bernard Le
Foll
18. Prazosin Blocks Development of Ethanol, Morphine and Cocaine
Conditioned Place Preference in Mice
Murray A. Raskind, Van A. Redila, Valerie G. Olson
19. Dopamine D1 Receptor Signaling Through Gαq: A Behavioral Assessment
Gregg D. Stanwood, Tommy P. Saborido, Roger J. Colbran, Aliya L. Frederick
303
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
20. A History of Cocaine Intake produces Persistent Reductions in the
Functional Integrity of Glial Cells in Forebrain
Sierra M. Webb, Arianne D. Sacramento, Osnat Ben-Shahar, Tod E. Kippin, Karen K. Szumlinski
21. Interaction between Ethanol and Nicotine within the Mesolimbic
Dopamine System: Evidence for Synergy and Distinct Alterations in Gene
Expression
William A. Truitt, Sheketha R. Hauser, Gerald A. Deehan, ZhengMing Ding, Anantha Shekhar, William J. McBride, Zachary A. Rodd
22. A Premorbid Depressive State Facilitates the Development of
Dependence-like Cocaine Intake in Rats with Extended Access
Sunmee Wee, George F. Koob
23. Valproic Acid reverses Age-Related decreases in Haloperidol-Induced
c-Fos Expression in the Nucleus Accumbens
Janitza L. Montalvo-Ortiz, Keely M. Murphy, John G. Csernansky,
Hongxin Dong
24. 5-HT2AR and MGluR2 Play Opposing Roles in Regulating Anxiety
Caitlin E. McOmish, Noelia V. Weisstaub, Jay A. Gingrich
25. Neurotropic Viruses and Induced Pluripotent Stem Cells: Tools for
Probing Neurodevelopment
Leonardo D’Aiuto, Brianna Heath, Annie Watson, Bamne Mikhil,
Roberto Di Maio, Giorgio Raimondi, Vishwajit Nimgaonkar
26. AMPA Receptor mediated Mitogen-Activated Protein Kinase Signaling in
Oligodendrocytes
Tara M. DeSilva, Brandi J. Baker, Adam Funk
304
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
27. Proinflammatory Gene Expression in Response to Stressors Differs across
Brain Regions and is Programmed by Maternal Diet
Nicola M. Grissom, Robert George, Teresa M. Reyes
28. Neurexin 1 (NRXN1) Gene Expression across the Human Lifespan:
Implications for Neurodevelopment and Schizophrenia
Aaron Jenkins, Yanhong Wang, Thomas M. Hyde, Joel E. Kleinman,
Amanda J. Law
29. ZNF804a Regulates Transcription of the Schizophrenia-Associated Genes
PRSS16, COMT, NRG1, PDE4B, and DRD2
Matthew J. Girgenti, Joseph J. LoTurco, Brady J. Maher
30. Adenosine A2A Receptor Mediated Signaling in the Dorsomedial Striatum
in Mice Lacking ENT1 and Alcoholism
Hyung Wook Nam, David Hinton, Sun Choi, Nayoung Kang, DooSup Choi
31. MicroRNA-132 drives Compulsive Cocaine Use through Nuclear FactorκB Signaling
Heh-In Im, Purva Bali, Paul Kenny
32. Chronic Wheel Running reduces Compulsive Methamphetamine Drug
Seeking
Alex Englemann, Mark Aparicio, Cameron McKay, Airee Kim,
Jeff Sobieraj, Yanabel Grant, Sunmee Wee, George Koob, Chitra
Mandyam
33. Targeting Synaptic Actin Dynamics to Weaken Memories Driving
Methamphetamine Seeking Behavior
Erica Young, Erica Griggs, Gavin Rumbaugh, Courtney Miller
305
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
34. Characterization of Trace Amine Associated Receptor 1 Signaling and
its Differential Effects on Dopamine Transporter and Norepinephrine
Transporter Internalization
Gregory M. Miller, Bertha K. Madras, Xie Zhihua
35. Chronic Intermittent Ethanol Exposure produces Brain Region Specific
Alterations in Immunohistochemical Labeling of the Neuroactive Steroid
Allopregnanolone during Withdrawal in C57BL/6J Mice A. Leslie Morrow, Ana Maria Dumitru, Antoniette M. MaldonadoDevincci, Jason B. Cook, Marcelo F. Lopez, Howard C. Becker
36. HDAC2-Induced Chromatin and Synaptic Remodeling in Amygdala: A
Role in Anxiety and Alcoholism
Subhash C. Pandey, Sachin Moonat, Huaibo Zhang, Amul J. Sakharkar
37. Characteristic of Plasticity Facilitated by Intravenous Neural Stem Cell
Transplantation in FASD Model
Toshikazu Saito, Eri Hashimoto, Wataru Ukai, Tomohiro Shirasaka,
Toshihiro Yoshinaga, Masaru Tateno
38. Peripheral Markers of Psychostimulant Abuse
Bertha K. Madras, Brad Constant, Tim Walsh, Lisa Ogawa, Gregory
Miller, Eric Vallender, Susan Westmoreland
39. HMGB1 and TLR increase Neuroinflammation and Neurodegeneration in
Alcoholic Brain
Fulton Crews, Liya Qin, Jian Zou
306
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
40. A Double-Blind, Placebo-Controlled Trial of Topiramate for Pathological
Gambling
Heather A. Berlin, Ashley Braun, Daphne Simeon, Lorrin M. Koran, Marc N. Potenza, Susan L. McElroy, Timothy Fong, Stefano
Pallanti, Eric Hollander
41. Voltage Dependent Behavioral or Emotional Induction Associated
with Lack of Improvement in Subsyndromal Mood Ratings following
Subthalamic Nucleus Deep Brain Stimulation for Parkinson’s Disease
Osama A. Abulseoud, Kendall H. Lee, Bryan T. Klassen, Shirlene
Sampson, Julie A. Fields , Joesph Y.Matsumoto, Amit Chopra ,
Jarrett W. Richardson , Squire M. Stead , Cynthia J. Stoppel, Mark
A. Frye
42. Predictive Factors of Efficacy of Adjunct Extended Release Quetiapine
Fumarate (Quetiapine XR) in Patients with Major Depressive Disorder
Michael Bauer, Michael Thase, Madhukar Trivedi, Sherry Liu, Willie
Earley, Hans Eriksson
43. A Randomized Trial evaluated the Safety and Efficacy of Levomilnacipran
SR in the Treatment of Major Depressive Disorder
Anjana Bose, Norman Rosenthal, Carl Gommoll, Changzheng Chen,
Philip Ross
44. Mitochondrial Enhancement in Bipolar Disorder: A Placebo-Controlled
Trial of Acetyl-L-Carnitine and Alpha-Lipoic Acid in the Treatment of
Bipolar Depression
Brian P. Brennan, J. Eric Jensen, James I. Hudson, Caitlin Coit,
Ashley Beaulieu, Harrison G. Pope, Jr., Perry F. Renshaw, Bruce M. Cohen
307
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
45. Adjunctive Aripiprazole Response Rates in Major Depressive Disorder
Patients who Exhibit No or Minimal Improvement on Antidepressant
Monotherapy
Daniel E. Casey, Kimberly K. Laubmeier, Elizabeth E. Bellocchio,
James M. Eudicone, Robert D. McQuade, Ross A. Baker, Zia
Rahman
46. Reliability and Validity of the Fatigue Associated with Depression (FAsD)
Questionnaire in a Clinical Trial of Patients with Major Depressive
Disorder who Partially Respond to SSRI Treatment
Peter Classi, Denái Milton, Jeffrey M. Witkin, Richard Shelton,
Louis Matza
47. Cognitive-Behavioral Therapy in Women Discontinuing Antidepressant in
Anticipation of Pregnancy Christina Psaros, Marlene Freeman, Steven Safren, Maria Barsky,
Lee Cohen
48. A Double-Blind, Placebo-Controlled Study of Selegiline Transdermal
System (STS) in Depressed Adolescents
Melissa P. DelBello, Thomas J. Hochadel, Kimberly B. Portland,
Alain Katic, Arif Khan, Graham Emslie
49. Factorial Clinical Trials for Hybrid (Explanatory and Pragmatic) Research
Studies: Design for the “Optimizing Treatment for Complicated Grief”
Study
Naihua Duan, Barry Lebowitz, Charles F. Reynolds, Naomi Simon,
Yuanjia Wang, Sidney Zisook, M. Katherine Shear
308
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
50. A Double-Blind, Randomized, Placebo-Controlled Long-Term Study of
Aripiprazole in Children with Bipolar Disorder
Robert L. Findling, Eric A. Youngstrom, Nora K. McNamara, Robert
J. Stansbrey, Christine A. Demeter, Brieana M. Rowles, Thomas W. Frazier, Joseph R. Calabrese
51. Aripiprazole Augmentation Improves Aspects of Executive Function in
Major Depressive Disorder: A Pilot Study
Tracy L. Greer, Prabha Sunderajan, Bruce D. Grannemann,
Madhukar H. Trivedi
52. The V1b Receptor Antagonist SSR149415 in the Treatment of Major
Depressive and Generalized Anxiety Disorders: Results from Three
Double-Blind, Placebo-Controlled Studies
Guy Griebel, Stephen Stahl, Lisa Arvanitis
53. Algorithm-Driven Treatment of Bipolar Disorder in Correctional Setting:
Impact on Psychotropic Medication Utilization
Jayesh Kamath, Sara Wakai, Wanli Zhang, Robert Trestman
54. Early Improvement as a Predictor of Later Treatment Response in Acute
Manic or Mixed Episodes using CGI Assessments: A Pooled, Post-Hoc
Analysis
Armin Szegedi, Craig Karsson, Jun Zhao
55. Efficacy and Safety of Lisdexamfetamine Dimesylate in Adults with
Executive Dysfunction and Partial or Full Remission of Major Depressive
Disorder
Richard Keefe, Manisha Madhoo, Robert Roth, Angelo Sambunaris,
James Wu, Madhukar Trivedi, Colleen Anderson, Robert Lasser
309
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
56. Baseline Interleukin-6, Cortisol, and Insulin in Major Depressive Disorder
and Response to Pioglitazone: Preliminary Support for Insulin Sensitizers
as Modulators of Mood
David E. Kemp, Stephen J. Ganocy, Keming Gao, Faramarz IsmailBeigi, Carla Conroy, Sarah Obral, Joseph R. Calabrese
57. Bipolar Clinical and Health Outcomes Initiative in Comparative
Effectiveness (Bipolar CHOICE): Rationale and Design
Terence A. Ketter, Andrew C. Leon, Joseph R. Calabrese, David
E. Kemp, Michael E. Thase, Charles L. Bowden, Mauricio Tohen,
Edward S. Friedman, James H. Kocsis, Richard C. Shelton, Melvin
G. McInnis, Susan L. McElroy, Noreen A. Reilly-Harrington, Louisa
G. Sylvia, Thilo Deckersbach, Andrew A. Nierenberg
58. Efficacy of Adjunctive Aripiprazole in Patients with Major Depressive
Disorder whose Symptoms Worsen with Antidepressant Monotherapy: A
Pooled Analysis of Three Trials
J. Craig Nelson, Elizabeth E. Bellocchio, Zia Rahman, Kimberly
K. Laubmeier, James M. Eudicone, Robert D. McQuade, Robert M. Berman, Ross A. Baker, John Sheehan
59. A Prospective Study of the Role of Life Events in Precipitating Suicidal
Behavior
Maria A. Oquendo, Ernest Poh, M. Mercedes Perez-Rodriguez,
Gregory M. Sullivan, J. John Mann, Hanga C. Galfalvy
60. Efficacy of Vilazodone in Patients with Moderate, Moderately Severe and
Severe Depression - Pooled Analyses from 2 Randomized Phase III Trials
Donald Robinson, Carol Reed, Wenjie Song, John Edwards, Philip
Ross
310
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
61. Comparison of Computer vs Site-based Rater Administration of the
MADRS in Three Placebo Controlled Trials
Gary S. Sachs, Michelle Arkow, Dan DeBonis
62. Suicide, Depression, and Complicated Grief
M. Katherine Shear, Natalia Skritskaya, Yuanjia Wang, Christine
Mauro, Naihua Duan, Barry Lebowitz, Charles F. Reynolds, Naomi
Simon, Sidney Zisook, Nicole LeBlanc, John Worthington, Kim
Glickman, Angela Ghesquiere
63. Oxcarbazepine for Acute Affective Episodes of Bipolar Disorder: A
Cochrane Review and Meta-analysis
Akshya Vasudev, Karine A.N. Macritchie, Kamini Vasudev, Stuart
Watson, John Geddes, Allan H. Young
64. Do Current Internet-Based Patient Recruitment Methods Impact Patient
Retention and Completion Rates in Depression Studies and Are These
Metrics Truly Indicative of Positive Outcomes?
Charles S. Wilcox, Nader Oskooilar, My-Linh Tong, Judy L. Morrissey, Don F. De Francisco, Mellissa M. Henry, Daniel E. Grosz
65. 7 Deadly Sins: Guidelines for Reporting Clinical Trials Methodology
Research
Danielle Popp, Janet Williams, Michael Detke
66. The Maternal and the Paternal Brain: Synchrony, Specificity, and Links
with Oxytocin and Vassopresin
Shir Atzil, Talma Hendler, Yonatan Winetraub, Ruth Feldman
311
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
67. Capacity-Based Differences in Functional Network Activation during
Spatial Working Memory
Katherine H. Karlsgodt, Eliza Congdon, Russell A. Poldrack,
Angelica A. Bato, Fred W. Sabb, Edythe London, Robert Bilder,
Tyrone D. Cannon
68. fMRI Evidence of Long-Lasting, rTMS-Caused Remediation of
Performance Deficits in Working Memory Induced by Sleep Deprivation
Bruce Luber, Adrienne Tucker, Jason Steffener, Christian Habeck,
Zhi-De Deng, Yaakov Stern, Sarah H Lisanby
69. Phasic Dopamine Transmission encodes Cached Value following StateBased Reinforcer Devaluation
Vicente Martinez, Paul Phillips
70. The Corticotropin Releasing Factor 1 Receptor (CRF1R) Antagonist
GW876008 modulates Brain Response during Extinction in Patients with
Chronic Abdominal Pain, but not Healthy Control Subjects
Jennifer S. Labus, Catherine S. Hubbard, Michael S. Fanselow,
Bahar Ebrat, Joshua Bueller, Kirsten Tillisch, Jean Stains, George E. Dukes, Dennis L. Kelleher, Bruce D. Naliboff, Emeran A. Mayer
71. Posterior Cingulate Cortex is Critical for Associative Learning
Sarah Heilbronner, Michael Platt
72. Neural Representation of Value and Prediction Error Signals in Late-Life
Depression With and Without Suicidal Behavior
Alexandre Dombrovski, Katalin Szanto, Luke Clark, Charles
Reynolds, Greg Siegle
312
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
73. Neural Substrates of Real-Life Decisions to Use Marijuana: A
Neuroeconomic Study
Gillinder Bedi, Meg Haney
74. Linkages between Genetic Factors, Neurocognitive Activation, and
Response to Interventions to Reduce Adolescent Alcohol Use and Related
Risk Behavior
Angela Bryan, Eric Claus, Renee Magnan, Sarah Feldstein Ewing,
Kent Hutchison
75. Chronic Alcohol Exposure disrupts Executive Cognitive Function and D2
receptor Modulation of Neuronal Firing in the Prefrontal Cortex of the Rat
Heather Trantham-Davidson, Sam Centanni, Judson Chandler
76. Extended Release Naltrexone decreases Rewarding Properties of Sucrose
in Patients with Opioid Dependence
Daniel D. Langleben, Elliot L. Bush, Charles P. O’Brien, Anna Rose
Childress, James Cornish, Igor Elman
77. Association between Impulsivity and Risk-taking in a Sample of
Adolescent-onset Marijuana Users
Erin McGlade, Melissa Lopez-Larson, Deborah Yurgelun-Todd
78. Insula Functional Connectivity with Default-Mode Network is Modulated
by Varenicline and Nicotine in Abstinent Smokers
Matthew T. Sutherland, Allison J. Carroll, Betty Jo Salmeron,
Thomas J. Ross, Elliot A. Stein
79. Molecular Genetic Evidence for a Psychosis Phenotype Pamela DeRosse, Christopher Morell, Todd Lencz, Anil Malhotra
313
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
80. Unemployment and Substance Misuse and Disorder in the United States
during Economic Recession
Wilson Compton, Joe Gfroerer, Kevin Conway
81. Item Response Theory Analysis of DSM-IV Amphetamine Use Disorder
Criteria in an American Indian Community Sample
David Gilder, Cindy Ehlers
82. Risk Factors for Illicit Anabolic-Androgenic Steroid Use in Men: Results
from a Cross-Sectional Cohort Study James I. Hudson, Gen Kanayama, Harrison G. Pope Jr.
83. Nonmedical Prescription Opioid Use and Use Disorders Secondary to
Nonmedical Use among U.S. Young Adults by Educational Attainment
Silvia S. Martins, Lian-Yu Chen, Miriam C. Fenton, Katherine M. Keyes, Carla L. Storr
84. Cocaine Dependence specifically predicts Historical Suicide Attempts
among African-American Participants in a Community Corrections
Program
Cheryl B. McCullumsmith, James H. Meador-Woodruff, C. Brendan
Clark, Karen L. Cropsey
85. Childhood Emotional Abuse and Alcohol Dependence in Adulthood:
Mediating Effects of Neurotic Personality Traits and Anxiety Symptoms
among Treatment-Seeking Alcohol Dependent Individuals
Melanie L. Schwandt, Daniel Hommer, Markus Heilig, Ted George,
Vijay A. Ramchandani
314
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
86. Whole Transcriptome and Methylome Sequencing in the Alcohol Postdependent Rat Model
Estelle Barbier, Jenica Tapocik, Jesse Schank, Kornel Schuebel,
Zhifeng Zhou, Qiaoping Yuan, David Goldman, Markus Heilig
87. The Involvement of Alpha3-Containing Nicotinic Acetylcholine Receptors
in the Habenulo-Interpeduncular Pathway in Nicotine Self-Administration
Christie D. Fowler, Qun Lu, Paul J. Kenny
88. Studies Performed in Rodents and Primates Point to a Potential Role for
FAM111A in the Etiology of Alcohol Use Disorders
Stephen G. Lindell, Zhifeng Zhou, Qiaoping Yuan, Mitsuru Kimura,
J. Dee Higley, David Goldman, Christina S. Barr
89. Role of the Alpha-2 Nicotinic Acetylcholine Receptor Subunit in Nicotine
Reinforcement and Withdrawal
Shahrdad Lotfipour, Christie D. Fowler, Niall P. Murphy, Paul J. Kenny, Jim Boulter
90. Small and Whole Transcriptome RNA Sequencing identifies Key
Regulation Patterns in the Medial Prefrontal Cortex of the Alcohol
Dependent Rat
Jenica Tapocik, Estelle Barbier, Jesse Schank, Kornel Schuebel,
Zhifeng Zhou, Qiaoping Yuan, David Goldman, Markus Heilig
91. Evidence from Mouse and Man for a Role of Neuregulin 3 in Nicotine
Dependence
Jill R.Turner, Riju Ray, Bridgin Lee, E. Paul Wileyto, Don Baldwin,
Klaus Kaestner, Caryn Lerman, Julie A. Blendy
315
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
92. Csnk1e is a Genetic Regulator of Sensitivity to Psychostimulants and
Opioids
Camron D. Bryant, Clarissa C. Parker, Lili Zhou, Christopher Olker,
Ramalakshmi Y. Chandrasekaran, Travis T. Wager, Valerie J. Bolivar,
Martha H. Vitaterna, Fred W. Turek, Abraham A. Palmer
93. Exploratory Association Study of Genetic Variation in OPRM1 Gene and
Interpersonal Dysfunction in a Personality Disorder Enriched Sample Antonia S. New, M. Mercedes Perez-Rodriguez, Luis Ripoll,
Colin Hodgkinson, Zhifeng Zhou, Marianne Goodman, Harold W. Koenigsberg, David Goldman, Larry J. Siever
94. Exploratory Association Study of 130 Candidate Genes in Patients with
Borderline Personality Disorder and Healthy Controls
M. Mercedes Perez-Rodriguez, Panos Roussos, Laura Bevilacqua,
Qiaoping Yuan, Zhifeng Zhou, Colin Hodgkinson, Luis Ripoll,
Marianne Goodman, Harold W. Koenigsberg, David Goldman, Larry
J. Siever, Antonia S. New
95. Gene x Disease Interactions on Stress Reactivity in Cocaine Addiction Nelly Alia-Klein, Elena Shumay, Muhammad Parva, Amelia N. Morales, Patricia A. Woicik, Thomas Maloney, Gene-Jack Wang,
Joanna S. Fowler, Nora D. Volkow, Rita Z. Goldstein
96. TTC12-ANKK1-DRD2 and CHRNA5-CHRNA3-CHRNB4 influence
Different Pathways leading to Smoking Behavior from Adolescence to
Mid-Adulthood
Francesca Ducci, Marika Kaakinen, Juha Veijolai, Matti Isohannii,
Gunter Schumann, Marjo-Riitta Jarvelin
316
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
97. Linkage Analyses of Stimulant Dependence, Craving and Heavy Use in
American Indians Cindy L. Ehlers, Ian R. Gizer, David A. Gilder, Kirk C. Wilhelmsen
98. HPA Axis Response to Acute Stress and Hippocampal Expression of
Stress-Related Genes in Alcoholics and Cocaine Addicts
Mary-Anne Enoch, Zhifeng Zhou, Deborah Mash, Qiaoping Yuan,
David Goldman, Rajita Sinha
99. Galanin Receptor 1 (GALR1) SNP is Associated with Craving and
Smoking Relapse
Allison B. Gold, E. Paul Wileyto, Christopher Jepson, Adriana Lori,
Joseph F. Cubells, Caryn Lerman
100. Association of DRD4 Exon 3 VNTR and Alcohol’s Effects on Temporal
Cognition
John E. McGeary, Robert M. Swift, William H. Zywiak, Andrea
Grenga, Junghyun Kim, James Murphy
101. Dopaminergic and Cholinergic Genetic Variation moderates SmokingInduced Striatal Dopamine Release
Erika L. Nurmi, Karyn S. Mallya, Thomas M. Levin, Karen Ta,
Jaime La Charite, James T. McCracken, Arthur L. Brody
102. Genome-Wide Association Study of d-Amphetamine Response in Healthy
Human Participants identifies Association with Cadherin 13 (CDH13)
Amy Hart, Barbara Engelhardt, Margaret Wardle, Greta Sokoloff,
Andrew Skol, Matthew Stephens, Harriet de Wit, Abraham Palmer
317
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
103. Quit Success Genotype Score predicts Success and Lower/Slower Paced
Involvement with Common Addictive Substances
George R. Uhl, Donna Walther, William Eaton, Nicholas Ialongo,
Jed Rose
104. Effects of PPP1R1B Genetic Variation on In Vivo Human Central
Dopamine System Function
Daniel P. Eisenberg, Joseph C. Masdeu, Philip D. Kohn, Angela
Ianni, Erica B. Baller, Bhaskar Kolachana, Daniel R. Weinberger,
Karen F. Berman
105. Acamprosate Acts as a Partial Agonist of the NMDA Receptor: Evidence
from a Spectroscopy Study in Schizophrenia
Bernard A. Fischer, Laura M. Rowland, William R. Keller, Henry H. Holcomb, Robert W. Buchanan
106. Expression Profile of Metabotropic Glutamate Receptors in the Human
Post Mortem Schizophrenia Brain
Subroto Ghose, Kelly Gleason, Bryan Potts, Carol Tamminga
107. Expression of Dopamine Receptor D1 and D2 Transcripts in Brains of
Patients with Schizophrenia and Mood Disorders
Sanne S. Kaalund, Erin N. Newburn, Tianzhang Ye, Liqin Wang,
Ran Tao, Chao Li, Radhakrishna Vakkalanka, Amy Deep-Soboslay,
Llewellyn B. Bigelow, Richard Straub, Thomas M. Hyde, Daniel R. Weinberger, Barbara K. Lipska, Joel E. Kleinman
108. Familial Abnormalities of Endocannabinoid Functioning in Schizophrenia
and Bipolar Disorder
Dagmar Koethe, Franziska Pahlisch, Martin Hellmich, Cathrin
Rohleder, Andreas Meyer-Lindenberg, Daniele Piomelli, F. Markus
Leweke
318
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
109. Subcellular Deficits of Glutamate Transporter Expression in Schizophrenia
Robert McCullumsmith, Dan Shan, Vahram Haroutunian, James
Meador-Woodruff
110. Stress-Induced Dopamine (DA) Response in Subjects at Clinical High
Risk (CHR) for Schizophrenia with Concurrent Cannabis Use
Romina Mizrahi, Ivonne Suridjan, Isabelle Boileau, Pablo Rusjan,
Tony George, Alan Wilson, Sylvain Houle
111. Deficits in Transcriptional Regulators of Cortical Interneuron
Subpopulations in Schizophrenia
David W. Volk, Takurou Matsubara, Takanori Hashimoto, Elizabeth
J. Sengupta, David A. Lewis
112. Diminished In-Vivo Gamma-Aminobutyric Acid Concentrations in
the Visual and Prefrontal Cortices in Recent Onset Schizophrenia and
Correlations with Gamma Band Power
Jong H. Yoon, Richard Maddock, Michael J. Minzenberg, Michael
Buonocore, Renata Ooms, Glenn Gomes, Cameron S. Carter
113. Central Pathways of Pain and Pleasure Interact
Eric C. Porges, Jean Decety
114. A Multi-Modal Investigation of Treatment Response Associated with
Electroconvulsive Therapy in Major Depressive Disorder
Chris Abbott, Robert J. Thoma, Kaitlyn DePlonty, Jeffrey Lewine,
Jessica Turner
319
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
115. Cross-Sectional and Longitudinal Abnormalities in Brain Structure in
Children with Severe Mood Dysregulation or Bipolar Disorder
Nancy E. Adleman, Varun Razdan, Stephen J. Fromm, Reilly Kayser,
Daniel Dickstein, Daniel S. Pine, Ellen Leibenluft
116. Gray Matter Alterations in Type 2 Diabetes and Major Depression
Assessed by Voxel-Based Morphometry
Olusola Ajilore, Aifeng Zhang, Shaolin Yang, Anand Kumar
117. Amygdala and Prefrontal Dysfunction during Face processing in Pediatric
and Adult Patients with Bipolar Disorder: Role of Face Emotion and
Attentional Demands
Melissa A. Brotman, Aviva K. Olsavsky, Julia G. Rutenberg, Eli J. Muhrer, Stephen J. Fromm, Carlos A. Zarate, Jr., Daniel S. Pine,
Ellen Leibenluft
118. Major Depressive Disorder and Bipolar Disorder Subjects exhibit
Different Functional Brain Activation during a Depressive Episode
Michael Cerullo, Eliassen James, Fleck David, Wade Weber, Caleb
Adler, Melissa DelBello, Stephen Strakowski
119. Cortical Thickness in Medicated and Unmedicated Adolescents with
Major Depressive Disorder
Kathryn R. Cullen, Bonnie Klimes-Dougan, Kelvin Lim
120. Measurement of Brain GABA Concentration by Magnetic Resonance
Spectroscopy in Postpartum Depressed Women: A Feasibility Study of the
Worcester Foundation for Biomedical Research
Kristina M. Deligiannidis, Elif M. Sikoglu, Anthony J. Rothschild,
Scott A. Shaffer, Bruce A. Barton, Richard A.E. Edden, Constance
M. Moore
320
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
121. The Functional Neural Circuitry of Anhedonia in Adolescent Depression
Vilma Gabbay, Benjamin A. Ely, Saroja Bangaru, F. Xavier
Castellanos, Michael P. Milham
122. Structural Plasticity of the Right Inferior Frontal Gyrus in the Course of
Bipolar Disorders - Interplay between Compensatory Changes, Illness
Burden and Lithium Treatment
Tomas Hajek, Jeffrey Cullis, Ryan Blagdon, Anne Duffy, Lukas
Propper, Miloslav Kopecek, Tomas Novak, Cyril Hoschl, Martin
Alda
123. Increased Glutamate in the Dorsal Anterior Cingulate Cortex is Associated
with IFN-alpha-Induced Depression using Single Voxel Magnetic
Resonance Spectroscopy
Ebrahim Haroon, Reena Anand, Xiangchuan Chen, Samir Parekh,
Bobbi Woolwine, James R. Spivey, Xiaoping Hu, Andrew H. Miller
124. Social Rejection activates Endogenous Opioid Systems
David T. Hsu, Tiffany M. Love, Heng Wang, Kortni K. Meyers,
Kathleen E. Hazlett, Lisong Ni, Sara J. Walker, Steven T. Korycinski,
Robert A. Koeppe, Jennifer K. Crocker, Scott A .Langenecker, JonKar Zubieta
125. Brain Gray and White Matter Abnormalities identified with Ex Vivo
Magnetic Resonance Imaging in the GT-tg Mouse after HIV1-Tat
Expression Amanda N. Carey, Xiaoxu Liu, Elizabeth Sypek, Jay P. McLaughlin,
Marc J. Kaufman
126. CANDIShare: Enabling Probabilistic Neuroanatomy in Child Psychiatry
David N. Kennedy, Steven Hodge, Christian Haselgrove, Pallavi
Rane, Jean A. Frazier
321
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
127. Can ASL Measures of Anterior Cingulate Cortex Perfusion Predict
Treatment Outcome in Major Depressive Disorder?
F. Andrew Kozel, Peiying Liu, Uma Rao, Kimberly S. Mapes,
Patrick Marsh, Jorge R.C. de Almeida, Paul E. Croarkin, Carol A. Tamminga, Madhukar H. Trivedi, Hanzhang Lu
128. Sertraline-Induced Normalization of Medial Visceromotor Network
Dysfunction in Major Depression
Richard Lane, Tor Wager, Ramon Mercado, Scott Schafer, Julian
Thayer, John Allen
129. Amygdala and Subgenual Cingulate Activation to Emotion Processing is
Related to Cortisol Reactivity to Stress
Scott Langenecker, Kathleen Hazlett, Brennan Haase, Aaron
Vederman, Erich Avery, Robert Welsh, Sara Weisenbach, Jon-Kar
Zubieta
130. White Matter Structural Abnormalities in Bipolar Illness Revealed using
Diffusion Weighted MR Imaging
Alex Leow, Olusola Ajilore, Liang Zhan, Donatello Arienzo,
Johnson Gad Elkarim, Aifeng Zhang, Teena Moody, Jeffrey Fischer,
Anand Kumar, Paul Thompson, Jamie Feusner, Lori Altshuler
131. Monoamine Oxidase A (MAO-A) Binding in Prefrontal and Anterior
Cingulate Cortex in Postpartum Depression
Jeffrey H. Meyer, Julia Sacher, Alan A. Wilson, Pablo M. Rusjan,
Leslie Jacobs, Sylvain Houle
132. Genetic Variation of the 5-HT2C Receptor Modulates Human Striatal
Dopaminergic Activation to Stress
Brian J. Mickey, Benjamin J. Sanford, Tiffany M. Love, Pei-Hong
Shen, Colin Hodgkinson, David Goldman, Jon-Kar Zubieta
322
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
133. Lower Anterior Corpus Callosum Fractional Anisotropy in Major
Depressive Disorder as Measured by Tract Based Spatial Statistics and
Diffusion Tensor Imaging
Denis Peruzzo, Binod Thapa-Chhetry, M. Elizabeth Sublette,
Gregory M. Sullivan, J. John Mann, Ramin V. Parsey
134. White Matter Alterations in Youth at High-Risk for Bipolar Disorder
Donna J. Roybal, Naama Barnea-Goraly, Ryan Kelley, Layla
Barapour, Dylan Alegria, Meghan Howe, Allan L. Reiss, Kiki D. Chang
135. Ictal Perfusion SPECT reveals Focal Seizures associated aith a New Form
of ECT: Focal Electrically-Administered Seizure Therapy [FEAST]
E. Baron Short, Ken Spicer, Carol Burns, Melanie Archer, Matthew
Schmidt, Harold Sackeim, Mark S. George, Ziad Nahas
136. Longitudinal Amygdalar Neuroanatomy in Adolescents with Bipolar I
Disorder
Manpreet K. Singh, Layla Bararpour, Ryan Kelley, Erica Marie
Sanders, Meghan E. Howe, Allan Reiss, Kiki D. Chang
137. Abnormal Hypothalamus Functional Connectivity in Psychotic Major
Depression
Keith Sudheimer, Jennifer Keller, Alan Schatzberg
138. Abnormal Temporal Lobe White Matter as a Biomarker for Genetic Risk
of Bipolar Disorder
Philip Szeszko, Katie Mahon, Katherine Burdick, Toshikazu Ikuta,
Patricia Gruner, Anil Malhotra
323
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
139. Changes in Brain Activation Following Family-Focused Treatment in
Youth at-risk for Bipolar Disorder
Amy Garrett, Allan Reiss, David Miklowitz, Meghan Howe,
Manpreet Singh, Ryan Kelley, Dawn Taylor, Elizabeth George, Kiki
Chang
140. Hippocampal Activation predicts Treatment Response to Antidepressant
for Depression
Shigeru Toki, Yasumasa Okamoto, Keiichi Onoda, Shinpei
Yoshimura, Aihiko Kunisato, Go Okada, Shigeto Yamawaki
141. The D3 Dopamine Receptor in Cocaine Users: PET Studies with [11C](+)-PHNO
Arian Behzadi, Stephen Kish, Doris Payer, Sylvain Houle, Pablo
Rusjan, Junchao Tong, Peter Selby, Tony George, Martin Zack, Tina
McCluskey, Romina Mizrahi, Dennis James, Alan Wilson, Isabelle
Boileau
142. Up-Regulation of Nicotinic Acetylcholine Receptors in Menthol Cigarette
Smokers
Arthur L. Brody, Alexey G. Mukhin, Jaime La Charite, Karen Ta,
Judah Farahi, Catherine A. Sugar, Evan Vellios, Meena Archie,
Maggie Kozman, Mark A. Mandelkern
143. 16 msec “Unseen” Cocaine and Sexual Cues Recruit Limbic Motivational
Circuitry
Anna R. Childress, Ronald N. Ehrman , Ze Wang, Yin Li, Jesse J. Suh, Teresa R. Franklin, Marina Goldman, Regina Szucs-Reed,
Daniel D. Langleben, Shing Chun Lam, Kimberly A. Young, Charles
P. O’Brien
324
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
144. Neural Responses to Acute IV Alcohol in Healthy Moderate Drinkers
Emma Childs, Xiaodong Guo, Sean J. O’Connor, Elliot A. Stein,
Harriet de Wit
145. Denicotinized vs Average Nicotine Tobacco Smoking differentially
Releases Striatal Dopamine
Edward F. Domino, Lisong Ni, Joseph S. Domino, Wendy Yang,
Catherine Evans, Sally K. Guthrie, Robert A. Koeppe, Jon-Kar
Zubieta
146. DAT Genotype-Dependent Baclofen-Induced Inhibition of Ventral
Striatum and Medial Orbitofrontal Cortex Brain Responses to Smoking
Cues: Moving Toward a Personalized Medicine Approach to Cigarette
Addiction
Teresa Franklin, Jesse J. Suh, Joshua Shin, Rebecca Hazan,
Kanchana Jagannathan, Zachary Singer, Yin Li, Ze Wang, Marina
Goldman, Ronald Ehrman, Charles P. O’Brien, Falk Lohoff, Anna
Rose Childress
147. A Role for Cannabinoid CB1 Receptor Signaling in Social Behavior
Yoav Litvin, Matthew Hill, Bruce McEwen, Donald Pfaff
148. Oxytocin at the Initiation of Romantic Love: Relations to Couple
Interactive Reciprocity
Inna Schneiderman , Orna Zagoory-Sharon , James Leckman , Ruth
Feldman
149. Puberty: A Sensitive Period in the Neurobiology of Stress and Emotion. A
Study of Internationally Adopted Youth at the Pubertal Transition
Karina Quevedo, Anna Johnson, Michelle Loman, Lafavor Theresa,
Gunnar Megan
325
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
150. HPA-Axis Dysregulation, Posttraumatic Stress, and Maladaptive Grief in
Parentally Bereaved Children: Individual and Environmental Correlates
Julie Kaplow, Alan Prossin, Danielle Shapiro, Britney Wardecker,
James Abelson
151. Differential Effects of Estrogen Preparation on Changes in Regional
Cerebral Metabolism in Postmenopausal Women
Natalie L. Rasgon, Heather A. Kenna, Tonita Wroolie, Cheri Geist,
Elissa Epel, Candyce Kroenke, Jue Lin, Daniel Silverman
152. Chronic Pituitary Dysfunction after Blast-related Mild Traumatic Brain
Injury
Charles W. Wilkinson, Elaine R. Peskind, Elizabeth A. Colasurdo,
Kathleen F. Pagulayan, Jane B. Shofer
153. The Effect of Citalopram on CRF R1 and R2 Expression in the Dorsal
Raphe of a Primate Model of Differential Stress Sensitivity
Olga Senashova, Cynthia L. Bethea
154. Risk for Psychosis: HPA-Axis Dysregulation and Childhood Trauma
Rachel L. Loewy, Rahel Pearson, Barbara K. Stuart, Daniel H. Mathalon, Sophia Vinogradov
155. Stress Reactivity in Persons at Familial Risk for Schizophrenia
Diana Perkins, Karen Grewen, Aysenil Belger, Stacy Ramsey,
Kathryn Lansing, Jevelo Evans
156. Predicting Seizure Threshold and Individualizing Seizure Therapy Dose
by Pulse Amplitude Adjustment
Angel V. Peterchev, Moacyr A. Rosa, Sarah H. Lisanby
326
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
157. First Human Use of Focal Electrically-Administered Seizure Therapy
[FEAST] Shows Feasibility, Safety, Clinical Benefits and Short
Reorientation Time
Ziad Nahas, Baron E. Short, Carol Burns, Melanie Archer, Matthew
Schmidt, Joan Prudic, Mitchell S. Nobler, D. P. Devanand, Linda
Fitzsimons, Sarah H. Lisanby, Nancy Payne, Tarique Perera, Mark S. George, Harold A. Sackeim
158. Relapse Rates in Psychotic Depression are Lower than in Non-psychotic
Depression after a Successful Course of Electroconvulsive Therapy (ECT)
Georgios Petrides, Max Fink, Rebecca Knapp, Mustafa Husain,
Teresa Rummans, Keith Rasmussen, Samuel Bailine, Martina
Mueller, Charles Kellner
159. Narp Mediates the Antidepressant Effect of ECT
Punit Vaidya, Edward Retzbach, Ashley Blouin, Sungho Han,
JongAh Lee, Kellie Tamashiro, Jay M. Baraban, Irving M. Reti
160. Effects of Omega-3 Fatty Acid Supplements on PET Quantification of
Regional Brain Glucose Metabolism in Depressed and Healthy Volunteers
M. Elizabeth Sublette, Thomas B. Cooper, Maria A. Oquendo, J. John Mann
161. Inflammatory Cytokines are Associated with Exercise Augmentation
Treatment Outcome for Major Depressive Disorder. Marisa S.P. Toups, Chad Rethorst, Tracy Greer, Ryan Huebinger,
Bruce Grannemann, Benji Kurian, Madhukar Trivedi
327
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
162. The Combination of rAAV/GDNF with Fetal Ventral Mesencephalic
Transplants in Parkinsonian Monkeys does not Significantly Improve
Behavioral Outcome over Either Treatment Alone
D. Eugene Redmond Jr., Caleb R.S. McEntire, Joseph P. Kingsbery,
Csaba Leranth, John D. Elsworth, Kimberly B. Bjugstad, Robert H. Roth, Richard J. Samulski, John R. Sladek, Jr.
163. Normobaric Hyperoxia Treatment of Schizophrenia
Galila Agam, Yehudit Bloch, Julia Applebaum, Yamima Osher,
Shirly Amar, Abed Azab, RH Belmaker, Yuly Bersudsky
164. Effect of Nutrients on Intrinsic Brain Activity in Lean and Obese Women
Emeran A. Mayer, Kristen Coveleskie, Lynn Connolly, Jennifer
S. Labus, Bahar Ebrat, Jean Stains, Zhiguo Jiang, Brandall Y. Suyenobu, Kirsten Tillisch, Lisa A. Kilpatrick
165. Objective Assessment of Social Disinhibition using the Human Behavioral
Pattern Monitor (hBPM) in Bipolar Disorder and Schizophrenia Patients
Virgine Patt, Arpi Minassian, Brook Henry, Mark A. Geyer, William
Perry
166. Further Evidence for the Reliability and Validity of the Massachusetts
General Hospital Cognitive and Physical Functioning Questionnaire
(CPFQ)
Lee Baer, Susan Ball, JonDavid Sparks, Joel Raskin, Margaret
Ferguson, Sanjay Dubé, Maurizio Fava1
167. In Vivo Monitoring of Sodium-Potassium ATPase in Euthymic LithiumTreated Bipolar Disorder Subjects
Fernando Boada, Mary Phillips, David Kupfer, Yongxian Qian,
Vicent Lee
328
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
168. Development of Novel Integrative Multistate Measures of Efficacy,
Tolerability and Functional Status in Maintenance Clinical Trials for
Bipolar Disorder
Charles L. Bowden, Jim Mintz, Mauricio Tohen, Joseph Calabrese,
John Davis
169. Clinical Staging for Bipolar Disorder: Defining Empirically Derived
Distinct Prognostic Patient Groups
Sophia Frangou, Maria Reinares, Efstathios Papachristou, Philip
Harvey, George Ploubidis, Eduard Vieta
170. Prevalence and Importance of Subsyndromal Manic Symptoms during
Bipolar Major Depressive Episodes
Lewis L. Judd, Pamela J. Schettler, Hagop S. Akiskal, William
Coryell, Jan Fawcett, Jess G. Fiedorowicz
171. Use of the Functional Adjusted Clinical States in Bipolar Disorder
Methodology in the Lamotrigine Registration Studies in Bipolar Disorder
Mauricio Tohen, Jim Mintz, Charles Bowden, Joseph Calabrese,
John Davis
172. Can We Detect Bipolar in Youths? Diagnostic Efficiency of Caregiver,
Youth, and Teacher Report of Manic Symptoms in Outpatient Settings
Eric Youngstrom, Anna Van Meter, Guillermo Perez Algorta, Heather
Marcinick, Andrew Freeman, Oren Meyers, Christine Demeter,
Jennifer Youngstrom, Norah Feeny, Joseph Calabrese, Robert L. Findling
173. A Randomized, Placebo Controlled Investigation of Intranasal Oxytocin in
Patients with Anxiety
David Feifel, Kai Macdonald, Rebecca McKinney, Nicolle Heisserer,
Verenea Serrano
329
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
174. Comparison of High-Dose Escitalopram, Bupropion, and their
Combination from Treatment Initiation in Major Depressive Disorder: A
Double-Blind Study
Pierre Blier, Patrick J McGrath, Richard Bergeron, Jonathan W. Stewart
175. Correlates of Ziprasidone Adherence in the Maintenance Treatment of
Bipolar Disorder: Analysis of STEP-BD Data
John O. Brooks
176. Neural Activity in Visual Cortical Areas during the Processing of
Emotional Expressions Predicts Antidepressant Response to the
Anticholinergic Scopolamine
Maura Furey, Erica Frankel, Elana Hoffman, Andrew Speer, Wayne
Drevets, Carlos Zarate Jr.
177. Effect of Lithium and Valproate on Brain Activation Patterns in fMRI in a
Working Memory Paradigm in Bipolar I Patients Carlos Lopez-Jaramillo, Juan Lopera-vasquez, Jorge Delgado,
Simon Rascovsky, Javier I. Escobar
178. Genetic Ancestry among Self-Reported African-Americans and
Pharmacogentic Response to SSRI Treatment of Major Depression
Eleanor Murphy, Francis McMahon
179. The Effect of Switching from Olanzapine, Quetiapine, or Risperidone
to Aripiprazole on Risk of Cardiovascular Disease: Results from the
Comparison of Antipsychotics for Metabolic Problems (CAMP) Study
T. Scott Stroup, Robert M. Hamer, Neepa Ray, Susan M. Esscok,
Jeffrey A. Lieberman
330
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
180. Gender Differences in a Placebo-Controlled Trial of Fluoxetine for Body
Dysmorphic Disorder Katharine A. Phillips, Megan M. Kelly
181. Lamotrigine in Bipolar Disorder, Depressed or Mixed Phase and Cocaine
Dependence
E. Sherwood Brown, Prabha Sunderajan, Lisa T. Hu, Sharon Sowell,
Thomas Carmody
182. Oral Naltrexone Alters the Subjective but not the Physiological Effects of
Oral d-Amphetamine in Humans
Sandra D. Comer, Phillip A. Saccone, Perrine Roux, Jermaine D. Jones, Ziva D. Cooper, Suzanne K. Vosburg, Maria A. Sullivan,
Eric Rubin, Jeanne M. Manubay, Shanthi Mogali, Margaret Haney,
Richard W. Foltin
183. Effects of D-Amphetamine on Responses to Emotional Stimuli
Margaret Wardle, Harriet de Wit
184. The Effects of the NK1 Antagonist Aprepitant on Opioid Withdrawal in
Patients Maintained on Methadone
Jermaine D. Jones, Taylor Speer, Edward Nunes, Sandra D. Comer,
Stephen Ross, John Rotrosen, Malcolm Reed
185. Naltrexone Reduces Women’s Weight Gain in Smoking Cessation at Six
Month Follow-Up
Andrea C. King, Dingcai Cao, Lingjiao Zhang, Stephanie S. O’Malley
331
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
186. Effects of MDMA on Social and Emotional Processing in Humans
Matthew G. Kirkpatrick, Margaret C. Wardle, Royce Lee, Harriet de
Wit
187. Baclofen as a Novel Pharmacotherapy for Alcohol Dependence:
Preliminary Findings from a Human Laboratory Double-Blind PlaceboControlled Randomized Study
Lorenzo Leggio, George Kenna, William Zywiak, Steven Edwards,
Samuel Fricchione, Tonya Tavares, Jessica Shoaff, John McGeary,
Robert Swift
188. Characterization of Operant Intravenous Alcohol Self-administration in
Humans: Open-Bar and Progressive-Ratio Paradigms
Bethany L. Stangl, Vatsalya Vatsalya, Molly Zametkin, Daniel W. Hommer, Vijay A. Ramchandani
189. The Calbindin-D28k Binding Site on Inositol Monophosphatase may
allow Inhibition Independent of the Lithium Site of Action
Orna Almog, Galila Agam
190. Organic Cation Transporters: Emergence of Novel Therapeutic Targets to
Improve Treatment of Depression
Lynette C. Daws, Deana M. Apple, Rebecca Horton, Sonio Cano,
Melissa Vitela, Glenn M. Toney, Georgianna Gould, Wouter Koek
191. Role of the5 HT7ReceptorSubtype in Mediating Antidepressant-Like
Effects of Lurasidone
Peter B. Hedlund, Lindsay Cates, Salvador Huitron-Resendiz,
Amanda J. Roberts, Herbert Y. Meltzer
332
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
192. ERK within the VTA Regulates Adult Behavioral Outputs Induced by
Fluoxetine Exposure during Adolescence
Sergio D. Iñiguez, Lyonna F. Alcantara, Vincent Vialou, Mary K. Lobo, Eric J. Nestler, Carlos A. Bolaños-Guzmán
193. Development of Novel Protein Peptide with Antidepressant-Like Effect
Fang Liu
194. Effects of Peripherally Restricted Kappa Opioid Receptor Agonists on
Pain-Stimulated and Pain-Depressed Behavior in Rats
S. Stevens Negus
195. Reduced Phosphodiesterase-2 Activity in the Amygdala Results in
Anxiolytic- and Antidepressant-Like Effects on Behavior in Mice
James M. O’Donnell, Anbrin Masood, Qiang Wu, Han-Ting Zhang,
Steven P. Wilson
196. Dynorphin and Orexin Interactions in the Development and Expression of
Depression-Related Anhedonia
Jaak Panksepp, Christine Nocjar
197. 5-HT Regulation of TrkB Signaling: Role of TG2/Rac-1 Pathway
Anilkumar R. Pillai, Chirayu Pandya, Alvin Terry, Jr., Ammar
Kutiyanawalla
198. Double Dissociation between the Roles of BDNF in the Antidepressant
Effects of Electroconvulsive Treatment and Desipramine
Dekel Taliaz, Sharon Haramati, Alon Chen, Abraham Zangen
333
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
199. Quantitative Proteomic and Metabolomic Profiling of Antidepressant Drug
Action Reveals Novel Targets beyond Monoamine Elevation
Chris W. Turck, Christian Webhofer
200. RG1678, a Novel and Potent Glycine Reuptake Inhibitor (GRI), Enhances
the Efficacy of Antipsychotics in Animal Models of Schizophrenia
Daniela Alberati, Jean-Luc Moreau, Roland Mory, Joseph G. Wettstein
201. Characterization of the Relationship between Target Occupancy/
Modulation and Preclinical/Clinical Responses for the Glycine Transporter
1 (GlyT1) Inhibitor Org 25935
Peter Dogterom, Fiona Thomson, Richard Hargreaves, Terence
Hamill, Cyrille Sur, Jason Uslaner, Donnie Eddins, Josh Vardigan,
Srini Jayaraman, John Morrow, Huub Jan Kleijn, Jacques Schipper,
Larry Ereshefsky, Stan Jhee, Darryle Schoepp
202. Selective TAAR1 Activation Reveals a Novel Approach for
Neuropsychiatric Therapy
Florent G. Revel, Jean-Luc Moreau, Raul R. Gainetdinov, Tatyana
D. Sotnikova, Antonio Ferragud, Clara Velazquez-Sanchez, Stephen
R. Morairty, Thomas S. Kilduff, Roger D. Norcross, Amyaouch
Bradaia, Juan J. Canales, Tanya Wallace, Marc G. Caron, Joseph G. Wettstein, Marius C. Hoener
203. Subchronic Treatment with Lurasidone has both Preventive and Enduring
Reversal Effects on the Phencyclidine (PCP)-Induced Deficit in Novel
Object Recognition (NOR) in Rats
Masakuni Horiguchi, Kayleen E. Hannaway, Adesewa E. Adelekun,
Herbert Y. Meltzer
334
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
204. Relationship between In Vivo Receptor Occupancy and Efficacy of Novel
Metabotropic Glutamate Receptor Subtype 5 Allosteric Modulators with
Different in Vitro Profiles of Activity at mGlu5
Jerri M. Rook, Mohammed N. Tantawy, Mohammad S. Ansari, Y. Sandra Zhou, Ryan D. Morrison, Shaun R. Stauffer, J. Scott Daniels,
Craig W. Lindsley, P. Jeffrey Conn
205. High-Throughput Screening for Allosteric Modulators of the D2
Dopamine Receptor
David R. Sibley, R. Benjamin Free, Noel Southall, Trevor Doyle,
Rebecca A. Roof, Jennie L. Conroy, Yang Han, Jonathan A. Javitch,
Marc Ferrer
206. The Novel Opioid Receptor Modulator RDC-0313 (ALKS 33) reduces
Olanzapine-Induced Weight Gain and Adipose Accretion in a Novel
nonhuman Primate Model of Antipsychotic-Related Weight Changes
Mark Todtenkopf, Reginald Dean, Michael Brunner, Michael Knopp,
Daniel Deaver
207. Effects of Oxytocin on Social Symptoms in Adults with Schizophrenia:
Cognition and Physiology
Joshua Woolley, Brandon Chuang, Joshua Resa, Katherine Rankin,
Sophia Vinogradov
208. Hard-Wired for Hedonism: The Role of Cortical D1 Dopamine Receptors
in Reward Processing
Heather C. Brenhouse, Nadja Freund, Britta Thompson, Kai C. Sonntag, Susan L. Andersen
209. A Role for Noradrenergic Systems in Kappa-Opioid Dependent Stress and
Drug Seeking Behaviors
Ream Al-Hasani, Jordan G. McCall, Michael R. Bruchas
335
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
210. The Effects of Small Molecules on Trace Amine-Associated Receptor 1
(TAAR1) Inform on its Contribution to Methamphetamine Abuse
David K. Grandy, Katie R. Tallman, Madeline S. Grandy, Troy A. Wahl
211. Potent Reinforcing Effects of the Synthetic Cathinone Methylenedioxypyrovalerone (MDPV) in Rats
Lucas R. Watteron, Peter R. Kufahl, Natali E. Nemirovsky, Kaveish
Sewalia, M. Foster Olive
212. Behavioral and Thermoregulatory Effects of Novel Cathinone Derivative
Drugs 4-MMC and MDPV
Shawn A. Aarde, M. Jerry Wright, Jr., Matthew W. Buczynski,
Deepshikha Angrish, Loren H. Parsons, Karen L. Houseknecht,
Tobin J. Dickerson, Michael A. Taffe
213. Opioid Regulation of GABAergic Circuitry in the Extended Amygdala
Kristen Pleil, Chia Li, Thomas Kash
214. Stress Adaptations in Endocannabinoid Signaling in the Amygdala:
Implications for Novel Treatment Approaches for Affective Disorders
Sachin Patel
215. Social Isolation during Adolescence Alters Dopamine Modulation in the
Adult Medial Prefrontal Cortex
Danielle S. Counotte, Petra J.J. Baarendse, Louk J.M.J. Vanderschuren, Patricio O’Donnell
336
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
216. Essential Role of Ventral Tegmental Area Dopamine Neurons in Mediating
the Induction and Rapid Reversal of Depression-Like Behaviors
Allyson K. Friedman, Dipesh Chaudhury, Jessica J. Walsh, Barbara
Juarez, Mary Kay Lobo, Herbert E. Covington III, Vincent F. Vialou,
Hsing-Chen Tsai, Karl Deisseroth, Eric J. Nestler, Ming-Hu Han
217. Elevation of Oxidative Stress in Tissue Cultures from Persons with Major
Depression
Richard C. Shelton, Sara A. Gibson, Zeljka Korade
218. Deficits in Vesicle Priming Underlie decreases in Glutamate Release in
Dysbindin-Mutant Mice
Shalini Saggu, Ty Cannon, J. David Jentsch, Antonieta Lavin
219. Acute Stress alters Plasticity in the Nucleus Accumbens via a
Dopaminergic Mechanism Kathryn M. Gill, Anthony A. Grace
220. Psychophysiological Predictors of PTSD Risk in Active Duty Marines: Preliminary Findings from the Marine Resiliency Study
Victoria Risbrough, Dewleen Baker , Caroline Nievergelt, Abigail
Goldsmith, Brett Litz, William Nash, Mark Geyer
221. Unique Changes in Fast-Spiking Interneuron Activity during SleepDependent Consolidation of Plasticity in the Developing Visual Cortex
Sara J. Aton, Michelle Dumoulin, Julie Seibt, Tammi Coleman,
Adam Watson, Marcos G. Frank
337
ACNP 50th Annual Meeting • Final Program
Poster Session III—Wednesday
222. The Roles of the Infralimbic Cortex in Impulsive Action
Yu Ohmura, Iku Tsutsui-Kimura, Takeshi Izumi, Toshiya
Matsushima, Hidetoshi Amita, Takayuki Yoshida, Mitsuhiro
Yoshioka
223. Safety Learning requires Synaptic Plasticity in the Posterior Insular Cortex
John Christianson, Johanna Flyer, Linda Watkins, Steven Maier
224. The Impact of Multi-System Infections on the Regulation of Affective
States is Explored using Chronic Neurologic Lyme Disease as a Model
James R. Moeller, Brian A. Fallon, John G. Keilp
225. TRP Channels as Mediators of Oxytocin-Induced Anxiolysis
Erwin H. Van den Burg, Julia Stindl, Marisa Brockmann, Inga D. Neumann, Olaf Strauss
226. Reduced Play Behavior and Altered Monamine Neurochemistry in Fisher
344 Rats
Cynthia A. Crawford
227. Dopamine Axons Innervating the Rat Lateral Habenula: Ultrastructural
Features Common to and Distinct from Other Forebrain Dopamine
Projections
Susan Sesack, George Krakowski, Judith Joyce Balcita-Pedicino
338
ACNP 50th Annual Meeting • Final Program
Notes
339
ACNP 50th Annual Meeting • Final Program
Notes
340
Nonmember
Participants
Nonmember
Participants
Notes
ACNP 50th Annual Meeting • Final Program
Jean Addington
Professor
University of Calgary
3280 Hospital Drive N.W.
Calgary, AB T2N4Z6
Nicole M. Avena
Assistant Professor
University of Florida
Department of Psychiatry, L4-100
100 S. Newell Dr.
Gainesville, FL 32608
Daniela Paola Alberati
F. Hoffmann-La Roche
Grenzacherstrasse 124
Basel, 4070
Dewleen Baker
Cincinnati VA Medical Center
3200 Vine Street
PTSD Unit 7E
Cincinnati, OH 45220
Cristina Alberini
Professor
Mount Sinai School of Medicine
Department of Neuroscience
Box 1065
New York, NY 10029
Debra A. Bangasser
Postdoctoral Fellow
The Children’s Hospital of Philadelphia
3615 Civic Center Blvd.
ARC 402E
Philadelphia, PA 19104
David G. Amaral
Professor
UC Davis
2825 50th St.
Sacramento, CA 95618
Ben A. Barres
Professor of Neurobiology
Stanford University School of Medicine
Dept of Neurobiology, Fairchild D235
299 Campus Drive
Stanford, CA 94305-5125
Noelle C. Anastasio
Postdoctoral Fellow
University of Texas Medical Branch
301 University Blvd. Route 0615
Galveston, TX 77555-0615
Francesco Benedetti
Professor
Scientific Institute
University Vita-Salute San Raffaele
Dept. of Clinical Neuroscience
San Raffaele Turro via Stamira d’Ancona 20
Milano, 20125
Mark Sascha Ansorge
Assistant Professor
Columbia University
1051 Riverside Drive
New York, NY 10032
Ottavio Arancio
Associate Professor
Columbia University Medical Center
630 W. 168th Street
New York, NY 10032
Alessandro Bertolino
Azienda Ospedaliera-Ospedale Policlinico
Consorziale
Dept. of Neurological & Psychiatric Sc.
Piazza Giolio Cesare, 11
Bari, 70124
Paola Arlotta
Assistant Professor
Harvard University
185 Cambridge Street
Boston, MA 02114
Kent Berridge
Professor
University of Michigan
East Hall
Ann Arbor, MI 48109
341
ACNP 50th Annual Meeting • Final Program
Kristin L. Bigos
Post-Doctoral Fellow
National Institute of Mental Health
9000 Rockville Pike
Building 10, Room 3C101
Bethesda, MD 20892
Claudia Buss
Assistant Professor
University of California Irvine
333 The City Blvd. W.
Suite 1200
Orange, CA 92868
John Blangero
Director, AT&T Genomics Computing Center
Texas Biomedical Research Institute
7620 N.W. Loop 410
San Antonio, TX 78227
Juan Bustillo
Professor
University of New Mexico
Dept Psychiatry MSC09
1 University of New Mexico
Albuquerque, NM 87131
David Borsook
Associate Professor
Harvard University
115 Mill Street
Belmont
Boston, MA 02478
Gyorgy Buzsaki
Professor
Center For Molecular And Behavior
Neuroscience, Rutgers University
Newark, NJ Newark
Chas Bountra
University of Oxford, SGC
ORCRB
Roosevelt Drive
Oxford, OX3 7DQ
Tyrone D. Cannon
Professor
UCLA
1285 Franz Hall
Los Angeles, CA 90095
Stephen Brimijoin
Professor
Mayo Clinic
200 First Street S.W.
Rochester, MN 55905
Mario Capecchi
Professor
University of Utah/Howard Hughes Medical
Institute
15 North 2030 East, #5440
Salt Lake City, UT 84112
Camron D. Bryant
University of Chicago
5801 South Ellis Avenue
Chicago, IL 60637
Arthur Caplan
University of Pennsylvania
3401 Market Street, Suite 320
Philadelphia, PA 19104
Neil Buckholtz
Chief, Dementias of Aging Branch
National Institute on Aging
7201 Wisconsin Avenue
Suite 350
Bethesda, MD 20892
Sue Carter
Professor
University of Illinois at Chicago
1601 W. Taylor St.
Department of Psychiatry
Chicago, IL 60612
342
ACNP 50th Annual Meeting • Final Program
Sohini Chowdhury
Vice President, Research Partnerships
The Michael J. Fox Foundation
for Parkinson’s Research
90 Broad Street
10th Floor
New York, NY 10004
Ralph DiLeone
Associate Professor
Yale University School of Medicine
34 Park St.
New Haven, CT 06519
C. Neill Epperson
Associate Professor
University of Pennsylvania
3535 Market Street
Rm 3001
Philadelphia, PA 19104
Jessica Connelly
Assistant Professor
University of Virginia
409 Lane Road
MR4 Building, Room 6031A
Charlottesville, VA 22908
Emad N. Eskandar
Associate Professor
Massachusetts General Hospital
55 Fruit Street
White 502
Boston, MA 01908
Kelly L. Conrad
Vanderbilt University Medical Center
23rd and Pierce Ave.
754B, RRB
Nashville, TN 37232
Damien Fair
Assistant Professor
Oregon Health and Science University
3181 S.W. Sam Jackson Park Road
UHN80R1
Portland, OR
Mark R. Cookson
Senior Investigator
Laboratory of Neurogenetics
35 Convent Drive
Bethesda, MD 20892
Rita M. Cowell
Assistant Professor
University of Alabama at Birmingham
1720 7th Avenue South
SC 729
Birmingham, AL 35294
Daniel Falk
Health Scientist Administrator
National Institute on Alcohol Abuse and
Alcoholism, NIH
5635 Fishers Lane
Room 2040
Rockville, MD 22201
Bruce Newell Cuthbert
Director, DATR
NIMH
6001 Executive Blvd.
MSC 9632
Bethesda, MD 20892-9632
Ruth Feldman
Professor
Bar-Ilan University
Department of Psychology
Gonda Brain Sciences Center
Ramat-Gan, 52900
Anders Dale
Professor
UCSD
9500 Gilman Drive
La Jolla, CA 92093
Dianne Figlewicz Lattemann
Senior Research Career Scientist/ Research
Professor
VA Puget Sound Health Care System
1660 So. Columbian Way (151)
Seattle, WA 98108
343
ACNP 50th Annual Meeting • Final Program
Kyle J. Frantz
Associate Professor
Georgia State University
Neuroscience Institute
PO Box 5030
Atlanta, GA 30302-5030
André Felix Gentil
Post-Graduate Student
Department of Psychiatry
University of Sao Paulo Medical School
Rua DR Carlos N. S. Aranha 179
Sao Paulo
Sao Paulo, 05450010
Samantha Fung
Research Officer
NeuRA & SRI
Corner of Baker & Easy
Sydney, 2031
Daniel H. Geschwind
Professor
UCLA
Gonda 2506
Los Angeles, CA 90095
Aurelio Galli
Professor
Vanderbilt
MRBIII 7124
465 21st Ave. South
Nashville, TN 37232
Richard Alexander Gibbs
Professor
Baylor College of Medicine
One Baylor Plaza
Houston, TX 77030
Adriana Galvan
Assistant Professor
UCLA
1285 Franz Hall
Box 951563
Los Angeles, CA 90095
David B. Goldstein
Director
Duke University
Box 91009
Durham, NC 27708
Larry Goldstein
UCSD School of Medicine
9500 Gilman Drive
La Jolla, CA 92093
Wenbiao Gan
Associate Professor
New York University School of Medicine
Skirball Institute
New York, NY 10016
Guillermo Gonzalez-Burgos
Assistant Professor
University of Pittsburgh
200 Lothrop Street
W1651 Biomedical Science Tower
Pittsburgh, PA 15261
Amy Garrett
Senior Scientist
Stanford University
401 Quarry Road
Palo Alto, CA 94305-5795
Steven Grant
Branch Chief
NIDA/NIH/DHHS
6001 Executive Blvd.
Room 3155
Bethesda, MD 20892
Shaoyu Ge
Assistant Professor
SUNY Stony Brook
Department of Neurobiology and Behavior
SUNY Stony Brook
Stony Brook, NY 11794
344
ACNP 50th Annual Meeting • Final Program
Hank Greely
Professor
Stanford University
Stanford Law School
559 Nathan Abbott Way
Stanford, CA 94305-8610
Kenji Hashimoto
Professor
Chiba University Center for Forensic Mental
Health
1-8-1 Inohana
Chiba, 260-8670
Ben Greenberg
Associate Professor
Brown Medical School
Butler Hospital
345 Blackstone Blvd.
Providence, RI 02906
Jonathan Hollander
The Scripps Research Institute
130 Scripps Way #2A2
Jupiter, FL 33458
Andrew Holmes
Senior Investigator
NIAAA
5625 Fishers Lane
Rockville, MD 20852
Christopher T. Gregg
Assistant Professor
University of Utah
School of Medicine
20 North 1900 East, 401 MREB
Salt Lake City, UT 84132-3401
William Horan
University of California, Los Angeles
11301 Wilshire Blvd., Bldg 210
Los Angeles, CA 90073
Eugenia V. Gurevich
Associate Professor
Vanderbilt University
2200 Pierce Ave., PRB417C
Nashville, TN 37232
Elaine Hsiao
Doctoral fellow
Caltech
1200 E. California Blvd.
BIO21 MC216-76
Pasadena, CA 91125
Chang-Gyu Hahn
Associate Professor
University of Pennsylvania
125 South and 31st
Philadelphia, PA 19104-3403
Richard Huganir
Professor and Director
Johns Hopkins University School of Medicine
725 N. Wolfe Street
Hunterian 1009A.
Baltimore, MD 21205
Ming-Hu Han
Mount Sinai School of Medicine
One Gustave L. Levy Place
New York, NY 10029
Christina M. Hultman
Professor
Department of Medical Epidemiology and
Biostatistics
Karolinska Institutet
Stockholm, Se 171 77
Charles R. Harman
Director, External Relations
NAMI
3803 N. Fairfax Drive
Suite 100
Arlington, VA 22203
Michael Jackson
Sanford-Burnham Medical Research Institute
10901 N. Torrey Pines Road
La Jolla, CA 92037
345
ACNP 50th Annual Meeting • Final Program
Sharna Jamadar
Olin Neuropsychiatry Research Center
Whitehall Building - Institute of Living
200 Retreat Avenue
Hartford, CT 06106
Patrick J. Kennedy
Former Congressman
1 Mind 4 Research
1101 K. St. MW
Gephardt Associates
Washington, DC 20004
David Jentsch
UCLA Psychology
P.O. Box 951563
Los Angeles, CA 90095
Barbara Ann Koenig
Professor of Biomedical Ethics and Medicine
Mayo Clinic
Dept. of General Internal Medicine
200 First St., S.W.
Rochester, MN 55905
Sheena Josselyn
Associate Professor
Hospital for Sick Children
555 University Ave.
Toronto, ON M5G 1X8
Ann Kring
Professor
University of California, Berkeley
Dept. of Psychology
3210 Tolman Hall
Berkeley, CA 94720
Tanja Jovanovic
Emory University School of Medicine
49 Jesse Hill Jr Dr.
Suite 331
Atlanta, GA 30303
Michael Kaplitt
Weill Cornell Medical College
1300 York Avenue
New York, NY 10065
Louis Kunkel
Professor of Pediatrics and Genetics
Children’s Hospital Boston
3 Blackfan Circle
CLS 15027.1
Boston, MA 02115
Atsushi Kamiya
Johns Hopkins University School of
Medicine
600 North Wolfe Street, Meyer 3-161
Baltimore, MD 21287
Story Landis
National Institute on Neurologic Disorders
and Stroke
Building 31, Room 8A52
Bethesda, MD 20892
Maria Karayiorgou
Columbia University
1051 Riverside Drive
Unit 28
New York, NY 10032
Francis Lee
Professor
Weill Cornell Medical College
1300 York Avenue
New York, NY 10065
Takaoki Kasahara
Lab for Molecular Dynamics of Mental
Disorders
RIKEN Brain Science Institute
Hirosawa 2-1
Wako-shi, 351-0198
Michael L. Lehmann
National Institutes of Health
9000 Rockville Pike
Bldg 35, Room 1C911
Bethesda, MD 20892
346
ACNP 50th Annual Meeting • Final Program
Frances Leslie
Professor
UC Irvine
Department of Pharmacology
Rm 360 MS2, School of Medicine
Irvine, CA 92697
Karen G. Martinez
University of Puerto Rico
170 Avenue Arterial Hostos
Apartment B6
San Juan, PR 00918
Jennifer Blair McCormick
Assistant Professor
Mayo Clinic
Plummer 3
200 First Street S.W.
Rochester, MN 55905
Chunyu Liu
Assistant Professor
University of Chicago
924 E. 57th Street
R012
Chicago, IL 60637
Michael Meaney
Professor
McGill University
Douglas Hospital Research Centre
6875 Boulevard LaSalle
Montreal, Montreal (Quebec) H4H 1R3
Mary Kay Lobo
Mount Sinai School of Medicine
One Gustave L. Levy Place
New York, NY 10029
Jamie Maguire
Assistant Professor
Tufts University School of Medicine
Department of Neuroscience
136 Harrison Ave., SC205
Boston, MA 02111
Peter Meerlo
Assistant Professor
University of Groningen
Center for Behavior and Neurosciences
P.O. Box 11103
Groningen, 9700 CC
Robert W. Mahley
Senior Investigator
Gladstone Institute of Neurological Disease
Professor
University of California, San Francisco
The J. David Gladstone Institutes
1650 Owens Street
San Francisco, CA 94158
Michael Merzenich
Professor Emeritus
UCSF
Keck Center for Integrative Neurosciences
828 HSE, UCSF
San Francisco, CA 9413-0732
David Michelson
Vice President, NS Clinical
Merck Research Laboratories
UG4C-06 P.O. Box 1000
North Wales, PA 19454
Roberto Malinow
Professor
UCSD
9500 Gilman Drive
La Jolla, CA 92093
Holly Moore
Associate Professor
Columbia University Psychiatry
New York Psychiatric Institute
1051 Riverside Dr.
Mail Unit 14
New York, NY 10032
Charles Raymond Marmar
Professor and Chair
NYU Langone Medical Center
550 First Ave., OBV A643
New York, NY 10016
347
ACNP 50th Annual Meeting • Final Program
John H. Morrison
Professor and Dean
Mount Sinai School of Medicine
One Gustave L. Levy Place
Box 1065
New York, NY 10029
Raj Narendran
Associate Professor
University of Pittsburgh
200 Lothrop Street
B-938
Pittsburgh, PA 15213
Stephen J. Moss
Professor
Tufts University School of Medicine
Dept. of Neuroscience
136 Harrison Avenue
Boston, MA 02030
Roberta Ness
University of Texas Health Science Center
1200 Hermann Pressler Street
P.O. Box 20186
Houston, Texas 77225
Jeffrey S. Nye
Vice President and Global Head, External
Innovation
Johnson and Johnson Pharma R&D
1125 Trenton Harbourton Rd.
Titusville, NJ 08560
Robin Macgregor Murray
Professor
Institute of Psychiatry
De Crespigny Park
London, BR3 3BG
Aye Mu Myint
Ludwig-Maximilians University
Psychiatric Hospital
Nussbaumstrasse 7
Munich, 80336
Scott Orr
Harvard Medical School
Massachusetts General Hospital
VA Medical Ctr, Research Service (151)
718 Smyth Road
Manchester, NH 03104
Karim Nader
Professor
McGill University
1205 Dr. Penfield Ave.
Montreal, QC H3A1B1
Anand Pandya
Vice-Chair
Cedars-Sinai Medical Center
8730 Alden Drive, C-301
Los Angeles, CA 90048
Michael A. Nader
Professor
Wake Forest University School of Medicine
Department of Physiology & Pharmacology
Medical Center Blvd., 546 NRC
Winston-Salem, NC 27157-1083
Diana Perkins
Professor
University of North Carolina
CB 7160
Chapel Hill, NC 27514
Kazu Nakazawa
Chief
NIMH
9000 Rockville Pike
Bldg 35, Room 1C915
Bethesda, MD 20892-3710
Jamie Peters
Post-Doc
VU University Medical Center
Van der Boechorststraat 7
MF Bldg, Room C-460
Amsterdam, 1081BT
348
ACNP 50th Annual Meeting • Final Program
Jean-Baptiste Poline
Neurospin, I2BM
Bâtiment 145- Point Courrier 156
Gif-sur-Yvette Cedex, 91191
Antonio Rangel
Professor
Caltech
1200 E. California Blvd.
228-77
Pasadena, CA 91125
Jonathan Pollock
Chief, GMNRB
NIDA
6001 Executive Blvd.
Bethesda, MD 20892
Russell Ray
Research Fellow
Harvard Medical School
77 Ave Louis Pasteur
Boston, MA 02459
Stephen W. Porges
Professor
University of Illinois at Chicago
1601 W. Taylor St MC 912
Department of Psychiatry
Chicago, IL 60612
Eric M. Reiman
Executive Director
Banner Alzheimer’s Institute
901 E. Willetta Street
Phoenix, AZ 85006
Frank Porreca
Professor
University of Arizona
1501 N. Campbell Avenue
Tucson, AZ 85724
Kathryn J. Reissner
Medical University of South Carolina
173 Ashley Avenue
40 BSB
Charleston, SC 29425
Louis Ptacek
Professor
University of California, San Francisco
1550 Fourth Street
Rock Hall - Room 548
San Francisco, CA 94158
Neil J. Risch
Professor
University of California at San Francisco
Box 0794
513 Parnassus Ave.,
San Francisco, CA 94143
Shaun Matthew Purcell
Assistant Professor
Massachusetts General Hospital
185 Cambridge St.
CPZ-N, Simches 6.254
Boston, MA 02114
Emilie Rissman
Professor
University of Virginia
Department of BMG
Charlottesville, VA 22908
Donald Rainnie
Associate Professor
Emory University
Yerkes Neuroscience Building, Rm 5220
954 Gatewood Road
Atlanta, GA 30329
Laura Roberts
Chairman & Professor
Stanford University School of Medicine
401 Quarry Road
Department of Psychiatry and Behavioral
Sciences
Stanford, CA 94304
349
ACNP 50th Annual Meeting • Final Program
Gavin Rumbaugh
The Scripps Research Institute, Florida
130 Scripps Way
#3C2
Jupiter, FL 33458
Yavin Shaham
Branch Chief and Senior Investigator
IRP-NIDA
Behavioral Neuroscience Branch
251 Bayview Blvd.
Baltimore, MD 21224
Barbara J. Sahakian
Professor
University of Cambridge
Department of Psychiatry (Box 189)
Addenbrooke’s Hospital, Hill’s Road
Cambridge, CB24 8SA
Geoffrey Shoenbaum
Professor
University of Maryland School of Medicine
20 Penn St.
HSF2 S251
Baltimore, MD 21201
Michael William Salter
Senior Scientist/Professor
Hospital for Sick Children
555 University Ave.
Toronto, ON M5G 1X8
Bernard Silverman
Alkermes, Inc.
Medical Affairs
88 Sidney Street
Cambridge, MA 02492
Stephen Scherer
Professor
Hospital for Sick Children
MaRS Ctr., TMDT, 14-701,
101 College Street,
Toronto, ON M5G 1L7
Edward Shorter
University of Toronto
Faculty of Medicine
88 College Street, Room 207
Toronto, ON M5G 1L4
Gunter Schumann
Professor
Institute of Psychiatry
MRC-SGDP Centre
London, SE58AF
Steven Michael Silverstein
Professor
UMDNJ-Robert Wood Johnson Medical
School
151 Centennial Avenue
Piscataway, NJ 08854
Jonathan Sebat
Assistant Professor
UCSD
9500 Gilman Dr.
La Jolla, CA La Jolla
Tracy Simpson
Associate Professor
VA Puget Sound Health Care System
1660 S. Columbian Way
116-WTRC
Seattle, WA 98108
Srijan Sen
Assistant Professor
University of Michigan
5047 BSRB
109 Zina Pitcher Place
Ann Arbor, MI 48109
Yoland Smith
Emory University
7158 MRB III
954 Gatewood Road NE
Atlanta, Georgia 30329
350
ACNP 50th Annual Meeting • Final Program
Beth Stevens
Assistant Professor
Children’s Hospital Boston
300 Longwood Avenue
CLS 12257
Boston, MA 02115
Gustavo Turecki
Associate Professor
McGill University
Douglas Institute
6875 LaSalle Blvd.
Montreal, QC H4H 1R3
Yousin Suh
Associate Professor
Albert Einstein College of Medicine
Departments of Medicine and Genetics
1301 Morris Park Ave. Price 475
Bronx, NY 10461
Jill R. Turner
University of Pennsylvania
3451 Walnut Street
Philadelphia, PA 19104
Peter Uhlhaas
Associate Professor
MPI for Brain Research
Deutschordenstr 46
Frankfurt, 60528
Stephen J. Suomi
Professor/Laboratory Chief
NICHD/NIH
6705 Rockledge Drive
Suite 8030, MSC 7971
Bethesda, MD 20892-7971
Linda Van Eldik
Director, Sanders-Brown Center on Aging
University of Kentucky
Sanders-Brown Center on Aging
800 S. Limestone Street
Lexington, KY 40536
Konrad Talbot
Assistant Professor
University of Pennsylvania
Translational Research Laboratories
125 South 31st Street
Philadelphia, PA 19104-3403
Jeremy Veenstra-VanderWeele
Vanderbilt University
Kennedy Center for Research on Human
Development
7158 MRB III
465 21st Avenue S.
Nashville, TN 37232
Seong-Seng Tan
Professor
The University of Melbourne
Parkville
Parkville, 3010
Eric Vermetten
Associate Professor
Military Mental Health/University Medical
Center
Heidelberglaan 100
Utrecht, 3584 CX
Irene Tracey
Professor
University of Oxford
FMRIB Centre
Nuffield Department Clinical Neurosciences
John Radcliffe Hospital
Oxford, OX3 9DU
Vincent Vialou
Mount Sinai School of Medicine
One Gustave L. Levy Place
New York, NY 10029
Kuei Y. Tseng
Assistant Professor
RFUMS / The Chicago Medical School
3333 Green Bay Rd.
North Chicago, IL 60064
351
ACNP 50th Annual Meeting • Final Program
Michael P. Vitek
Associate Professor
Duke University Medical Center
Campus Box 2900, Research Drive
Bryan Neurosciences Research Building
Durham, NC 27710
Leanne M. Williams
Professor
University of Sydney Medical School
Brain Dynamics Center
Acacia House Westmead Hospital
Sydney, 2145
Brent Alan Vogt
Professor
SUNY Upstate Medical University
750 E. Adams Street
Syracuse, NY 13210
Lois Winsky
National Institute of Mental Health
Division of Neuroscience
MSC 9641, Room 7185
6001 Executive Blvd, NSC
Bethesda, MD 20892
Elaine F. Walker
Professor
Emory University
36 Eagle Row
Atlanta, GA 30322
Jared William Young
Assistant Professor
University of California, San Diego
9500 Gilman Drive
La Jolla, CA 92093-0804
Jon Wallis
Associate Professor
UC Berkeley
132 Barker Hall
MC #3190
Berkeley, CA 94720-3190
L. Trevor Young
Professor and Chair
University of Toronto
1 King College St.
Toronto, ON M5S 1A8
Melissa Warden
Postdoctoral Scholar
Stanford University
Department of Bioengineering
Clark Center, Room W080
318 Campus Drive West
Stanford, CA 94305
Larry Scott Zweifel
Assistant Professor
University of Washington
1959 N.E. Pacific Street
Box 357280
Seattle, WA 98115
Kenneth Warren
Acting Director
National Institute on Alcohol Abuse and
Alcoholism
5635 Fishers Lane
Bethesda, MD 20892
David Weinshenker
Associate Professor
Emory University
Dept. of Human Genetics, Whitehead 301
615 Michael St.
Atlanta, GA 30322
352
ACNP 50th Annual Meeting • Final Program
Notes
353
ACNP 50th Annual Meeting • Final Program
Notes
354
Explanation of Conflict of Interest Disclosure Parts:
Part One: All Financial Involvement with a pharmaceutical or biotechnology company, a
company providing clinical assessment, scientific, or medical companies doing business with or
proposing to do business with ACNP over past 2 years (Jan. 2008-Present)
Part Two: Income Sources & Equity of $10,000 or greater
Part Three: Financial Involvement with a pharmaceutical or biotechnology company, a
company providing clinical assessment, scientific, or medical products or companies doing
business with or proposing to do business with ACNP which constitutes more than 5% of
personal income (Jan. 2008-Present)
Part Four: Grants from pharmaceutical or biotechnology company, a company providing
clinical assessment, scientific, or medical products directly, or indirectly through a foundation,
university, or any other organization (Jan. 2008-Present)
Part Five: My primary employer is a pharmaceutical/biotech/medical device company
ACNP 2011 Council Disclosures
John Csernansky: Sanofi Aventis, Eli Lilly & Company, Part 1
Ronald Duman: Eli Lilly & Company, Lundbeck, Wyeth, Johnson & Johnson, Taisho,
Psychogenics, Pfizer, Bristol Myers Squibb, Forest, Part 1; Taisho, Part 3; Eli Lilly &
Company, Johnson & Johnson, Psychogenics, Pfizer, Bristol Myers Squibb, Repligen, Forest,
Part 4
Alan Frazer: Lundbeck, Takeda, Cyberonics, Part 1; Lundbeck, Takeda, Part 2
Mark Geyer: Acadia, ACNP (Neuropsychopharmacology), Chakra Inc., J&J, Medivation,
Merck, Omeros, San Diego Instruments, Sepracor, Teva, Pfizer (spouse), Astra-Zeneca (spouse),
Part 1; Omeros, San Diego Instruments, Part 2; Intracellular Therapies, Part 4
Anthony Grace: J&J, Taisho, Lundbeck, Pfizer, GSK, Puretech Ventures, Merck, Takeda,
Dainippon Sumitomo, Part 1; J&J, Taisho, Part 2; J&J, Taisho, Part 3; Lundbeck, GSK, Part 4
Herbert Kleber: Abbott, Alkermes, Ascend Media, Neuromed, U.S. World Med, Cephalon,
Gruenthal, J&J, Purdue Pharma, Part 1
David Kupfer: Sevier (spouse), Guilford Press (spouse), Part 1; American Psychiatric
Association, Sevier (spouse), Part 2
David Lewis: BMS, Bioline RX, SK Life Science, Part 1; BMS Foundation, BMS, Curridium
LTD, Pfizer, Part 4
Eric Nestler: PsychoGenics, Merck, Neurologix, Psylin Neurosciences, Part 1; PsychoGenics,
Merck, Neurologix, Epix, Part 2; Astra Zeneca, Part 4
Disclosures
John Krystal: Abbott, Aisling Capital, LLC, Astra Zeneca, Brintall & Nicolini, Inc., BMS,
Easton Associates, Inc., Eli Lilly & Co., F. Hoffman-La Roche Ltd, Gilead Sciences, Inc., GSK,
Janssen, Lohocla, Lundbeck, Medivation, Merz, MK Medical Communications, Naurex, Inc.,
Pfizer, SK Holdings Co., Takeda Industries, Tetragenex, Teva, Transcept, Part 1; Biological
Psychiatry Editor, Part 2; AstraZeneca, Janssen Research Foundation, Part 4
David Rubinow: Dialogues of Clinical Neuroscience, Azevan, Amgen, CME Outfitters
(Chair’s Summit), Part 1; Servier/Dialogues of Clinical Neuroscience, Part 2; Foundation of
Hope, Part 4
Disclosures
Council Members with No Disclosures
Karen Berman
Linda Brady (Council-Elect 2011)
David Braff
Cindy Ehlers
Ronnie Wilkins, Executive Director
ACNP 50th Annual Meeting • Final Program
Explanation of Conflict of Interest Disclosure Parts:
Part One: All Financial Involvement with a pharmaceutical or biotechnology company, a
company providing clinical assessment, scientific, or medical companies doing business with or
proposing to do business with ACNP over past 2 years (Jan. 2008-Present)
Part Two: Income Sources & Equity of $10,000 or greater
Part Three: Financial Involvement with a pharmaceutical or biotechnology company, a
company providing clinical assessment, scientific, or medical products or companies doing
business with or proposing to do business with ACNP which constitutes more than 5% of
personal income (Jan. 2008-Present)
Part Four: Grants from pharmaceutical or biotechnology company, a company providing
clinical assessment, scientific, or medical products directly, or indirectly through a foundation,
university, or any other organization (Jan. 2008-Present)
Part Five: My primary employer is a pharmaceutical/biotech/medical device company
ACNP 2011 Program Committee Disclosures
Anissa Abi-Dargham: BMS-Otsuka, Sunovion, Merck, Lundbeck, Boehsinger-Ingelheim, Part
1; BMS-Otsuka, Part 2; BMS-Otsuka, Part 3; GSK, Part 4
David Baker: Promentis Pharamaceutical, Part 1; Promentis Pharamaceutical, Part 2;
Promentis Pharamaceutical, Part 3; Promentis Pharamaceutical, Part 4;
Randy Blakely: Lundbeck-Scientific Advisory Board, Jubilant Innovation – Scientific
Consultant, Part 1
Joseph Buxbaum: Mount Sinai School of Medicine, Part 2
William Carlezon: The American College of Neuropsychopharmacology, Huya Bioscience
International, Infinity Pharmaceuticals, Lantheus Medical Imaging, Myneurolab.com, The
Society for Neuroscience, Transcept Pharmaceuticals, Part 1; Howard Hughes Medical Institute
(2008-2012), Part 4
Marie-Francoise Chesselet: Michael J. Fox Foundation, Psychogenics, Genentech, BachmannStrauss Foundation, Adolor, Inc., Part 1; Adolor, Inc., Part 2; NIH, Michael J. Fox Foundation,
CHDI, Inc. Allon Therapeutics, Amicus Therapeutics, Isis Pharmaceuticals, Part 4
Michael Davidson: JNJ, Pfizer, Lundbeck, Teva, BiolineRx, Eli Lilly, Sanofi-Aventis, Roche,
GSK, Servier, stock in Tangent Data and BiolineRx, Part 1; Servier, Tangent Data, BiolineRx,
Part 2; Tangent Data, Part 3
Michael Egan: Merck, Part 2
Igor Elman: Astra Zeneca, Eli Lilly and Company, Part 4
Lisa Monteggia: Speaker-Sepracor, Consultant-Sunovion, Part 1
Stephan Heckers: Associate Editor, Archives of General Psychiatry, Part 2
355
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Program Committee Disclosures (continued
Walter Kaye: Astra Zeneca, Part 1; UCSD, Part 2
Richard Keefe: Abbott, Allon, Astellas, AstraZeneca, BiolineRx, BrainCells, BMS, CHDI,
Cypress Bioscience, Eli Lilly, EnVivo, GSK, Lundbeck, Memory Pharmaceuticals, Merck,
NeuroCog Trails, Inc., NeuroSearch, Novartis, Orion, Otsuka, Pfizer, Roche, Shire, Solvay,
Sunovion, Takeda, Wyeth, Part 1; Abbott, BiolineRx, Cypress Bioscience, Eli Lilly, EnVivo,
Lundbeck, Merck, NeuroCog Trials, Inc., Pfizer, Roche, Shire, Sanofi/Aventis, Sunovion, Part
2; NeuroCog Trials, Inc., Part 3; Allon, AstraZeneca, GSK, Novratis, Part 4
Maria Oquendo: Eli Lilly, Bristol Myers Squibb, Janssen, Astra Zenenca, Shire, Part 1; Bristol
Myers Squibb (spouse), Part 3
Kerry Ressler: Burroughs Wellcome Foundation, NARSAD, NIMH, NIDA, Howard Hughes
Medical Institute, Extinction Pharmaceuticals, Part 1; Howard Hughes Medical Institute, Part 2
Darryle Schoepp: Merck, Part 1; Merck, Part 2; Merck, Part 3
David Self: Springer, Neuroscience Letters, Elsevier, Noorik Biopharmaceuticals Ltd., Riehen,
Switzerland, Part 1; Noorik Biopharmaceuticals Ltd., Riehen, Switzerland, Part 2
Arielle Stanford: Elmhurst Hospital, Part 1; NARSAD, Part 4
Trisha Suppes: Jones and Bartlett, Wolters Kluwer--Pharma Soluntions, Part 1; Astra Zeneca,
National Institute of Mental Health, Pfizer Inc., Sunovion Pharmaceutials Inc. (pending), Part 4
Audrey Tyrka: Cyberonics, Medtronic, Neuronetics, Sepracor, UCB Pharma, Lundbeck,
Takeda, Part 1; Cyberonics, Medtronic, Neuronetics, Sepracor, UCB Pharma, Lundbeck,
Takeda, Part 2
Dean Wong: John Hopkins University School of Medicine, Part 2; Amgen, Avid, GE,
Intracellular, Johnson and Johnson, Lilly, Lundbeck, Merck, Orexigen, Otsuka, Roche, Sanofi/
Aventis Part 4
Vanderbilt CME has determined that there is no conflict of interest.
Nothing to Disclose
Ted Abel
Elizabeth Abercrombie
Victoria Arango
Aysenil Belger
Karen Berman
Kristin Cadenhead
Karl Deisseroth
Jay Gingrich
David Goldman
Paul Kenny
Joel Kleinman
Thomas Lehner
Arnold Mandell
Peter Schmidt
Pamela Sklar
Daniel Weinberger
Rachel Yehuda
Carlos Zarate
356
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures
George Aghajanian: State of Connecticut, NIMH RO1 MH-17871, TIAA-CREF, Part 2
Daniela Alberati: F. Hoffmann-La Roche, Part 5
James Anthony: Reckitt-Benckhiser buprenophine post-marketing surveillance and risk
management, Part 1; Retirement mutual funds (TIAA-CREF / Valic), Part 2; NIH/NIDA K05
Senior Scientist and Mentorship Award, NIH/NIDA R01 Research Funding, CISA Foundation,
Sao Paulo, Brazil, Part 4
Raymond Anton: Eli Lilly, Johnson & Johnson, Alkermes, Schering, Lundbeck,Glaxo Smith
Kline, Abbott Labs, Part 1 ACNP, Part 2; Merck, Eli Lilly, Hythiam Inc, Part 4
Ben Barres: Stanford University, Part 2
Sheryl Beck: NIMH, Part 4
Alessandro Bertolino: Eli Lilly, Janssen Pharmaceuticals, Astra Zeneca, Bristol Myers Squibb,
Part 1; Janssen Pharmaceuticals, Astra Zeneca, Eli Lilly, Part 4
Robert Bilder: CHDI, Cypress Bioscience, Inc., Dainippon-Sumitomo Pharma, Hoffman La
Roche, Johnson & Johnson, Merck, Pfizer Inc, Part 1; Johnson & Johnson, Part 2
Floyd Bloom: Alkermes, Inc., Elan Pharmaceuticals, Neotope Inc., Part 1; Alkermes Inc., Elan
Pharmaceuticals, Part 2
Chas Bountra: Grunenthal, Spinifex, Part 1; GSK, Novartis, Merck, Pfizer, Lilly, Part 4
Kathleen Brady: Catalyst Pharmaceuticals, Ovation Pharmaceuticals, Part 4
Nicholas Brandon: Pfizer Inc., Part 1; Pfizer Inc., Part 3; Pfizer Inc., Part 5
David Bredt: Johnson and Johnson, Eli Lilly and Company, Part 1; Johnson and Johnson, Eli
Lilly and Company, Part 2; Johnson and Johnson, Eli Lilly and Company, Part 3; Johnson &
Johnson Pharmaceuticals Group, Eli Lilly and Company, Part 5
Juan Bustillo: Novartis, Part 1
Arthur Caplan: Johnson and Johnson, Glaxo, Merck, Biosen, Part 1
William Carlezon: Leica, Part 1
William Carpenter: Lundbeck, Merck, BMS, Lilly, AstraZeneca, Part 1
Kiki Chang: Lilly, BMS, GSK, Merck,GSK, Part 1; GSK, Part 4
357
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Linda Chang: Neurobehavioral Research, Inc., Part 1; Neurobehavioral Research, Inc., Part 2;
Neurobehavioral Research, Inc., Part 3
Dennis Charney: Pending use patent of Ketamine for the treatment of depression, Part 4
Guang Chen: Johnson & Johnson, Part 1; Johnson & Johnson, Part 2; Johnson & Johnson,
Part 3; Johnson & Johnson, Part 4; Johnson & Johnson, Part 5
Peter Jeffrey Conn: Seaside Therapeutics, Johnson and Johnson, Part 1; Seaside Therapeutics,
Johnson and Johnson, Part 4
Barbara Cornblatt: Merck, Bristol-Meyers Squibb, J & J, Part 1
Kathryn Cunningham: Synosia Therapeutics, Part 4
Anders Dale: CorTechs Labs, Inc., Part 1
Harriet de Wit: Unilever, Part 4
Melissa DelBello: BMS, Eli Lilly, Pfizer, GSK, Amylin, Johnson and Johnson, Somerset,
Merck, Schering Plough, AstraZeneca, Pfizer, Part 1; BMS, Merck, Part 2; BMS, Merck, Part
3; Eli Lilly, Astra Zeneca, Amylin, Part 4
Darin Dougherty: Medtronic, Cyberonics, Eli Lilly, Part 1; Medtronic, Part 2; Medtronic,
Cyberonics, Eli Lilly, Part 4
Wayne Drevets: Pfizer, Eisai, Rules Based Medicine, Johnson & Johnson, Part 1; Oklahoma
University Health Sciences Center, Part 2
Ronald Duman: Lilly, Lundbeck, Wyeth, Johnson and Johnson, Taisho, Psychogenics, Pfizer,
Bristol Myers Squibb, Part 1; Taisho, Part 2; Lilly, Repligen, Lundbeck, Johnson & Johnson,
Part 4
C. Neill Epperson: Shire, Eli Lilly, Johnson and Johnson, Part 1; Shire, Eli Lilly, Novartis,
Part 4; University of Pennsylvania, Part 5
Robert Findling: Abbott, Addrenex, Alexza, AstraZeneca, Biovail, Bristol-Myers Squibb,
Forest, GlaxoSmithKline, Johnson & Johnson, KemPharm Lilly, Lundbeck, Merck,
Neuropharm, Novartis, Noven, Organon, Otsuka, Pfizer, Rhodes Pharmaceuticals, SanofiAventis, Schering-Plough, Seaside Therapeutics, Sepracore, Shire, Solvay, Sunovion, Supernus Pharmaceuticals, Transcept Pharmaceuticals, Validus, Wyeth, Part 1; Shire, Part 2; Abbott,
Addrenex, AstraZeneca, Bristol-Myers Squibb, Forest, GlaxoSmithKline, Johnson & Johnson,
Lilly, Merck, Neuropharm, Otsuka, Pfizer, Rhodes Pharmaceuticals, Schering-Plough, Shire,
Supernus Pharmaceuticals,Wyeth, Part 4
358
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Ellen Frank: Servier International, Guilford Press, Part 1; Servier International, American
Psychiatric Assocation (Spouse), Part 2; Forest Research Institute, Part 3
Guido Frank: NIMH K23 Award, Klarman Foundation Award, Part 4
Adriana Galvan: Philip Morris USA, Part 1
Daniel Geschwind: Biological Psychiatry, Part 2; Repligin, Part 4
Richard Gibbs: SeqWright, Inc; Life Technologies, Part 1; Baylor College of Medicine;
SeqWright Inc, Part 2; SeqWright, Inc, Part 3
John Gilmore: Sunovion Pharmaceuticals, Part 4
Donald Goff: Indevus Pharmaceuticals, H. Lundbeck, Schering-Plough, Eli Lilly, Takeda,
Biovail, Solvay, Hoffman- La Roche, Cypress, Dainippon Sumitomo, Bristol-Meyers Squibb,
Abbott Laboratories, Takeda, Genentech, Merck, Endo Pharmaceuticals, Otsuka, Pfizer,
Novartis, Janssen, GlaxoSmithKline, Part 1; Pfizer, Novartis, Janssen, GlaxoSmithKline, Part 4
Terry Goldberg: Merck, GSK, Part 1; Pfizer/Eisai, Part 4
Anthony Grace: Johnson & Johnson, Lundbeck, Pfizer, GSK, Puretech Ventures, Merck,
Takeda, Dainippon Sumitomo, Puretech Ventures, EMD Serano, NeuroSearch, Part 1; Johnson
& Johnson, Part 2; EMD Serano, Lundbeck, GSK, Neurosearch, Part 4
Steven Grant: Marriot, Inc, Apple, Inc,TIAA-CREF, Federal Thrift Savings Pla, Part 2
John Greden: Eli Lilly, Merck, Neuronetics, Informed Medical Decision Making Foundation,
Part 1
Henry Greely: Celera Corporation, Part 1; Permanente Medical Group(Spouse), Part 2;
Stanford University, Part 5
Michael Green: Abbott Laboratories, Cypress, Dainippon Sumitomo Pharma, Lundbeck,
Otsuka, Sunovion, Sanofi-aventis, Takeda, Teva, Janssen-Cilag, Otsuka, Sunovion, Part 1
Raquel Gur: Pfizer, AstraZeneca, Part 4
Ruben Gur: Johnson & Johnson, Part 1; Medical legal consultations to US courts, federal and
state public defenders, Part 2; Current Designs, Inc., Part 3 Pfizer, Merck, AstraZeneca, Part 4
Chang-Gyu Hahn: Asztrazeneca, GSK and Phizer, Part 1; Asztrazeneca, GSK and Phizer, Part 4
Richard Huganir: Millipore Corporation,The Johns Hopkins University, R.L.H, Part 1;
Millipore Corporation,The Johns Hopkins University, R.L.H Part 2
359
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Steven Hyman: Harvard University, Part 2
Daniel Javitt: Schering-Plough, Takeda, NPS Pharma, Solvay, Sepracor, AstraZeneca, Pfizer,
Cypress, Merck, Sunovion, Eli Lilly, BMS, Pfizer, Roche, Jazz, Promentis, Glytech, Part 1;
Pfizer, Glytech, Part 2; Pfizer, Glytech, Part 3; Pfizer, Roche, Jazz, Part 4
Michael Jackson: Ortho McNeil Janssen Pharmaceuticals, Part 1; Ortho McNeil Janssen
Pharmaceuticals, Part 4
David Janowsky: QRx Pharma, Part 1; QRx Pharma, Part 2
Michael Kaplitt: Neurologix, Cornell University, Part 1; Neurologix, Part 2; Neurologix, Part 3; Neurologix, Part 4
Walter Kaye: Astra Zeneca, Denver Eating Disorder Center, Part 1; Astra Zeneca, Part 4
Lawrence Kegeles: Pfizer, Amgen, Part 1; Columbia University NY State Psychiatric Institute,
Part 2; Pfizer, Amgen, Part 4
Robert Kessler: Merck, Part 3; Novo Nordisk, Part 4
George Koob: Addex Pharmaceuticals, Alkermes, Arkeo Pharmaceuticals, Casa Palmera,
Embera Neurotherapeutics, GlaxoSmithKline, Lilly, Psychogenics, Part 1
Stephen Koslow: Brain Resource, Biomedical Synergy, Enhanced Pharaceuticals, Part 1;
American Foundation for Suicide Prevention, Brain Resource Ltd, Part 2; Brain Resource Ltd,
Part 3; Self Employed, Part 5
Thomas Kosten: NIH, Baylor, VAMC, Reckitt Benckizer, Catalyst Pharma, Part 2
Henry Kranzler: Gilead Sciences, GlaxoSmithKline, Alkermes, Inc. Eli Lilly, Janssen,
Schering Plough, Lundbeck, Alkermes, GlaxoSmithKline, Abbott, Johnson & Johnson,
ACTIVE, Part 1; Alkermes, Part 2; Merck & Co, Part 4
Louis Kunkel: SynapDx, Part 1; SynapDx, Part 2
David Kupfer: American Psychiatric Association, Part 2
Bernard Le Foll: Pfizer, Richter Pharmaceutical, Part 1; Pfizer, Part 3; Pfizer, Part 4
James Leckman: Talecris, Biotherapeutics, Part 1; Talecris, Biotherapeutics, Part 4
Robert Lenox: Sanofi-aventis Pharmaceuticals, Jubilant Innovation, QRxPharma, Part 1;
Sanofi-aventis Pharmaceuticals, Jubilant Innovation, QRxPharma, Part 2; Sanofi-aventis
Pharmaceuticals, Jubilant Innovation, Part 3
360
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Pat Levitt: Pediatric Biosciences, Puretech, Bioventures, Part 1
David Lewis: AstraZeneca, BioLine RX, Bristol-Myers Squibb, Merck, Neurogen, SK Life
Science, Part 1; BMS Foundation, Bristol-Myers Squibb, Curridium LTD, Pfizer, Part 4
Xiaohua Li: NIH, NARSAD, AFSP, Corcept, Otsuka, Novartis, OMJ, Part 1; NIH, NARSAD,
AFSP, Corcept, Otsuka, Novartis, OMJ, Part 2
Jeffrey Lieberman: Bioline,GlaxoSmithKline, Intracellular Therapies, Eli Lilly, Pierre Fabre,
Psychogenics, Part 1; Allon, GlaxoSmithKline, Eli Lilly, Merck, Novartis, Pfizer, Roche,
Sepracor, Tragacept, Part 4
Sarah H. Lisanby: ANS/St. Jude Medical, Neuronetics, Cyberonics, Brainsway, Magstim,
MagVenture, Part 4
Robert Mahley: Merck, Part 1; The J. David Gladstone Institutes, Part 2; Merck, The J. David
Gladstone Institutes, Mahley laboratory, Part 4
Robert Malenka: Pfizer, Inc., Part 1; Pfizer, Inc., Part 2
Husseini Manji: Johnson and Johnson, Part 1; Johnson and Johnson, Part 2; Johnson and
Johnson, Part 3; Johnson & Johnson Pharmaceuticals Group, Part 5
Helen Mayberg: St. Jude Medical Inc., Part 1; St. Jude Medical Inc., Part 2; St. Jude Medical
Inc., Part 3
John March: Pfizer, Lilly, GlaxoSmithKline, BMS, Johnson and Johnson, Psymetrix, Attention
Therapeutics, Avenir, Alkiermes, Vivus and MedAvante, Eli Lilly, Pfizer; Eli Lilly, Pfizer,
NARSAD, NIMH, NIDA, Part 1; MultiHealth Systems, Part 2; Pfizer, Part 4
Stephen Marder: Pfizer, Abbott, Roche, Genetech, Amgen, Otsuka, Lundbeck, MedAvante,
Part 1; GSK, Novartis, Psychogenics, Part 4
Dan Mathalon: Pfizer, AstraZeneca, Part 1; AstraZeneca, Part 4
Colleen McClung: GlaxoSmithKline, Pfizer, Servier, Part 1; GlaxoSmithKline, Pfizer, Part 4
Christopher McDougle: Bristol-Myers Squibb Co., Part 1; Bristol-Myers Squibb Co., Part 2;
Bristol-Myers Squibb Co., Part 3; Bristol-Myers Squibb Co., Part 4
James Meador-Woodruff: Editor-in-chief Neuropsychopharmacology, Part 2
Herbert Meltzer: ACADIA,Amgen,BioLIne Rs, Dainippon Sumitomo, Cypress, Janssen,
EnVivo, Novartis, Otsuka, Roche, Sunovion, Part 1; Dainippon, EnVivo,Forest, Roche, Otska,
Part 4
361
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Emilio Merlo-Pich: GSK, Part 1; GSK, Part 2; GSK, Part 3; GlaxoSmithKline, Part 5
Michael Merzenich: DNA, Part 1; Posit Science Corporation, Brain Plasticity Institute, Scientific
Learning Corporation, Part 2; DNA, Part 3; DNA, Part 4; Posit Science Corporation, Part 5
Roger Meyer: Northwestern University, PennState, Pfizer, AstraZeneca, Dainippon Sumitomo,
Eli Lilly, Forest Research Institute, Janssen, Division of Ortho-McNeil-Janssen Pharmaceuticals,
Inc, administered by Ortho-McNeil-Janssen Scientific Affairs, LLC, Roche Pharmaceuticals,
Sanofi-Aventis, Schering-Plough, Sepracor; and United BioSource, Part 1; Best Practice Project
Mgmt, Inc. Penn State Hershey Medical Center, TIAA CREF, Part 2; Northwestern University,
PennState, Forest, Pfizer, Part 4
Andreas Meyer-Lindenberg: Roche, Pfizer, AstraZeneca, J+J, Novartis, Part 1; EU Innovative
Medicine Initative, Part 4
David Michelson: Merck and Co, Part 1; Merck and Co, Part 2; Merck and Co, Part 3; Merck
and Co., Part 5
Emmanuel Mignot: Jazz Pharmaceutical, Merck, GSK, Part 1
Andrew Miller: Abbott Laboratories, AstraZeneca, Centocor Inc., GlaxoSmithKline, Lundbeck
Research USA, F. Hoffmann-La Roche Ltd., Schering-Plough Research Institute (now Merck)
and Wyeth/Pfizer Inc, Part 1; Centocor Inc., GlaxoSmithKline, and Schering-Plough Research
Institute (now Merck), Part 4
Lisa Monteggia: Sepracor, Part 1;
Holly Moore: Lilly, Pfizer, Part 1
Stephen Moss: Pfizer, Part 3; Pfizer, Astrazeneca, Part 4
Robin Murray: AZ, BMS, Lilly, Janssen, Novartis, Part 1
Aye-Mu Myint: MOODINFLAME, Advanced Practical Diagnostics Part 2; Advanced Practical
Diagnostics, Part 5
Raj Narendran: GlaxoSmithKline, Sunovion (Sepracor), Part 4
Charles Nemeroff: American Psychiatric Publishing, Astra Zeneca, SK Pharma, Novadel
Pharma, Cenerx, Xhale, Pharmaneuroboost, Part 2
Eric Nestler: PsychoGenics, Merck, Psylin Intracellular Therapies, Neurologix, Part 1; Mount
Sinai School of Medicine, PsychoGenics,Merck, Neurologix, Part 2; Astra-Zeneca, Part 4
Thomas Neylan: Supply of Phase III drug (almorexant) from Actelion for DoD funded
study. Supply of Phase II drug (GSK561579) from Glaxo Smith Kline for a VA funded study,
Part 4
362
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Jeffrey Nye: Johnson and Johnson, Part 1; Johnson and Johnson, Part 2; Johnson and Johnson,
Part 3; J&J, Part 4; Johnson and Johnson, Part 5
Charles O’Brien: Alkermes, Reckitt, Catalyst, Part 1; University of Pennsylvania, Part 2
Stephanie O’Malley: American College of Neuropsychopharmacogy, Alkermes, Abbott
Laboratories, Eli Lilly & Company, GlaxoSmithKline, Johnson & Johnson Pharmaceuticals,
Lundbeck, Schering Plough, Applied Behavioral Research, Nabi Biopharmaceuticals, Gilead
Pharmaceuticals, Alkermes, GlaxoSmithKline, Brown University, University of Chicago,
Scientific Panel of Advisors, Hazelden Foundation, Part 1; NABI, Part 4
Eric Olson: Vertex Pharmaceuticals, Part 1; Vertex Pharmaceuticals, Part 2; Vertex
Pharmaceuticals, Part 3; Vertex Pharmaceuticals, Part 5
Anand Pandya: Janseen, Part 1
Steven Paul: Sigma Aldrich, Alnylam Pharmaceuticals-, Constellation Pharmaceuticals,Seaside
Therapeutics, Third Rock Ventures, Karuna, Part 1; Sigma Aldrich, Alnylam Pharmaceuticals-,
Constellation Pharmaceuticals,Seaside Therapeutics, Third Rock Ventures, Karuna, Weill
Cornell Medical College, Part 2
Diana Perkins: Lilly, Sunovion, Merck, Genentech, Endo Pharmaceuticals,Lilly, Part 1; Lilly,
Part 2
Mary Phillips: Cardiff University, Part 2
Jean-Baptist Poline: IPSEN Laboratory, Part 4
Frank Porreca: NeurAxon, Grunenthal, Part 1; NeurAxon, Grunenthal, Part 4
William Potter: Amgen, Bristol Myers Squibb, Elan, En Vivo, J & J, Medavante, Orasi, Pfizer
Roche, Theravance, Part 1; Merck, Part 2; Merck, Part 5
Mark Rapaport: Clintara, OND-003 Transcept Study SIV, Part 4
Mark Rasenick: Pax Neuroscience, Eli Lilly, Lundbeck, Sepracor, Part 1; Eli Lilly, Lundbeck,
Part 4
Scott Rauch: Vanderbilt University, VA Roundtable, Oxford Press, APPI, Wolters Kluwer, ACNP, UTSW Dallas Grand Rounds, Cleveland Clinic Grand Rounds, NIMH Conte Center
Advisory Board, Oxford University Press, APPI, Hall Mercer, Oxford University Press, NIMH
RDoC, APPI, Part 1; McLean Hospital/Partners Healthcare, Part 2; McLean Hospital/Partners
Healthcare, Part 3; Cyberonics, Medtronic, Northstar Neuroscience, Part 4
363
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Eric Reiman: Amnestix/Sygnis, AstraZeneca, Bayer, Eisai, Elan, Eli Lilly, GlaxoSmithKline,
Intellect, Link Medicine, Novartis, Siemens, Takeda, Part 1; AstraZeneca, Avid, Kronos Life
Sciences National Institute on Aging, Anonymous Foundation, Nomis Foundation, Banner
Alzheimer’s Foundation, State of Arizona, Part 4
Kerry Ressler: Extinction Pharmaceuticals, Part 1
Robert Ring: Pfizer, Part 1; Pfizer, Part 2; Pfizer, Part 3; Pfizer, Part 4; Pfizer, Part 5
Trevor Robbins: Cambridge Cognition, Pfizer, Lilly, Lundbeck, GlaxoSmithKline, Part
1; Pfizer, Cambridge Cognition, Part 2; Pfizer, Cambridge Cognition, Part 3; Lundbeck,
GlaxoSmithKline, Lilly, Part 4
Laura Roberts: Medical College of Wisconsin, Terra Nova Learning Systems, Stanford
University, Part 2
Brian Roth: Mediavation, Galenea Pharmaceuticals, Nereus Pharmaceuticals, Venrock
Investments, Merck, AMRI, Invitrogen, Part 1; Finnegan Law Firm, Part 2
John Rush: Otsuka, University of Michigan and Brain Resource, Guilford Publications,
University of Texas Southwestern Medical Center, Part 1; Brain Resource, Part 2
Barbara Sahakian: Cambridge Cognition, Novartis, Shire, GlaxoSmithKline, Lilly,
Boehringer-Ingelheim, Hoffmann-La Roche, Massachusetts General Hospital, Medical Research
Council Neurosciences and Mental Health Board, Science Co-ordination Team for the Foresight
Project on Mental Capital and Wellbeing European Research Council, Journal of Psychological
Medicine, Part 1
Mary Sano: Medivation, Bayer, Medpace, Pfizer, Takeda, United Biosource, Part 1
Akira Sawa: Astellas, Tanabe-Mitsubishi, Takeda, Part 4
Alan Schatzberg: Stanford University, American Psychiatric Association, PharmaNeuroBoost,
Neuronetics, Somaxon, Amnestix, BrainCells, CeNeRx, Corcept, Novadel, Jazz, Lundbeck,
McKinsey; Sanofi-Aventis; Synosia, CNS Response, Eli Lilly, GSK, Takeda, Pfizer,
Neurocrine, Merck, Forest, Part 1; Stanford University, American Psychiatric Association,
PharmaNeuroBoost, Neuronetics, Amnestix, BrainCells, CeNeRx, Corcept Therapeutics,
Forest, Neurocrine, Pfizer, Somaxon, BrainCells, Merck, Synosia, Part 2; American Psychiatric
Association; PharmaNeuroBoost, Part 3
Darryle Schoepp: Merck and Company, Inc, Part 1; Merck and Company, Inc, Part 2; Merck
and Company, Inc, Part 3; Merck and Company, Inc., Part 5
364
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Nina Schooler: Astra Zeneca, Bristol Meyers Squibb, Eli Lilly & Company, Hoffman LaRoche,
H. Lundbeck, Merck, Johnson and Johnson, OrthoMcNeil Janssen, Pfizer, Shire, Part 1; Astra
Zeneca, Bristol Meyers Squibb, Eli Lilly & Company, H. Lundbeck, OrthoMcNeil Janssen,
Pfizer, Part 4
Yavin Shaham: NIH, Part 5
Edward Shorter: Faculty of Medicine, University of Toronto, the Social Sciences and
Humanities Research Council of Canada (SSHRC), the Canadian Institutes for Health Research
(CIHR), Part 4; Faculty of Medicine, University of Toronto, Part 5
Steven Siegel: NuPathe, Merck, Part 1; NuPathe, Merck, Part 2; Nupathe, Part 3
Bernard Silverman: Alkermes, Part 1; Alkermes, Part 2; Alkermes, Part 3
Steven Silverstein: AstraZeneca, Part 1; AstraZeneca, Part 4
Phil Skolnick: DOV Pharmaceutical, Inc. Sepracor, Part 1; DOV Pharmaceutical, Inc, Part 2;
DOV Pharmaceutical, Inc, Part 3
Gwenn Smith: Pfizer, Part 1; Pfizer, Part 4
Matthew State: Yale University, Athena Diagnostics, Part 1
Murray Stein: Journal deputy editorship: Depression and Anxiety, Up To Date Part 1; Journal
deputy editorship: Depression and Anxiety, Up To Date Part 2
Beth Stevens: Ellison Medical Foundation Dana Foundation Smith Family Foundation, Part 4
James Swanson: Noven, Johnson and Johnson, Part 1
Susan Swedo: Talecris Biotherapeutics, Part 4; Employee of US Federal Government, Part 5
John Sweeney: Takeda, Pfizer, Janssen, Part 1; Janssen, Part 4
Carol Tamminga: Intracellular Therapies, PureTech Ventures, Eli Lilly Pharmaceutical,
Sunovion, Astellas, Cypress Bioscience, Merck, International congress on Schizophrenia
Research, NAMI, APA, Finnegan, Henderson, Farabow,Garrett & Dunner, LLP, Part 1; UTSW,
APA, Intracellular Therpies Finnigan Henderson, Part 2
Seong-Seng Tan: The University of Melbourne, Part 5
Irene Tracey: Pfizer, Lilly, Grunenthal, Part 1; Pfizer, IMI Consortium, Part 4
Kuei Y. Tseng: Rosalind Franklin University, NIH R01-MH086507, Part 4
365
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Jeremy Veenstra-VanderWeele: Seaside Therapeutics, Roche Pharmaceuticals, Novartis, Part
1; Seaside Therapeutics, Roche Pharmaceuticals, Novartis, Part 4
Michael Vitek: Duke, Cognosci, NIH, Part 1; NIH,IRS, Part 2; Duke, Cognosci, Part 3;
Cognosci, Inc., Part 5
Gene-Jack Wang: Orexigen Therapeutic Inc., Part 4
Rikki Waterhouse: Abbott Labs, Part 2; Abbott Labs, Part 5
David Weinshenker: Pending license on a patent concerning the use of selective dopamine
beta-hydroxylase inhibitors for the treatment of cocaine dependence, Part 1
Myrna Weissman: Multi Health Systems, Part 3
Leanne Williams: Brain Resource Ltd., Pfizer, Part 1; Brain Resource Ltd, Part 2
Dean Wong: Amgen, Part 1; Johns Hopkins University School of Medicine, Part 2; Amgen,
Avid, BMS, GE, Intracellular, Johnson and Johnson, Lilly, Lundbeck, Merck, Orexigen, Otsuka,
Roche, Sanofi-Aventis, Part 4
Rachel Yehuda: Eli Lilly Pharmaceutical, Part 4
L. Trevor Young: Eli Lilly, Astra-Zeneca, Part 3
Joseph Zohar: Lundbeck, Servier, Solvay/Abbot, Sanofi-Aventis, Bristol Myer Squibb, Part 1;
Lundbeck, Servier, Part 4
Charles Zorumski: NIMH, NIAAA, NIGMS, NINDS Bantly Foundation, Part 4
366
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Following Faculty Had No Disclosures:
Ted Abel
Jean Addington
Schahram Akbarian
Cristina Alberini
David Amaral
Noelle Anastasio
Susan Andersen
Mark Ansorge
Paul Appelbaum
Ottavio Arancio
Paola Arlotta
Nicole Avena
Thomas Ban
Dewleen Baker
Debra Bangasser
Carrie Bearden
Sheryl Beck
Francesco Benedetti
Kent Berridge
Kristin Bigos
Barry Blackwell
John Blangero
David Borsook
Linda Brady
Stephen Brimijoin
Camron Bryant
Neil Buckholtz
William Bunney
Claudia Buss
Joseph Buxbaum
Kristin Cadenhead
Vince Calhoun
Tyrone Cannon
Mario Capecchi
Marilyn Carroll
Sue Carter
Xavier Castellanos
Sohini Chowdhury
De-Maw Chuang
Jessica Connelly
Kelly Conrad
Mark Cookson
Rita Cowell
Bruce Cuthbert
John Davis
Michael Davis
Karl Deisseroth
Ralph DiLeone
Joel Elkes
Emad Eskandar
Damien Fair
Daniel Falk
Ruth Feldman
Dianne Figlewicz
Max Fink
Kyle Frantz
Robert Freedman
Samantha Fung
Aurelio Galli
Wenbiao Gan
Amy Garrett
Shaoyu Ge
Andre Gentil
Elliot Gershon
Samuel Gershon
Jay Giedd
Jay Gingrich
David Glahn
David Goldstein
Lawrence Goldstein
Rita Goldstein
Guillermo Gonzalez-Burgos
Ann Graybiel
Benjamine Greenberg
Christopher Gregg
Eugenia Gurevich
Suzanne Haber
Ming-Hu Han
James Harris
Vahram Haroutunian
Kenji Hashimoto
Jonathan Hollander
Andrew Holmes
William Horan
Elaine Hsiao
Christina Hultman
Kent Hutchison
Thomas Hyde
367
Thomas Insel
Sharna Jamadar
J. David Jentsch
Sheena Josselyn
Tanja Jovanovic
Peter Kalivas
Atsushi Kamiya
Maria Karayiorgou
Takaoki Kasahara
Martin Katz
Patrick Kennedy
Paul Kenny
Matcheri Keshavan
Donald Klein
Joel Kleinman
Barbara Koenig
Conan Kornetsky
Ann Kring
Gonzalo Laje
Story Landis
Francis Lee
Michael Lehmann
Thomas Lehner
Ellen Leibenluft
Frances Leslie
Barbara Lipska
Chunyu Liu
Mary Kay Lobo
Jamie Maguire
Anil Malhotra
Roberto Malinow
Charles Marmar
Karen G. Martinez
Venkata Mattay
Jennifer McCormick
Michael Meaney
Peter Meerlo
Kathleen Merikangas
Bita Moghaddam
John Morrison
Karim Nader
Michael Nader
Kazutoshi Nakazawa
Roberta Ness
ACNP 50th Annual Meeting • Final Program
ACNP 2011 Presenter Disclosures (continued)
Following Faculty Had No Disclosures:
John Neumaier
Alexander Neumeister
Amy Newman
Ralph Nixon
Patricio O’Donnell
Scott Orr
Godfrey Pearlson
Jamie Peters
Daniel Pine
Roger Pitman
Jonathan Pollock
Stephen Porges
Frank Porreca
William Potter
Louis Ptacek
Shaun Purcell
Donald Rainnie
Antonio Rangel
Judith Rapoport
Russell Ray
Kathryn Reissner
Kerry Ressler
Victoria Risbrough
Neil Risch
Emilie Rissman
Lorna Role
Gavin Rumbaugh
Michael Salter
Carl Salzman
Stephen Scherer
Thomas Schulze
Gunter Schumann
Jonathan Sebat
Larry Seidman
Srijan Sen
Yavin Shaham
Anantha Shekhar
Geoff Shoenbaum
Tracy Simpson
Yoland Smith
Yousin Suh
Fridolin Sulser
Stephen Suomi
Konrad Talbot
368
Gustavo Turecki
Jill Turner
Peter Uhlhaas
Rita Valentino
Linda van Eldick
Eric Vermetten
Vicent Vialou
Nora Volkow
Brent Vogt
Elaine Walker
Jonathan Wallis
Melissa Warden
Kenneth Warren
Cynthia Weickert
Daniel Weinberger
David Weinshenker
Lois Winsky
Jared Young
Lei Yu
Carlos Zarate
R. Suzanne Zukin
Larry Zweifel
Author Index
Author Index
ACNP 50th Annual Meeting • Final Program
Aarde, Shawn A.
Abazyan, Bagrat
Abazyan, Sofya
Abbott, Chris
Abbs, Brandon
Abdallah, Chadi G.
Abdel-Hamid, Mona
Abel, Ted
Abelson, James
Abend, Rany
Abercrombie, Heather C.
Aberdam, Daniel
Abi-Dargham, Anissa
Abrams, Debra J.
Abulseoud, Osama A.
Aceti, Max
Acheson, Shawn K.
Achtyes, Eric
Adams, David H.
Adams, Matthew
Addington, Jean
Adelekun, Adesewa E.
Adleman, Nancy E.
Adler, Caleb
Adler, Lenard
Adli, Mazda
Agam, Galila
Agarwal, Vishesh
Aghajanian, George
Agid, Ofer
Agster, Kara L.
Ahmari, Susanne E.
Ahmed, Saeeduddin
Ajilore, Olusola
Akbarian, Schahram
Akce, Leyla
Akil, Huda
Akiskal, Hagop S.
Akula, Nirmala
Al-Hasani, Ream
Alarcon, Gabriela
Alberati, Daniela
Alberini, Cristina
Alburges, Mario
Albusaysi, Salwa
Alcantara, Lyonna F.
Alda, Martin
Alegria, Dylan
336
266
266
319
59, 297
241
240
114, 269
242, 326
232
247
267
57, 58, 109, 287
237
307
224
295
129, 271
239
303
102, 274
334
320
247, 292, 320
271
248
224, 328, 332
77, 250
207
230
223
221
230, 231
320, 322
177, 235
61, 234
41, 222, 241
329
236
335
283
205, 334
97, 215
257
236
333
321
323
Aleman, Gabriela Gonzalez
Alexander-Bloch, Aaron F.
Alexander, Kathleen S.
Algorta, Guillermo Perez
Alia-Klein, Nelly
Alim, Tanya
Allard, Carolyn B.
Allen, John
Allen, Katherine
Allen, Wade M.
Almeida, Jorge R. C.
Almli, Lynn M.
Almog, Orna
Altshuler, Lori
Aluisio, Leah
Alvarez-Estrada, Miguel
Aman, Michael G.
Amar, Shirly
Amaral, David
Amir, Nader
Amita, Hidetoshi
Anand, Amit
Anand, Reena
Anastasio, Noelle
Anders, Martin
Andersen, Susan L.
Anderson, Adam
Anderson, Colleen
Anderson, George
Anderson, Jeffery
Anderson, Stewart J.
Anderson, Susan
Andreasen, Nancy
Angelakos, Christopher C.
Angrish, Deepshikha
Angstadt, Mike
Ansari, Mohammad S.
Ansorge, Mark
Anthony, James
Anton, Raymond
Anwar, Salman
Aparicio, Mark
Appasamy, Martin
Appelbaum, Paul
Apple, Deana M.
Applebaum, Julia
Apud, Jose A.
Arad, Michal
369
238
284
264
329
316
294
241
54, 256, 322
240
251
290, 322
279
332
234, 276, 322
258
254
270
328
183
242
338
245
321
112, 296
250
335
251
309
261
246
233
200
237
261
336
231
335
210, 223
31, 32
104, 272
272
305
222
101, 235
332
328
299
261
ACNP 50th Annual Meeting • Final Program
Arancio, Ottavio
Arango, Victoria
Archer, Clay
Archer, Melanie
Archie, Meena
Arevalo, Jesusa
Arguello, Amy
Arienzo, Donatello
Aris, Virginie
Arizpe, Joseph
Arkow, Michelle
Arlotta, Paola
Armitage, Roseanne
Arnedt, J. Todd
Arnold, L. Eugene
Arrant, Andrew
Arvanitis, Lisa
Åsberg, Marie
Ascher-Svanum, Haya
Asgharnejad, Mahnaz
Ashworth, Judy B.
Atack, John R.
Aton, Sara J.
Atzil, Shir
Aupperle, Robin L.
Aurelian, Laure
Avena, Nicole
Avery, Erich
Ayalew, Mikias
Azab, Abed
Azar, Marc R.
Baarendse, Petra J.J.
Baca-Garcia, Enrique
Baddam, Suman
Baer, Lee
Baganz, Nicole
Baghal, Basel
Bahn, Sabine
Bailer, Ursula F.
Bailey, Christopher
Bailine, Samuel
Baillie, George S.
Bakalian, Mihran J.
Bakchine, Serge
Baker, Brandi J.
Baker, Chris
Baker, David A.
Baker, Dewleen
187
225
303
323, 327
324
281
257
287, 322
236
233
311
181
259
259
270
221, 295
309
247
228
292
250
258
269, 337
311
241, 242
249
98, 199
322
278
328
258
336
281
279
328
225
238
119, 262
282
232, 242
327
269
225
269
304
233
299
102, 337
Baker, Karen
Baker, Ross A.
Bakshi, Vaishali P.
Balaraman, Yokesh
Balcita-Pedicino, Judith
Baldwin, Don
Bali, Purva
Ball, Susan
Baller, Erica B.
Balu, Darrick T.
Ban, Thomas A.
Banasr, Mounira
Bangaru, Saroja
Bangasser, Debra
Bao, Y.
Bar-Haim, Yair
Baraban, Jay M.
Barapour, Layla
Barbier, Estelle
Barch, Deanna M.
Barchas, Jack D.
Barnea-Goraly, Naama
Baros, Alicia
Barr, Cathy
Barr, Christina S.
Barres, Ben
Barrios, Brandon
Barsky, Maria
Barton, Bruce A.
Basheer, Radhika
Basu, Aditi
Bates, Andrew T.
Bath, Kevin
Bato, Angelica A.
Bauer, Michael
Baumann, Pierre
Bavamian, Sabine
Bearden, Carrie
Beaulieu, Ashley
Bechtholt-Gompf, Anita
Beck, Sheryl
Becker, Eni S.
Becker, Howard C.
Bedi, Gillinder
Beesdo-Baum, Katja
Begley, Amy
Behr, Guilherme A.
Behrens, M. Margarita
370
263
308, 310
221
278
338
315
305
328
283, 318
264
201
263
321
209
277
232
327
323
37, 315
274
241
323
272
281
315
185
270
308
320
39, 227
288
223
48, 277
312
248, 307
251
268
163, 284
307
261
210, 224
248
256, 302, 306
253, 313
248
233
227
266
ACNP 50th Annual Meeting • Final Program
Behrooznia, Michelle
Behzadi, Arian
Belálcazar, Helen
Belger, Aysenil
Belin, David
Bell, Chris
Bell, Lauren
Bellelli, Giuseppe
Bellgowan, Patrick S.F.
Bellocchio, Elizabeth E.
Belmaker, R.H.
Ben-David, Shelly
Ben-Shachar, Dorit
Ben-Shahar, Osnat
Benedek, Irma H.
Benedetti, Francesco
Benedict, Melissa
Benítez, Bruno A.
Bennett, Ryan S.
Bentley, Graham D.
Bergeron, Richard
Bergman, Jack
Berlin, Heather A.
Berman, Karen
Berman, Robert M.
Berman, Steven
Bernstein, Ira
Berridge, Kent
Bersudsky, Yuly
Bertolino, Alessandro
Berton, Olivier
Bertrand, Daniel
Bertrand, Sonya
Betancur, Catalina
Bethea, Cynthia L.
Beurel, Eleonore
Beveridge, Thomas
Bevilacqua, Laura
Bhagwagar, Zubin
Bhatia, Kamal
Bhatt, Mahima
Bhuvaneswaran, Chaya
Bidzan, Leszek
Bigelow, Llewellyn B.
Bigos, Kristin
Bilder, Robert
Bilello, John A.
241
324
238
326
303
288
258
269
234
308, 310
224, 328
274
267
258, 304
250
114
247
238
253
273
330
257
231, 307
246, 278, 283,
298, 318
310
248
234
199
328
111
224
255
255
35, 280
326
224, 295
258
281, 316
70, 255, 291
257
291
74, 251, 255
269
318
218, 240
215, 312
247
Billingslea, Eileen
Bilsky, Edward
Binder, Elisabeth B.
Bishop, Jeffrey R.
Biskup, Caroline Sarah
Bivens, Nancy M.
Bixler, Edward
Bjugstad, Kimberly B.
Black, Sandra E.
Blacktop, Jordan M.
Blagdon, Ryan
Blakely, Randy
Blanco, Carlos
Blangero, John
Bleakman, David
Blendy, Julie A.
Blennow, Kaj
Blier, Pierre
Blitzer, David N.
Bliwise, Nancy
Blizinsky, Katherine
Bloch, Michael
Bloch, Yehudit
Blom, Thomas J.
Bloom, Floyd
Blouin, Ashley
Blum, Justine
Blumenthal, Jonathan
Boada, Fernando
Bodurka, Jerzy
Boellaard, Ronald
Boerwinkle, Eric
Bogdan, Ryan
Bogerts, Bernhard
Boggs, Douglas L.
Boileau, Isabelle
Bolaños-Guzmán, Carlos
Boldrini, Maura
Bolivar, Valerie J.
Bolo, Nicolas R.
Bonaventure, Pascal
Borckardt, Jeffrey J.
Borsook, David
Bortz, David
Bose, Anjana
Bossong, Matthijs M.
371
240
296
279
235
221
115, 127, 128,
264
297
328
269
302
321
225
269
111
263
315
226
192, 330
123, 273
274
287
57, 70, 71, 240,
291
328
292
211
327
282
244
328
234
287
280
236
66, 249
228
288, 319, 324
333
225
316
286
258
249
189
264
307
287
ACNP 50th Annual Meeting • Final Program
Bostrom, John A.
Boulter, Jim
Bountra, Chas
Bowden, Charles
Brachman, Rebecca A.
Bradaia, Amyaouch
Bradley, Bekh
Brady, Kathleen
Brady, Linda
Braff, David L.
Braga, Raphael
Brand, Eva J.
Brand, Jesse
Brandon, Nicholas
Brasic, James
Braun, Ashley
Bravova, Margarita V.
Bredemann, Teruko M.
Bredt, David
Bredy, Timothy W.
Breed, Mason C.
Breier, Alan
Breier, Michelle R.
Brenhouse, Heather C.
Brennan, Brian P.
Brennan, Molly
Brent, David
Brimijoin, Stephen
Bristow, Linda J.
Britton, Jennifer C.
Brockmann, Marisa
Brody, Arthur L.
Brohawn, David G.
Brooks, John O.
Brooks, William
Brotman, Melissa A.
Brower, Kirk
Brown, Alan S.
Brown, Clayton H.
Brown, Delisa
Brown, E. Sherwood
Brown, Jesse
Brown, Ritchie
Brown, Walter A.
Bruce, Amanda
Bruchas, Michael R.
Bruinenberg, Vibeke M.
Brunner, Michael
239
315
189
310, 329
248
334
279
103, 233, 249
103, 192
238, 239, 282
233
239
235
191, 263
289
307
242
261
208
221, 302
238
192
255
335
307
258
236
108, 296
255, 256
241
338
254, 317, 324
279
330
282
320
259
277
228, 230
233
331
287
39, 227
293
282
335
269
52, 335
Bruno, Davide
Bruno, John P.
Brutsche, Nancy
Bryan, Angela
Bryant, Camron D.
Bryant, Melanie
Buchanan, Robert
Buchsbaum, Monte
Buckholtz, Neil
Buckley, Peter F.
Buckmaster, Christine L.
Buckner, Randy
Buczynski, Matthew W.
Bueller, Joshua
Bui, Eric
Bujarski, Spencer
Buka, Stephen
Bullmore, Ed
Bulova, Peter D.
Bunney, William
Buonocore, Michael
Burch, Daniel J.
Burdick, Katherine
Burke, Kelly M.
Burmeister, Margit
Burns, Carol
Burstein, Marcy
Bush, Elliot L.
Bushong, Mark
Buss, Claudia
Bustillo, Juan
Butler, Merlin
Butt, Tanya H.
Butters, Meryl A.
Buxbaum, Joseph
Buzhdygan, Tetyana
Bymaster, Frank P.
Cacace, Angela M.
Caceda, Ricardo
Cadenhead, Kristin
Cahn, Wiepke
Cai, Guiqing
Cain, Rachel E.
Calabrese, Joseph
372
226
264
293
313
142, 144, 145,
316
296
228, 229, 318
285
30
226, 230
302
246
336
312
292
254
59, 297
273, 284
246
31, 236, 241,
268
319
292
233, 235, 273,
323
225
226, 288
323, 327
237
313
275
101, 243, 273
194, 230
282
225
233
33, 35, 36, 197,
280, 291
226
296
264
293
102, 274
287
291
223
309, 310, 329
ACNP 50th Annual Meeting • Final Program
Calhoun, Vince
Calkins, Monica E.
Callicott, Joseph H.
Calvó, María
Camchong, Jazmin
Campbell, Joannalee C.
Campbell, Nicholas G.
Campbell, Phillip
Canales, Juan J.
Cañive, Jose M.
Cannistraci, Christopher
Cannon, Tyrone
Cano, Sonio
Cao, Dingcai
Capecchi, Mario
Capitanio, John P.
Caplan, Arthur
Caputo, Angelika
Carey, Amanda N.
Carlezon, William
Carli, Vladimir
Carmody, Thomas
Carneiro, Ana
Caron, Marc
Carpenter, Anne E.
Carpenter, Janet
Carpenter, Linda
Carpenter, William
Carr, Gregory
Carroll, Allison J.
Carroll, F. Ivy
Carroll, Marilyn
Carson, Richard E.
Carson, William H.
Carter, Cameron
Carter, Sue
Caruana, Amanda L.
Carver, Frederick
Casey, Daniel E.
Casper, Regina
Cassidy, Brittany S.
Castellanos, Xavier
Cates, Lindsay
Cazorla, Pilar
Centanni, Sam
195
237
286, 299
238
284, 288
302
280
121, 298
334
274
251
102, 274, 312,
337
332
331
185
301
170, 171, 172
269
321
33, 34, 97, 113,
221
281
331
225, 280
54, 256, 334
268
136, 298
192, 240, 279
188
264
313
221
108, 296
242, 247, 283
294
275, 287, 319
214
258
298
308
234
298
101, 148, 244,
321
332
271
313
Cerullo, Michael
Ceskova, Eva
Chae, Sharon S.
Chai, X.J.
Chait, Brian T.
Chamroonrat, Wichana
Chan, Mico
Chandler, Judson
Chandramohan, Dharshan
Chandrasekaran, R. Y.
Chang, Kiki
Chang, Lee C.
Chang, Linda
Chang, Wei-li
Chansard, Matthieu
Charlton, Rebecca
Charney, Dennis
Chaudhury, Dipesh
Chaudhury, Nashid
Chavkin, Charles
Chelliah, James
Chen, Alon
Chen, Anthony G.
Chen, Changzheng
Chen, David T.
Chen, Guang
Chen, Haiming
Chen, Jingshan
Chen, Kevin
Chen, Lei
Chen, Lian-Yu
Chen, Ligong
Chen, Meng
Chen, Peining
Chen, Ping
Chen, Qiang
Chen, Shufen
Chen, Xiangchuan
Chen, Xin
Chen, Yu-Han
Cherkerzian, Sara
Cheung, Iris
Cheung, Timothy H.C.
Cheung, Yin Bun
Chiavacci, Rosetta
Childress, Anna R.
Childs, Emma
373
320
250
235
249
250
289
240
313
246
316
216, 323, 324
253
101
255
234
272
83, 84, 232
43, 337
140, 286
46, 262
224
333
299
307
236
208
226
264
263
228
314
262
54, 256
239
255
238, 278, 299
279
321
54, 256
274
59, 297
235
303
249
237
290, 313, 324,
325
325
ACNP 50th Annual Meeting • Final Program
Chiles, Deborah
Chisholm, Denise
Chiu, Chi-Tso
Cho, Raymond Y.
Cho, Youngsun T.
Choi, Doo-Sup
Choi, Jennifer H.K.
Choi, Sun
Choi, Yong Kee
Chopra, Amit
Chow, Carolyn
Chow, Tiffany W.
Chowdhury, Nabilah I.
Chowdury, Sohini
Christian, Brad
Christianson, John
Christina, Barr S.
Christoffel, Daniel J.
Christopher, Paul
Chu, King-Wai
Chu, Richard
Chuang, Brandon
Chuang, De-Maw
Ciccocioppo, Roberto
Cissell, Shadha H.
Clair, David St.
Clark, C. Brendan
Clark, Janet A.
Clark, Luke
Clark, Nicholas
Clasen, Liv
Classi, Peter
Claus, Eric
Claycomb, Kierstyn
Clinton, Sarah
Cloninger, C. Robert
Clough, Shannon J.
Coakley, Alice
Cobia, Derin
Coccaro, Emil F.
Cocker, Paul J.
Cohen, Ann D.
Cohen, Bruce
Cohen, Lee
Coit, Caitlin
Colalillo, Sam A.
Colasurdo, Elizabeth A.
292
233
264
259
243
305
269
305
296
307
284
299
239
182
246, 285
338
301
119, 262
235
283
221
335
208, 264
258
241
238
314
225
312
294
244
308
313
291
33, 41, 42, 222
238
267
249
287
241
295
246
261, 268, 286,
307
227, 308
307
123, 273
326
Colbran, Roger J.
Cole, Steve W.
Coleman, Tammi
Colibazzi, Tiziano
Collins, Dan
Comer, Sandra
Compton, Wilson
Conesa, Horacio A.
Congdon, Eliza
Conley, Robert R.
Conn, Jeffrey
Conneely, Karen N.
Connelly, Jessica
Connolly, John
Connolly, Lynn
Connor, Caroline
Connor, Kathryn M.
Conrad, Kelly L.
Conroy, Carla
Conroy, Deirdre
Conroy, Jennie L.
Constant, Brad
Contet, Candice
Conti, Alana C.
Conway, Kevin
Cook, Edwin
Cook, Jason B.
Cook, Jim
Cookson, Mark
Coombs, Garth
Cooper, Alissa
Cooper, Jared
Cooper, Thomas B.
Cooper, Ziva
Coote, Marg
Coplan, Jeremy D.
Coppola, Richard
Corcoran, Cheryl
Cormier, Jim
Cornblatt, Barbara
Cornelius, Jack R.
Cornett, Elyse M.
Cornish, James
Corradi, John P.
Correll, Christoph
Coryell, William
374
303
281
337
284
299
250, 253, 331
314
238
312
228
19, 335
279
214
237
328
235
271
142, 143, 259
310
259
335
306
301
296
314
134, 273, 280
306
255
177
298
286
291
327
253, 331
237
241
298
140, 274, 284,
286
296
102, 274
293
301
313
264
58, 77, 78, 250,
251, 252
277, 329
ACNP 50th Annual Meeting • Final Program
Counotte, Danielle S.
Coupland, Nicholas J.
Coveleskie, Kristen
Covington III, Herbert
Cowan, Christopher W.
Cowell, Rita
Coyle, Joseph T.
Craig, David
Crawford, Cynthia A.
Crawford, Emily L.
Cressman, Victoria
Crews, Fulton
Croarkin, Paul E.
Crocker, Jennifer
Cropsey, Karen L.
Cross, Donna J.
Crow, Tim J.
Crowley, Michael
Crumbley, Alex
Csernansky, John
Cubells, Joseph
Cucchiaro, Josephine
Cullen, Kathryn
Cullis, Jeffrey
Cullum, Munro
Cummings, Jeffrey
Cunningham, Kathryn
Currie, Paul J.
Curtis, Andre L.
Cuthbert, Bruce
Cutter, Gary R.
Czachowski, Cristine L.
D’Aiuto, Leonardo
D’Antonio, Emily
Dabas, Puneet
Dale, Anders
Daly, Mark J.
Daniels, J. Scott
Daniels, Judith K.
Dannenfelser, Ruth
Dardo, Tomasi
Darvasi, Ariel
Daskalakis, Zafiris J.
David, Fleck
David, Sean P.
Davies, Peter
Davis, Adeola R.
336
242
328
43, 337
301
212
264
236
338
280
274
306
322
64, 321
314
283
285
261
274
287, 304
279, 317
229, 250, 251,
252
284, 320
321
234, 285
269, 270
112, 296
256
222
95
270
301
304
291
291
101
280
335
242
35, 280
290
238
274
320
288
279
259
Davis, Caroline
Davis, Elysia P.
Davis, John
Davis, Kenneth L.
Davis, Michael
Daws, Lynette C.
Dawson, Gerard R.
Day, Amanda
Day, Jonathan P.
de Araujo, Ivan
de B. Frederick, Blaise
de Erausquin, Gabriel A.
De Francisco, Don F.
De La Garza, Richard
de Lecea, Luis
De Luca, Vincenzo
de Mathis, Maria A.
de Wit, Harriet
79, 280
243, 273
201, 251, 329
291
97, 228
332
258
221
269
261
288
238
311
253, 254
232
238
276
112, 317, 325,
331, 332
Dean, Reginald
52, 335
Deaver, Daniel
52, 335
Debiec, Jacek
222
DeBold, Joseph F.
302
DeBonis, Dan
311
Decety, Jean
319
Deckersbach, Thilo
233, 310
Deehan, Gerald A.
300, 304
Deep-Soboslay, Amy
318
Deisseroth, Karl
39, 43, 113, 217,
227, 263, 337
del Re, Elisabetta
275
DelBello, Melissa
216, 247, 292,
308, 320
Delgado, Jorge
330
Deligiannidis, Kristina M. 320
DeLisi, Lynn E.
284
Dellovade, Tammy
294
Demeter, Christine
277, 309, 329
Demireva, Elena Y.
223
Deng, Zhi-De
312
Denny, Bryan
283
Denton, Rex
255
DePlonty, Kaitlyn
319
Der-Avakian, Andre
262
DeRosse, Pamela
233, 238, 243,
313
DeSilva, Tara M.
304
375
ACNP 50th Annual Meeting • Final Program
Detke, Michael
Detloff, Allison
Deutsch, Stephen I.
Devanand, D. P.
DeVito, Elise
Devlin, Bernie
Dgetluck, Nancy
Di Martino, Adriana
DiazGranados, Nancy
Dickerson, Tobin J.
Dickstein, Daniel
Diedrich, Andre
Dieter, Lesa
Dietrich, Peter
Dietz, David M.
Dietz, Karen C.
Digavalli, Sivarao
DiLeone, Ralph
Dincheva, Iva
Ding, Yu-Shin
Ding, Zheng-Ming
Diniz, Juliana B.
Dinov, Ivo
Dirks, Bryan
Ditzhazy, Jennifer
Diwadkar, Vaibhav A.
Dobson-Stone, Carol
Dodds, Chris M.
Dodman, Nicholas H.
Dogterom, Peter
Dombrovski, Alexandre
Domino, Edward F.
Domino, Joseph S.
Dong, Hongxin
Doty, Richard L.
Douaihy, Antoine
Dougherty, Darin
Dowd, Erin C.
Downing, AnnCatherine
Downs, Pamela
Doyle, Glenn A.
Doyle, Trevor
Drapeau, Elodie
Drevets, Wayne
Du, Fei
Du, Jing
227, 231, 271,
311
243
228, 230
327
288
280
74, 251
244
293
336
245, 282, 320
222
227
299
226, 262
301
255
112
34, 48, 49, 277
247, 261, 283
300, 304
276
242
229
258
286
278
273
278
334
312
325
325
304
276
293
203, 233
274
239
269
265
335
291
192, 207, 234,
330
286
264
Duan, Naihua
Dubé, Laurette
Dubé, Sanjay
Dubocovich, Margarita L.
Duc, Anne Marie
Ducci, Francesca
Dudley, Kevin J.
Duffy, Amanda M.
Duffy, Anne
Dugar, Ashish
Dukes, George E.
Dulawa, Stephanie C.
Duman, Ronald
Dumitru, Ana Maria
Dumoulin, Michelle
Dunbar, Geoffrey
Duncan, Erica
Dunn, Laura
Durling, Michelle J.
Durston, Sarah
Dwivedi, Saurabh
Dwivedi, Yogesh
Dwork, Andrew J.
Dzirasa, Kafui
Eack, Shaun
Earley, Willie
Easton, Amy
Eaton, William
Ebrat, Bahar
Edden, Richard A.E.
Eddins, Donnie
Edgar, J. Christopher
Edwards, John
Edwards, Scott
Edwards, Steven
Ehlers, Cindy
Ehrman, Ronald
Eiland, Lisa
Einat, Haim
Eisenberg, Daniel P.
Ekhator, Nosakhare N.
Eldik, Linda van
Elizalde, Diana
Elkarim, Johnson Gad
Elkes, Joel
Ellingrod, Vicki L.
Elliott, Mark
Ellis, James
376
308, 311
79, 280
328
267
294
316
302
233
321
229
312
222
186, 207, 263
306
337
72, 229
274
235
247
245
252
225, 240
225
227
291
307
255, 256
318
312, 328
320
334
274
310
222
68, 332
253, 314, 317
290, 324, 325
291
224
318
247
187
255
322
31, 32
228, 235
244, 285
232
ACNP 50th Annual Meeting • Final Program
Ellis, John
Elman, Igor
Elmslie, Gwendolynne
Elsworth, John D.
Ely, Benjamin A.
Ely, Tim
Emeson, Ronald B.
Emslie, Graham
Engel, Anzhelika
Engelhardt, Barbara
Englemann, Alex
Englund, Erika
Enoch, Mary-Anne
Entringer, Sonja
Epel, Elissa
Epperson, C. Neill
Epping, Eric
Epstein, Emerson M.
Ercan-Sencicek, A. Gulhan
Ereshefsky, Larry
Eriksson, Hans
Ermentrout, G. Bard
Ernst, Monique
Erwin, Brigette
Escobar, Javier I.
Eskandar, Emad
Espallergues, Julie
Esscok, Susan M.
Estévez, Marcel
Eudicone, James M.
Evans, Catherine
Evans, Jevelo
Evans, Suzette M.
Everitt, Barry J.
Evins, A. Eden
Ewing, Sarah Feldstein
Fagerness, Jesen
Fair, Damien
Falk, Daniel
Falk, Torsten
Fallon, Brian A.
Fan, Jin
Fan, Theresa
Fan, Xin
Fani, Negar
Fanselow, Michael S.
Farahi, Judah
Fatemi, S. Hossein
254
256, 313
254
328
321
242
296
245, 308
281
317
305
296
244, 281, 317
273
326
209
237, 281
274
280
334
307
259
231, 232, 243
269
238, 330
179, 203
224
252, 330
265
308, 310
325
326
298
138, 266, 303
249, 288
313
236
101, 244
104
296
338
231, 283
265
245
242
312
324
267
Faucett, James
Fava, Maurizio
Fawcett, Jan
Febo, Marcelo
Feder, Adriana
Federici, Lauren
Feeny, Norah
Feifel, David
Feldman, Ruth
Felix-Ortiz, Ada C.
Felix, Anna
Feng, Jian
Fenton, Miriam C.
Ferguson, Deveroux
Ferguson, Margaret
Ferguson, Michael
Ferguson, Susan
293
288, 294, 328
329
245
232
136, 298
277, 329
292, 329
214, 311, 325
245
255
262
314
223
328
246
115, 117, 118,
267
Fernandez-Mendoza, Julio 297
Fernandez-Navarro, Pablo 281
Fernandez, Thomas V.
280
Ferragud, Antonio
334
Ferrell, Robert
236
Ferrer, Marc
296, 335
Feusner, Jamie
287, 322
Fiedorowicz, Jess G.
277, 329
Fields, Julie A.
307
Figueroa-Guzman, Yazmin 302
Fijal, Bonnie
236, 239
Filin, Evgeniy E.
292
Findling, Robert L.
277, 292, 309,
329
Finegood, Eric
61, 234
Fink, Max
327
Finney, Charlene
222
Fisch, Marylynn C.
274
Fischer, Avital M.
279
Fischer, Bernard A.
318
Fischer, Bradford D.
258
Fischer, Jeffrey
322
Fish, Kenneth N.
267
Fisher, Carl
235
Fisher, Melissa
276
Fisher, Phil
244
Fitz, Stephanie
136, 298
Fitzgerald, Jacklynn
232
Fitzgerald, Kate D.
241, 242
Fitzsimons, Linda
327
377
ACNP 50th Annual Meeting • Final Program
Fivel, Peter A.
Flagan, Taru
Flavin, Karen S.
Fleck, David E.
Fleckenstein, Annette E.
Flood, Dorothy
Florenzano, Néstor V.
Flyer, Johanna
Foll, Bernard Le
Folsom, Timothy D.
Foltin, Richard
Fong, Timothy
Fontenot, Miles R.
Forbes, Erika
Forbes, Robert A.
Ford, Judith M.
Forrester, Tammy
Fossaluza, Victor
Foster, Jane A.
Foussias, George
Fowler, Christie
Fowler, J. Corey.
Fowler, Joanna
Fox, Andrew S.
Fox, Meredith A.
Fox, Nathan A.
Fox, Robert G.
Frånberg, Olivia
Francis, Alan
Franco-Chaves, Jose A.
Frangou, Sophia
Frank, Ellen
Frank, Guido
Frank, Marcos G.
Frankel, Erica
Frankle, W. Gordon
Franklin, Teresa
Frantz, Kyle
Fraser, Ian
Frazer, Alan
Frazier, Jean A.
Frazier, Thomas W.
Frederick, Aliya L.
Frederique, Behm M.
Fredrich, Sarah E.
Free, R. Benjamin
Freedman, Robert
257
241, 242
235
247, 292
257
255
238
338
94, 295
267
253, 298, 331
290, 307
301
233, 290
294
259
270
276
237
287
34, 50, 51, 315
257
290, 316
263
263
231, 232
296
254
284, 286
293
329
170, 277
199
337
330
282, 287, 290
290, 324, 325
200
258
31
321
309
303
289
249
335
206, 213, 238,
239, 282
Freeman, Andrew
Freeman, Marlene
Freund, Nadja
Frey, Benicio N.
Fricchione, Samuel
Fried, Ruby
Friedman, Allyson
Friedman, Edward S.
Friedman, Joseph I.
Frielingsdorf, Helena
Frölich, Lutz
Fromm, Stephen J.
Frye, Mark A.
Frye, Stephen
Fuchs, Rita
Fudge, Julie L.
Fujinaga, Yukako
Fung, Daniel
Fung, Samantha
Funk, Adam
Furey, Maura
Gabbay, Vilma
Gabrieli, John
Gabrieli, Susan W.
GadElkarim, Johnson
Gainetdinov, Raul R.
Galdzicka, Marzena
Galendez, Gail
Galfalvy, Hanga C.
Gallagher, Lizbeth
Gallego, Juan
Gallen, Courtney L.
Gallezot, Jean-Dominique
Galli, Aurelio
Gallitano, Amelia
Galloway, Matthew P.
Galvan, Adriana
Galvan, Thania
Gambale, Katherine
Gan, Wenbiao
Ganocy, Stephen J.
Gao, Joseph
Gao, Keming
Gao, Yang
Garcia, Luis F.
Gardiner, Sara
Garg, Pradeep K.
378
329
192, 308
335
227, 237
68, 332
278
43, 337
310
291
48, 277
269
243, 320
307
54, 256
257
243
248
249
181, 240
304
330
321
249, 284
284
287
334
278
129, 271
310
255
252, 291
244
242, 247, 261,
283
98, 109
255
296
200
245, 282
286
185
310
229
310
296
266
292
289
ACNP 50th Annual Meeting • Final Program
Garg, Sudha
Garrett, Amy
Gascoigne, Dennis K.
Gaskill, Christa F.
Gatt, Justine M.
Gau, Susan S.-F.
Gaudreau, Hélène
Gawryl, Maria
Gawuga, Cyrena
Ge, Shaoyu
Geday, Jacob
Geddes, John
Geist, Cheri
Gelenberg, Alan
Gentil, Andre
George, Danielle K.
George, David T.
George, Elizabeth
George, Mark S.
George, Robert
George, Sophie A.
George, Susan
George, Ted
George, Tony
Geracioti, Thomas D.
Gerhardt, Greg A.
Gershon, Elliot
Gershon, Samuel
Gerson, William A.
Geschwind, Daniel
Geyer, Mark
Gfroerer, Joe
Ghahremani, Dara G.
Ghesquiere, Angela
Ghoddoussi, Farhad
Ghorashi, Shahab
Ghose, Subroto
Giardino, Nicholas
Gibbs, Richard
Gibson, Sara A.
Giedd, Jay
Gilbert, Donald L.
Gildengers, Ariel
Gilder, David
289
216, 324
302
225
278
245
79, 280
74, 251, 255
240
217
299
311
326
297
203
281
253
324
249, 323, 327
305
222
265
314
252, 274, 319,
324
247
224
206
201
292
197
125, 221, 237,
239, 265, 266,
278, 328, 337
314
290
311
296
276
119, 262, 318
248
197, 280
337
100, 101, 244,
284
280
233
314
Gilder, David A.
Gill, Kathryn M.
Giller, Earl
Gillespie, Charles F.
Gilman, Jodi
Gilmore, John
Gilpin, Nicholas W.
Gingrich, Jay
Gingrich, Jay A.
Ginns, Edward I.
Ginsberg, Stephen D.
Giraldez, Antonio
Girgenti, Matthew J.
Girgis, Ragy R.
Giuffra, Luis
Giuvelis, Denise
Gizer, Ian R.
Gjedde, Albert
Glahn, David
Glaser, Paul E.A.
Gleason, Kelly
Glenthoj, Birte
Glick, Ira
Glickman, Kim
Glineburg, Paul
Glover, Ebony M.
Godfrey, Jodi
Goeders, Nicholas E.
Goff, Donald
Gogtay, Nitin
Gold, Allison B.
Gold, James M.
Goldberg, Concepcion
Goldberg, Terry
Golden, Sam A.
Goldenberg, Marina
Goldman, David
Goldman, Marina
Goldsmith, Abigail
Goldstein, David
Goldstein, Jill
379
317
337
231, 293
279
282
101
222
210
127, 223, 264,
304
278
226
131, 261
305
287
292
296
317
299
111, 195, 234,
276
224
318
250, 284
251
311
264
242
243
301
129, 218, 230,
271
284
317
228
279
96, 211, 235,
279
119, 262
291
37, 238, 244,
281, 315, 316,
317, 322
290, 324, 325
337
180
57, 59, 60, 282,
297
ACNP 50th Annual Meeting • Final Program
Goldstein, Lawrence
Goldstein, Rita
Gołembiowska, Krystyna
Gomes, Glenn
Gommoll, Carl
Gönenç, Atilla
Gonzalez-Burgos, G.
González-Maeso, Javier
Goodman, Marianne
Goodman, Wayne
Gopal, Srihari
Gordon, Evan
Gorelick, David A.
Gouder, Laura
Gould, Georgianna
Gould, Todd D.
Grabemann, Marco
Grace, Anthony
Graham, Danielle
Grandelis, Anthony
Grandy, David K.
Grandy, Madeline S.
Grannemann, Bruce
Grant, Jon E.
Grant, Paul
Grant, Steven
Grant, Yanabel
Gray, Audrey
Graybiel, Ann
Grayson, David
Greco, John
Greden, John
Greely, Hank
Green, Michael
Greenberg, Ben
Greene, Robert W.
Greenwood, Tiffany A.
Greer, Tracy
Gregg, Christopher
Grenga, Andrea
Grewen, Karen
Griebel, Guy
Griffin III, William C.
Griggs, Erica
Grillon, Christian
Grimes, Erin M.
83, 88, 89
179, 289, 316
296
319
307
246
212
255
283, 316
231
229
258
228
291
332
261
240
104, 337
294
287
336
336
309, 327
292
270
179
305
294
217
244
232
103
101
194, 228, 234,
238, 239, 276,
282
179
285
238, 282
309, 327
209
254, 317
326
309
256, 302
305
241
241
Grissom, Nicola M.
Groark, James
Gross, Robin
Grosz, Daniel E.
Grove, Tyler B.
Gründer, Gerhard
Gruner, Patricia
Gu, Hong
Guan, Cuntai
Guercio, Leonardo A.
Guerin, Glenn F.
Guerreri, Stephanie
Guerrero, Gonzalo
Guevara, Maria
Guha, Saurav
Gunawardane, Nisali
Gundapuneedi, Tejasvi
Gunduz-Bruce, Handan
Guo, Junjie U.
Guo, Xiaodong
Gur, Raquel
Gur, Ruben
Gurevich, Eugenia
Gustafsdottir, Sigrun
Guthrie, Sally K.
Gutierrez-Reid, Navarre
Gutman, David
Guyer, Amanda E.
Haase, Brennan
Habeck, Christian
Haber, Suzanne
Haddad, Stephen
Hadlock, Gregory C.
Haggarty, Stephen J.
Hahn, Chang-Gyu
Hahn, Maureen K.
Haile, Colin N.
Hajek, Tomas
Hakonarson, Hakon
Halaris, Angelos
Halberstadt, Adam L.
Hallmayer, Joachim
Hamani, Clement
Hamer, Robert M.
380
305
292
274
311
228
289
243, 323
244
249
269
301
283
238
289
238
235
275
259
225
325
99, 101, 111,
188, 237, 238,
239, 244, 275,
276, 282, 285
100, 237, 239,
244, 275, 285
209
268
325
303
242
232, 243
322
312
179
279, 288
257
268
99
222
254
321
237, 244
293
265
237
303
252, 330
ACNP 50th Annual Meeting • Final Program
Hamill, Terence
Han, Ming-Hu
Han, Sungho
Han, Yang
Handen, Ben
Haney, Margaret
Haney, Meg
Hanlon, Colleen A.
Hanna, Gregory L.
Hannaway, Kayleen E.
Hanson, Glen
Haramati, Sharon
Hardy, Nicholas
Hargreaves, Richard
Harlow, Bernard L.
Haroon, Ebrahim
Haroutunian, Vahram
Harris, James
Harris, Margret S.H.
Hart, Amy
Hart, Kim
Hartley, Sigan L.
Hartwell, Karen
Harvey, Philip
Haselgrove, Christian
Hasenkamp, Wendy
Hashimoto, Eri
Hashimoto, Kenji
Hashimoto, Takanori
Hasso, Anton
Hatch, John
Hauser, Sheketha R.
Havekes, Robbert
Hawkins, Rollin
Hayden, Benjamin Y.
Haynes, M. Ryan
Haynes, Virginia
Haynes, William G.
Hazan, Rebecca
Hazlett, Erin A.
Hazlett, Kathleen
Hazlett, Kathleen E.
Head, Kevin
Heath, Brianna
Heaton, Leanne
Heaton, Robert
334
33, 43, 44, 45,
155, 156, 157,
158, 337
327
335
246, 270
253, 331
313
249
241
334
257
333
265
334
227
321
96, 239, 319
214
285
317
283
246
249
251, 291, 329
321
274
306
205
319
243
293
300, 304
269
254
299
123, 273
228
277
325
283
64, 322
321
243
304
237
291
Hedlund, Peter B.
Hedou, Gael
Hegadoren, Kathleen M.
Heilbronner, Sarah
Heilig, Markus
Heim, Christine M.
Heiman, Gary A.
Heimberg, Richard
Heinloth, Alexandra
Heinssen, Robert
Heisserer, Nicolle
Heitzeg, Mary M.
Hellemann, Gerhard
Hellmich, Martin
Heman, Karen
Hempstead, Barbara
Hen, Rene
Hendler, Talma
Hendricson, Adam
Henn, Fritz A.
Henry, Brook
Henry, Mellissa M.
Henry, Shannan
Herbst, Nicole
Herkenham, Miles
Herman, Alex
Hermannsson, L.
Herring, W. Joseph
Hessler, Martha J.
Hewlett, William
Higley, J. Dee
Higley, James D.
Hill, Matthew
Hill, Michele
Hill, Scot
Hill, Shelley J.
Hillard, Cecilia J.
Hilt, Dana
Himes, Michael L.
Himle, Joseph A.
Hinds, Oliver
Hines, Samantha R.
Hinton, David
Hipolito, Maria
Ho, Beng-Choon
Ho, Jennifer
381
332
294
242
312
37, 253, 258,
314, 315
243
280
269
236
274
329
288
239
318
256
48, 277
221, 225
311
255
262
328
311
242
227
248
276
237
271, 272
235
225
315
301
291, 325
129, 271
275
237
291
58, 74, 75, 76,
251, 255
282, 290
242
249
255, 266
305
294
237, 285
284
ACNP 50th Annual Meeting • Final Program
Ho, S. Shaun
Ho, Y.-Y.
Hobart, Mary
Hobus, Joy
Hochadel, Thomas J.
Hochfeld, Marla
Hodes, Georgia E.
Hodge, Rachel
Hodge, Steven
Hodgkinson, Colin
Hoefler, Michael
Hoekstra, Pieter J.
Hoener, Marius C.
Hoff, David
Hoffman, Beth
Hoffman, Elana
Hoffman, Ellen J.
Hoffmann, Robert
Hoftman, Gil D.
Hoks, Roxanne M.
Holcomb, Henry H.
Hole, Anita
Hollander, Eric
Hollander, Jonathan
Holly, Elizabeth N.
Holm, Margo B.
Holmes, Andrew
Holroyd, Tom
Holsen, Laura
Holt, Daphne J.
Holtzheimer, Paul
Hommer, Daniel
Hong, Elliot
Hoonakker, Amanda
Horan, William
Horga, Guillermo
Horiguchi, Masakuni
Hornig, Mady
Horton, Rebecca
Hosanagar, Avinash
Hoschl, Cyril
Hosford, David
Houde, John
Hough, David
61, 234
277
294
293
308
230
115, 119, 120,
262
265
321
238, 244, 316,
322
248
280
334
283
103
330
116, 131, 132,
133, 261
259
267
247
318
290
307
98
302
233
204
298
59, 282, 297
298
192, 235
253, 282, 314,
332
228, 286
257
188, 234
284
334
59, 297
332
231
321
57, 72, 73, 229
276
229
Hough, Morgan E.
Houle, Sylvain
285
288, 319, 322,
324
Hounie, Ana G.
276
Houseknecht, Karen L.
336
Houslay, Miles D.
269
Houston, John
236, 256
Howe, Meghan
323, 324
Howell, Brittany R.
243, 244
Hoyer, Juergen
248
Hsiao, Elaine
183
Hsu, David
57, 64, 65, 321
Hsu, Jay
250, 252
Hu, Jian
242
Hu, Jiun-Yiing
248
Hu, Lisa T.
331
Hu, Xiaoping
243, 244, 321
Huang, Chih-Chia
294
Huang, Hao
245, 275
Huang, Kuo-Hao
294
Huang, Mei
255
Huang, Mingxiong
274
Huang, Wen
278
Huang, Xi-ping
54, 256
Huang, Xu-Feng
240
Huang, Yiyun
242
Hubbard, Catherine S.
312
Hubbard, Sydney
274
Huber, Kimberly M.
301
Hudson, James I.
307, 314
Hudson, Randall L.
267
Huebinger, Ryan
327
Huestis, Marilyn A.
228
Huganir, Richard
110
Hughes, Jonathan C.
226
Hughs, Zoe A.
263
Huitron-Resendiz, Salvador 332
Hultman, Christina
213
Hulvershorn, Leslie
245
Hummer, Tom
245
Hunter, Michael A.
274
Husain, Mustafa
234, 327
Hutchinson, Anthony J.
267
Hutchison, Kent
195, 313
Hutchison, Natalie
275
Hutzelmann, Jill
271, 272
Hwang, Kai
273
382
ACNP 50th Annual Meeting • Final Program
Hyde, Thomas
Hyman, Steven
Iacoviello, Brian
Iadarola, Michael J.
Ialongo, Nicholas
Ianni, Angela
Iarikova, Polina
Ibrahim, Hisham M.
Ibrahim, Lobna
Ikuta, Toshikazu
Im, Heh-In
Ingram, Wendy
Iñiguez, Sergio D.
Insel, Thomas
Inslicht, Sabra
Irwin, Michael R.
Ismail-Beigi, Faramarz
Ismail, Zahinoor
Isohannii, Matti
Iwuagwu, Christiana
Iyengar, Smriti
Izenwasser, Sari
Izumi, Takeshi
Jabbar, Gul A.
Jabbi, Mbemba
Jackowski, Andrea
Jackson, Michael
Jackson, Saheeda
Jacobs, Leslie
Jacobsen, Paula
Jacoby, Ryan J.
Jaeger, Judith
Jagannathan, Kanchana
Jahn, Allison L.
Jahshan, Carol
Jaime, Karelia Montane
Jakovcevski, Mira
Jalbrzikowski, Maria
Jamadar, Sharna
James, Anthony C.
James, Dennis
James, Eliassen
Janes, Amy C.
Jang, Mi-Hyeon
Janowski, Scott
191, 268, 279,
305, 318
182
232
296
318
298, 318
238
285
293
243, 323
305
255
333
30, 101, 182,
186, 197
297
281
310
299
316
255
236
298
338
284
298
241
208
271
322
269
292
230, 259
290, 325
247
276
253
235
284
155, 161, 162,
288
285
324
320
288
225
255
Janowsky, David
Jansen, Wim
Jarcho, Johanna M.
Jardemark, Kent
Jarvelin, Marjo-Riitta
Javitch, Jonathan A.
Javitt, Daniel
Jayaraman, Srini
Jayne, Millard
Jenjahn, Elsa
Jenkins, Aaron
Jenkins, Sarah
Jensen, J. Eric
Jensen, Niels
Jentsch, David
Jentsch, J. David
Jepson, Christopher
Jernigan, Courtney
Jernigan, Turner N.
Jessen, Tammy
Jeste, Dilip
Jhee, Stan
Jia, Jiemin
Jiang, Lihong
Jiang, Zhiguo
Jiantonio, Rachel
Jimenez, Dennisse
Jimerson, David C.
Jin, Jian
John, Catherine
John, Davis
Johnson, Anna
Johnson, Cynthia
Johnson, Kevin
Johnson, Philip
Johnson, Sheri L.
Johnson, Sterling C.
Jokinen, Jussi
Jones, Edward G.
Jones, Jeffrey L.
Jones, Jermaine D.
Jones, Kelli
Jope, Richard S.
Joseph, Jane E.
Joseph, Lisa
383
201
231
231
256
316
259, 335
99, 194, 205,
247
334
290
248
305
233
307
54, 256
170, 173, 174
337
317
301
251
225
267
334
264
240
328
288
265
292
34, 54, 55, 56,
256
261
252
325
270
249
116, 136, 137,
298
237
246
247
241
274
250, 331
255, 256
224, 295
290
270
ACNP 50th Annual Meeting • Final Program
Josselyn, Sheena
Jovanovic, Tanja
Juarez, Barbara
Judd, Lewis L.
Juelich, Richard
June, Harry L.
Jurjus, George J.
Kaakinen, Marika
Kaalund, Sanne S.
Kaestner, Klaus
Kahler, Christopher W.
Kahn, René S.
Kalali, Amir
Kalechstein, Ari D.
Kaleshian, Vasken
Kalin, Ned H.
Kalivas, Peter
Kamali, Masoud
Kamath, Jayesh
Kamath, Vidyulata
Kamel, Christy
Kamiya, Atsushi
Kanayama, Gen
Kanba, Shigenobu
Kane, John
Kang, Nayoung
Kaplan, Allan S.
Kaplitt, Michael
Kaplow, Julie
Kapur, Shitij
Kara, Nirit
Karayiorgou, Maria
Karlsgodt, Katherine
Karlsson, Rose-Marie
Karmacharya, Rakesh
Karolewicz, Beata
Karry, Rachel
Karsson, Craig
Kasahara, Dr. Takaoki
Kash, Thomas
Kassem, Layla
Kaszas, Krisztian
Katic, Alain
Katz, Martin M.
Kaufman, Marc J.
Kaye, Walter
97
204, 242
43, 337
329
284
249
227
316
318
315
253
287
229
253
226
263
190, 300
297
309
276
255
163, 168, 169,
268
314
248
77, 230, 231,
250, 252
305
280
83, 86, 87
326
230, 284
224
104
284, 312
265
268
301
267
231, 309
208
294, 336
236
296
308
201
288, 321
199, 282
Kayser, Reilly
Keator, David B.
Keedy, Sarah K.
Keefe, Richard
Kegeles, Lawrence
Kei, Justin
Keilp, John G.
Kelleher, Dennis L.
Keller, Jason M.
Keller, Jennifer
Keller, William R.
Kelley, Ryan
Kellner, Charles
Kelly, Deanna L.
Kelly, Megan M.
Kelly, Michelle P.
Kelly, Timothy
Kelsoe, John
Kelsoe, John R.
Kemp, David E.
Kenna, George
Kenna, Heather A.
Kennedy, David N.
Kennedy, James
Kennedy, Pamela J.
Kennedy, Patrick
Kennedy, Richard E.
Kenny, Paul
Keolasy, Daniel
Keshavan, Matcheri
Keshaviah, Aparna
Keslerwest, Marilyn
Kessler, Daniel
Kessler, Robert
Ketter, Terence A.
Keyes, Katherine M.
Khan, Arif
Khan, Asif
Khan, Shirin R.F.
Khine, Tin
Kiefer, Orion
Kilduff, Thomas S.
Kilpatrick, Lisa A.
Kilts, Jason D.
Kim, Airee
384
320
252
285
229, 275, 309
194, 287
243
338
312
296
323
318
323, 324
327
228
331
263
249
236
237
310
68, 332
326
321
79, 238, 239,
280, 287
226
103
270
50, 199, 305,
315
266
102, 275, 284,
286, 291
292
252
232
98
237, 310
314
293, 308
272
293
285
242
334
328
295
305
ACNP 50th Annual Meeting • Final Program
Kim, Ju-Young
Kim, Junghyun
Kim, Pilyoung
Kim, Suck Won
Kim, Sun-Hong
Kim, Sung-yon
Kim, Tae
Kimmons, Steven
Kimura, Mitsuru
King, Andrea C.
King, Anthony
King, Dalton
King, Margaret K.
King, Robert A.
Kingsbery, Joseph P.
Kinkead, Becky
Kinon, Bruce
Kinsey, Berma
Kippenhan, J. Shane
Kippenhan, Shane
Kippin, Tod E.
Kircher, Daniel
Kirkpatrick, Matthew G.
Kirsch, Courtney
Kis, Bernhard
Kish, Stephen
Kitabatake, Yasuji
Klahr, Kristin
Klar, Rebecca
Klassen, Bryan T.
Kleber, Herbert D.
Kleijn, Huub Jan
Klein, Donald
Klein, Sam
Kleinman, Joel
Klibanski, Anne
Klimes-Dougan, Bonnie
Kluge, Wolfgang
Klumpp, Heide
Klunk, William E.
Knackstedt, Lori A.
Knapp, Rebecca
Knesevich, Mary Ann
Knopp, Michael
Knox, Dayan
Ko, Eunice
Kobak, Kenneth A.
225
254, 317
233
292
268
263
39, 227
293
315
331
232, 248
255
295
280
328
241
228, 230, 239
296
283, 298
246
258, 302, 304
265
332
229
240
288, 324
225
284
222
307
201
334
31, 201
226
177, 206, 268,
279, 305, 318
59, 282, 297
320
119, 262
231
246
257
327
229
52, 335
222
282
227
Kobets, Andrew J.
Koch, Alisa
Kocsis, James
Koek, Wouter
Koenig, Barbara
Koenig, Gerhard
Koenigsberg, Harold W.
Koeppe, Robert
Koethe, Dagmar
Kohler, Christian
Kohn, Philip
Kohut, Stephen J.
Kokel, David
Kolachana, Bhaskar
Komek, Kubra
Konechnik, Thomas
König, Gerhard
Koob, George
Kopecek, Miloslav
Kopelowicz, Alexander
Korade, Zeljka
Koran, Lorrin M.
Korycinski, Steven
Koslow, Stephen
Kosten, Therese A.
Kosten, Thomas
Kowalska, Magdalena
Kozel, F. Andrew
Kozlenkov, Alexey
Kozman, Maggie
Krach, Sören
Kraemer, Markus
Krakowski, George
Kranzler, Henry
Kreek, Mary Jeanne
Kressig, Reto W.
Kring, Ann
Krishnan, K. Ranga
Kroenke, Candyce
Kronstein, Philip
Kroon, Rene
Krueger, Jessica J.
385
240
121, 298
48, 277, 310
332
101
74, 251
283, 316
64, 321, 325
318
285
246, 278, 283,
298, 318
257
263
238, 278, 299,
318
259
270
255
94, 222, 258,
301, 303, 304,
305
321
230
337
307
64, 321
95
278
108, 289, 296
296
322
255
324
289
240
338
104
250
269
188
249
326
293
271
222
ACNP 50th Annual Meeting • Final Program
Krystal, John
70, 192, 242,
259, 261, 283,
288, 291
Kubicki, Marek
284, 285
Kuczenski, Ronald
278
Kufahl, Peter R.
336
Kuhn, Cynthia
221, 295
Kumar, Anand
272, 320, 322
Kunii, Yasuto
268
Kunisato, Aihiko
324
Kunkel, Louis
183
Kuo, Yan
35, 280
Kupfer, David
95, 114, 277,
328
Kurbanov, Daniel B.
256
Kurian, Benji
327
Kurup, Pradeep
244
Kuskowski, Michael
284
Kutiyanawalla, Ammar
333
Kuwabara, Hiroto
289
La Charite, Jaime
317, 324
La, Christian
287
Labus, Jennifer S.
312, 328
Laezza, Fernanda
226
Lahti, Adrienne
268
Laje, Gonzalo
218
Lalonde, Francois
244
Lam, Shing Chun
290, 324
Lamar, Melissa
272
Lamb, Sarah N.
243
Lamberti, Steve
129, 271
Lammertsma, Adriaan A. 287
Landeros-Weisenberge, A. 271
Landis, Story
30
Lane, Hsien-Yuan
294
Lane, Richard
322
Lane, Scott D.
289, 295
Langenecker, Scott
64, 231, 297,
321, 322
Langleben, Daniel D.
313, 324
Lanius, Ruth A.
242
Lansing, Kathryn
326
LaPaglia, Melissa
256
LaPlant, Quincey
262
Larr, Allison S.
247
Larson, Erin
303
Laruelle, Marc
273, 287
Lasser, Robert
229, 309
Lasseter, Heather
Latham, Patricia
Lattemann, Dianne
Laubmeier, Kimberly K.
Lauriello, John
Lavin, Antonieta
Lavretsky, Helen
Law, Amanda J.
Lawrence, Philip
Lawson, Anthony
Lawson, Elizabeth
Le Foll, Bernard
Le-Niculescu, Helen
Le, David
LeBlanc, Nicole
Lebowitz, Barry
Lebron-Milad, Kelimer
Leckman, James
Ledley, Debra
Lee, Bridgin
Lee, Chi-Ming
Lee, Francis
Lee, JongAh
Lee, Junghee
Lee, Kendall H.
Lee, Mary R.
Lee, Royce
Lee, Tiffany T.Y.
Lee, Tih Shih
Lee, Tong H.
Lee, Vicent
Leggio, Lorenzo
Lehmann, Michael L.
Lehner, Thomas
Lehrer, Douglas
Leibenluft, Ellen
Leitman, David I.
LeMatty, Todd
Lemos, Julia
Lencer, Rebekka
Lencz, Todd
Lenoci, Maryann
Lenox, Robert
Leon, Andrew C.
386
257
272
98
308, 310
230
259, 337
281
286, 305
273
228
282
303
278
301
292, 311
308, 311
297
70, 214, 271,
276, 280, 291,
325
269
315
119, 262
48, 113, 277
327
228, 234
307
244
332
291
249
257
328
57, 68, 69, 332
148, 151, 152,
153, 248
180, 197
285
216, 231, 233,
320
275
249
46, 262
234
238, 291, 313
297
189
310
ACNP 50th Annual Meeting • Final Program
Leotti, Lauren
Leow, Alex
Lepack, Ashley
Lepping, Rebecca
Leranth, Csaba
Lerch, Jason
Lerman, Caryn
Leslie, Frances
Levenson, Jessica C.
Levent, Kirisci
Leventhal, Adam M.
Leventhal, Bennett L.
Levin, Thomas M.
Levine, Sarah
Levitan, Robert
Levitt, Pat
Leweke, F. Markus
Lewine, Jeffrey
Lewis, David
Li, Bo
Li, Chao
Li, Chia
Li, Xiang
Li, Xiaohua
Li, Xingbao
Li, Yin
Li, Yu-Wen
Li, Zheng
Liberzon, Israel
Liddie, Shervin
Lidge, Regina
Lieberman, Jeffrey
Liebowitz, Michael
Light, Gregory A.
Lii, Theresa R.
Lijffijt, Marijn
Lim, Choon Guan
Lim, Kelvin
Lim, Keunpoong
Lin, Jue
Lin, Keng-Han
Lin, Shu-Fei
Lindell, Stephen G.
221
287, 322
263
282
328
287
315, 317
200
277
293
253
261
317
255
58, 79, 80, 280
180, 210
318
319
191, 212, 267,
319
262
268, 318
336
302
19, 262
249
275, 290, 324,
325
255, 256
264
222, 232, 248
298
255
72, 140, 170,
229, 239, 252,
286, 330
269
239, 243
296
295
249
284, 288, 290,
320
247, 283
326
269
247
315
Lindenmayer, Jean-Pierre
Linder, Jonathan
Lindler, Kathryn
Lindquist, Martin
Lindsley, Craig W.
Lines, Christopher
Links, Kira A.
Linton, Edward
Lipska, Barbara
Lipska, Barbara K.
Lisanby, Sarah
Lish, James
Lissek, Shmuel
Lister, Jamey J.
Litvin, Yoav
Litz, Brett
Liu, Chunyu
Liu, Fang
Liu, Juan
Liu, Kenneth
Liu, Peiying
Liu, Sherry
Liu, Shuang
Liu, Xiaoxu
Liu, Xiu
Liu, Xun
Liu, Yanni
Livi, Carolina
Lobach, Iryna
Lobaugh, Nancy
Lobo, Mary K.
Lodge, Nicholas
Loebel, Antony
Loewy, Rachel L.
Logan, Jean
Lohoff, Falk
Lohoff, Falk W.
Lokuge, Sonali
Loman, Michelle
Lonce, Suzanna
London, Edythe
Lonergan, Colleen
Lopera-vasquez, Juan
Lopez-Castroman, Jorge
387
229
277
225
242
259, 335
272
299
269
177
268, 318
19, 26, 27, 234,
235, 271, 312,
326, 327
255
241
242
325
337
206, 238
333
249
272
322
307
256
321
301
283
241
279
272
287
43, 148, 154,
226, 333, 337
255
229, 250, 251,
252
326
290
325
265
237
325
222
248, 290, 312
303
330
281
ACNP 50th Annual Meeting • Final Program
Lopez-Jaramillo, Carlos
Lopez-Larson, Melissa
Lopez, Alberto J.
Lopez, Marcelo F.
Lopresti, Brian L.
Lori, Adriana
Loring, Erin
Lotfipour, Shahrdad
Loth, Annemarie
Lotrich, Francis
LoTurco, Joseph J.
Lotz, Meredith J.
Loughead, James
Louis, Lindsay St.
Love, Tiffany
Lovenberg, Timothy
Lu, Bai
Lu, Hanzhang
Lu, Qun
Luber, Bruce
Lublin, Henrik
Luckenbaugh, David A.
Lueken, Ulrike
Luks, Tracy
Luna, Beatriz
Lunagariya, Abhishek
Lund, Jesper
Lutgen, Victoria
Lyketsos, Constantine
Lyons, David M.
Ma, Hong-Tao
Ma, Junshiu
Ma, Liangsuo
Ma, Ting Martin
Ma’ayan, Avi
Maayan, Lawrence
Macciardi, Fabio
MacDonald, Angus
MacDonald, Kai
Maciag, Dorota
Macklin, Eric A.
Macor, John
Macritchie, Karine
Madayag, Aric
Madden, Katherine L
Maddock, Richard
Madhoo, Manisha
330
246, 313
294
306
246
317
283
254, 315
245
236
305
277
237, 244, 275,
285
296
64, 321, 322
258
265
322
50, 315
312
284
293
248
276
273
293
273
299
270
302
296
272
289
266
35, 280
247
236, 268
288
292, 329
226
129, 271
255, 256
311
299
268
319
309
Madras, Bertha K.
306
Magida, Jane
119, 262
Magnan, Renee
313
Mahableshwarkar, Atul R. 269
Mahajan, Gouri
227
Maher, Brady J.
305
Mahley, Robert
211
Mahon, Katie
323
Mahoney, Erin
271
Mahoney, James J.
253
Maier, Steven
338
Maio, Roberto Di
304
Makris, Nikos
282
Malaga, Maria J.
242
Malapani, Cara
221
Malaspina, Dolores
229
Maldonado-Devincci, A. M. 306
Malenka, Robert
110, 186
Malhotra, Anil
218, 233, 235,
238, 243, 252,
273, 291, 313,
323
Malik, Mansoor
294
Malinow, Roberto
196
Malison, Robert
247
Mallya, Karyn S.
317
Malmerfelt, Anna
256
Maloney, Thomas
316
Mamdani, Firoza
236, 268
Mamo, David C.
299
Mandel, Francine S.
230
Mandelkern, Mark
254, 290, 324
Mandyam, Chitra
305
Manji, Husseini
186, 207, 293
Mann, J. John
225, 310, 323,
327
Manschreck, Theo
230
Mantovani, Antonio
271
Mantsch, John R.
302
Manubay, Jeanne M.
250, 331
Mao, Wenje
235
Mao, Xiangling
241
Mapes, Kimberly S.
322
March, John
103
Marcinick, Heather
329
Marcinkiewcz, Catherine A. 294
Marcus, Monica M.
254, 256
Marcus, Sheila
61, 234
388
ACNP 50th Annual Meeting • Final Program
Marder, Stephen
Marek, Kubicki
Marenco, Stefano
Marinic, Tina
Mark, Opler
Markou, Athina
Markowitz, John C.
Marmar, Charles
Marona-Lewica, Danuta
Marquez, Kathleen
Marsh, H. Michael
Marsh, Patrick
Marsit, Carmen
Martin, David A.
Martin, Laura
Martinez, James
Martinez, Karen G.
Martinez, Luis A.
Martínez, Maribel
Martinez, Vicente
Martinowich, Keri
Martins, Silvia S.
Maruff, Paul
Marugan, Juan
Marvin, Robert
Marx, Christine E.
Masdeu, Joseph C.
Mash, Deborah
Mason, Graeme F.
Mason, N. Scott
Masood, Anbrin
Masten, Virginia L.
Mathalon, Daniel
Mathé, Aleksander A.
Mathew, Sanjay J.
Mathis, Chester A.
Mathis, Kristopher I.
Matho, Andrea
Matos, Jannifer
Matsubara, Takurou
Matsumoto, Joseph Y.
Matsushima, Toshiya
Mattay, Venkata
Matthews, Brittany A.
Matthews, Stephen
Matthies, Heinrich J.G.
Mattick, John S.
188, 228, 229
283
246
48, 277
229
262
242
102, 178, 297
265
290
296
322
279
265
282
236
163, 166, 167
223
238
312
248, 265
314
273
258, 296
285
295
278, 318
317
240
290
333
222
102, 259, 326
251
241
246, 282
276
291
298
319
307
338
96, 278, 286,
299
288
79, 280
259
302
Matuskey, David
Matza, Louis
Mauro, Christine
May, Warren
Mayberg, Helen
Mayer, Cynthia
Mayer, Emeran A.
Mayes, Linda
Mayford, Mark
Mayson, Sarah Jo
McAuley-Clark, Erica
McBride, William J.
McCall, Jordan G.
McCall, Nora M.
McCarley, Robert
247
308
311
227
83, 85, 235, 243
283
312, 328
261
267
283
278
300, 304
335
294
33, 39, 40, 227,
275
McClintock, Shawn M.
234
McClung, Colleen
114, 227, 303
McCluskey, Tina
324
McCormick, Jennifer
101
McCracken, James T.
317
McCullumsmith, Cheryl B. 314
McCullumsmith, Robert
319
McDaniel, Jonathan
275
McDougle, Christopher
183, 270
McElligott, Zoe A.
223
McElroy, Susan L.
307, 310
McEntire, Caleb
328
McEwen, Bruce
291, 325
McFadden, Lisa M.
257
McGaughy, Jill A.
223
McGeary, John
68, 317, 332
McGinty, Jacqueline F.
302
McGlade, Erin
246, 313
McGlashan, Thomas
274
McGrath, Patrick J
330
McInnis, Melvin
226, 297, 310
McKay, Cameron
305
McKeague, Ian W.
277
McKenna, James
39, 227
McKinney, Rebecca
329
McLaughlin, Jay P.
321
McMahon, Francis
236, 330
McMahon, Lori L.
261
McMahon, Robert P.
228
Mcmanus, Owen B.
296
McNally, James
39, 227
McNamara, Nora K.
309
389
ACNP 50th Annual Meeting • Final Program
McOmish, Caitlin E.
McPhie, Donna
McQuade, Robert D.
Meador-Woodruff, James
Meaney, Michael
Mears, Ryan
Meda, Shashwath
Meerlo, Peter
Megan, Gunnar
Mehra, Vishaal
Mehta, Pankaj D.
Mello, Nancy K.
Meltzer, Herbert
Menard, François
Mendelowitz, Alan J.
Meng, Xiangyi
Mennes, Maarten
Mentch, Frank D.
Mercado, Ramon
Mercer, Kristina B.
Merchenthaler, Istvan
Meresh, Edwin
Merikangas, Kathleen
Mervis, Carolyn
Merzenich, Michael
Messer, Michael
Mette, Christian
Metzler, Thomas
Meyer-Lindenberg, A.
Meyer, Jeffrey H.
Meyer, Paul J.
Meyer, Roger
Meyers, Kortni
Meyers, Oren
Miao, Feng
Michael, Thase E.
Michelson, David
Michopoulos, Vasiliki
Mickey, Brian J.
Miczek, Klaus A.
Mignot, Emmanuel
Miguel-Hidalgo, Jose J.
Miguel, Euripedes C.
Mihalakos, Perry
304
268
294, 308, 310
212, 239, 314,
319
79, 280
276
288
114, 269
325
293
226
257
74, 104, 239,
251, 255, 332,
334
230
230
230, 231
282, 283
237
322
279, 281
261
293
213, 237, 245
246
214, 215
292
240
297
198, 318
288, 322
261
104
64, 231, 321
329
293
293
96, 182, 271,
272
243
322
302
213
226, 227
276
275
Mikhil, Bamne
Miklowitz, David
Milad, Mohammed
Milham, Michael P.
Millen, Brian A.
Miller, Andrew
Miller, Bailey W.
Miller, Courtney
Miller, Del D.
Miller, Erin M.
Miller, Gregory
Milton, Denái
Minassian, Arpi
Mines, Marjelo A.
Ming, Guo-li
Minoshima, Satoshi
Mintz, Jim
Minzenberg, Michael
Miotto, Karen
Miranda, Dielle
Mirrione, Martine M.
Mirzabekov, Julie
Mizrahi, Romina
Mletzko, Tanja
Moberg, Kerstin Uvnäs
Moberg, Paul J.
Moeller, F. Gerard
Moeller, James R.
Moeller, Michael
Moeller, Scott J.
Mogali, Shanthi
Moghaddam, Bita
Mohanty, Suman
Mohr, Pavel
Moline, Jessica
Molino, Bruce F.
Molinuevo, José L.
Molski, Thaddeus
Momenam, Reza
Monk, Chris S.
Montalvo-Ortiz, Janitza L.
Monteggia, Lisa
Monteiro, Isabel
Monterosso, John R.
Montez, David
Moody, Teena
Moonat, Sachin
Moore, Constance M.
390
304
324
242, 297
244, 282, 321
239
186, 321
258
305
230, 277
224
306
308
237, 328
295
225
283
230, 329
275, 287, 319
254
299
262
263
319, 324
243
247
276
289, 295, 296
338
254
289
331
19, 22, 23, 190
134, 273
250
235
256
269
255
253, 282
241
304
196
272
253
273
322
306
320
ACNP 50th Annual Meeting • Final Program
Moore, Holly
Moore, Loretta
Morairty, Stephen R.
Morales, Amelia N.
Moran, Lauren V.
Mordi, Isioam
Moreau, Jean-Luc
Moreira, Jose C. F.
Morell, Christopher
Moreno, Alexis
Morgan, Paul
Morris, Colleen
Morris, David
Morrison, John
Morrison, Ryan D.
Morrissey, Judy L.
Morrow, A. Leslie
Morrow, Eric M.
Morrow, John
Mory, Roland
Mosconi, Matthew
Moss, Stephen
Motlagh, Maria
Mount, Daniel J.
Moy, Sheryl S.
Moya, Pablo R.
Mueller, Bryon A.
Mueller, Daniel J.
Mueller, Martina
Mueller, Ulf
Muftuler, L. Tugan
Muhrer, Eli J.
Mukherjee, Jogeshwar
Mukhin, Alexey G.
Mulholland, Patrick J.
Muller, Jeff M.
Mulsant, Benoit
Multani, Pushpinder K.
Munafò, Marcus
Munsie, Leanne M.
Murali, Dhanabalan
Murphy, Dennis L.
Murphy, Eleanor
Murphy, James
Murphy, Keely M.
Murphy, Niall P.
109, 140, 274,
286
240
334
316
286
238
334
227
313
247
249
246
291
207
335
311
306
245
334
334
116, 134, 135,
273
191
271
239
294
278
288
239
327
57, 66, 67, 249
243
320
252, 285
289, 324
256, 302
223
233, 287, 299
265
253
239
246
263, 278
330
317
304
315
Murray, Jennifer E.
Murray, Robin
Murrough, James
Muse, John
Muzik, Maria
Myers, Amanda J.
Myers, Richard M.
Myerson, Gayle
Myint, Aye-Mu
Nadar, Michael
Nader, Karim
Nader, Michael
Nagarajan, Srikantan
Nahas, Ziad
Nair, Govind
Nakazawa, Kazu
Naliboff, Bruce D.
Nam, Hyung Wook
Nanda, Steven A.
Napier, T. Celeste
Narayan, Ankita
Narayan, Sapna
Narayana, Ponnada A.
Narendran, Raj
Narendran, Rajesh
Nash, Tiffany
Nash, William
Nasr, Shahin
Nasrallah, Henry
Nathan, Pradeep J.
Nazih, Rachid
Neale, Ben M.
Negus, S. Stevens
Neisewander, Janet L.
Nelson, Brent G.
Nelson, Eric E.
Nelson, Erik B.
Nelson, J. Craig
Nelson, Marianela
Nemeroff, Charles
Nemirovsky, Natali E.
Nenov, Miroslav
Neria, Mariana J.
Neria, Yuval
Nerumalla, Chandra
Ness, Roberta
391
303
198
232, 242
278
61, 234
278, 281
241
291
186
107
97
258
276
323, 327
243, 244
212
312
305
263
295
225
224
289
109
282, 290
298
337
298
229, 230
273
289
280
333
303
290
232
247
310
223
102, 198, 241,
243, 278, 281
336
226
242
242
254
90
ACNP 50th Annual Meeting • Final Program
Nestler, Eric
Neufeld, Richard W.J.
Neumaier, John
Neumann, Inga D.
Neumeister, Alexander
New, Antonia S.
Newburn, Erin N.
Newcomer, John W.
Newell, Kelly
Newman, Amy
Newman, Lori A.
Newton, Samuel S.
Newton, Thomas F.
Neylan, Thomas
Nguyen, Dana
Ni, Lisong
Nicholls, Brianne
Nichols, Charles D.
Nichols, David E.
Nichols, Lisa
Nicholson, Shevon E.
Nicol, Ginger E.
Nicoletti, Mark
Niculescu, Alexander
Nie, Xingju
Nielsen, David A.
Nielsen, Mette
Niendam, Tara
Nierenberg, Andrew A.
Nierenberg, Jay J.
Nievergelt, Caroline
Nikisch, Georg
Nimgaonkar, Vishwajit
Nin, Mauricio S.
Ninan, Philip
Nisenbaum, Laura K.
Nixon, Ralph
Nizami, Maryam
Niznikiewicz, Margaret
Nobler, Mitchell S.
Nobrega, José N.
Nocjar, Christine
Nomura, Jun
Nopoulos, Peggy
30, 43, 83, 91,
110, 182, 192,
217, 223, 226,
262, 333, 337
242
117, 204, 267,
303
338
102, 232, 242
283, 316
318
235
240
107
223
226
254
102, 297
252
64, 321, 325
237
265
265
278
221
235
293
278
290
278
284
275
233, 310
226
337
251
304
223
192
239
187
225
275
327
303
333
266
281
Norcross, Maxine A.
Norcross, Roger D.
Nordström, Peter
Norman, Sonya B.
Norrholm, Seth D.
Novak, Gabriela
Novak, Tomas
Novakovic, Vladan
Nuamah, Isaac
Nuechterlein, Keith H.
Nunes, Edward
Nurmi, Erika L.
Nyberg, Fred
Nye, Jeffrey
Nyhuis, Allen
O’Brien, Charles
O’Brien, Elizabeth G.J.
O’Connor, Sean J.
O’Donnell, James M.
O’Donnell, Patricio
O’Malley, Stephanie
O’Neal, Jon
O’Neill, Barry V.
Obral, Sarah
Ochsner, Kevin N.
Odlaug, Brian L.
Ofer, Pasternak
Ogasa, Masaaki
Ogawa, Lisa
Ohmura, Yu
Ohtsu, Hiroshi
Okada, Go
Okamoto, Yasumasa
Oler, Jonathan A.
Olincy, Ann
Olino, Thomas M.
Olive, M. Foster
Olker, Christopher
Olsavsky, Aviva K.
Olson, Amanda
Olson, Richard
Olson, Valerie G.
Olvera, Rene
Omar, Yasmine
Ongur, Dost
392
241
334
247
241
242
265
321
291
229
238, 239, 282
331
317
272
103
228
94, 290, 313,
324, 325
268
325
333
181, 200, 265,
336
104, 142, 331
232
273
310
283
292
283
250
306
338
261
324
324
263
238, 239, 282
233
336
316
320
274
255, 256
303
279
254
246, 261, 268,
286
ACNP 50th Annual Meeting • Final Program
Onoda, Keiichi
Ooms, Renata
Oosting, Ronald S.
Oquendo, Maria A.
Oranje, Bob
Ordeberg, Gunnar
Orr, Scott
Orson, Frank
Osher, Yamima
Oskooilar, Nader
Osorio, Ricardo
Oswald, Lynn M.
Otto, Michael W.
Overholser, James C.
Owens, Michael J.
Oxenkrug, Gregory
Ozburn, Angela
Padilla, Eduardo
Padmanabhan, Aarthi
Padro, Bobbi S.
Pagulayan, Kathleen
Pahlisch, Franziska
Palfreyman, Michael
Pallanti, Stefano
Palmer, Abraham
Pandey, Ghanshyam N.
Pandey, Subhash C.
Pandya, Anand
Pandya, Chirayu
Panek, Laura
Panessiti, Micaella G.
Pani, Ariel
Paniagua, Beatriz
Panksepp, Jaak
Panos, John
Paolone, Giovanna
Papachristou, Efstathios
Papaleo, Francesco
Pardo, Jose V.
Parekh, Samir
Park, Alan J.
Parker, Clarissa C.
Parsey, Ramin V.
Parsons, Loren H.
Parva, Muhammad
Pasko, Bryce
Passarotti, Alessandra M.
Patel, Sachin
324
319
262
281, 310, 327
250
272
102, 292, 297
296
328
311
272
289
227
227
241
293
303
238
246, 273
301
283, 326
318
74, 251
307
316, 317
225, 240
306
103
333
247
263
246
140, 286
333
255
261
329
264
284
321
269
316
323
297, 336
316
299
232
336
Patel, Sagar
Patel, Shitalben
Patnaik, Samarjit
Patt, Virgine
Patterson, Beth
Patterson, Thomas
Paul, Steven
Paula, Pelavin
Pauls, David
Paulus, Martin P.
Pava, Matthew
Pavuluri, Mani N.
Payer, Doris
Payne, Nancy
Pearce, Brad
Pearlson, Godfrey
Pearson, Jay
Pearson, John M.
Pearson, Rahel
Pecenak, Jan
Peckham, Andrew D.
Pela, Marlena
Pentkowski, Nathan S.
Perera, Tarique
Perez-Edgar, Koraly
Perez-Rodriguez, M.
Perkins, Diana
Perlis, Roy H.
Perlmutter, Joel
Perry, William
Person, Ann L.
Peruzzo, Denis
Pescosolido, Matthew F.
Peskind, Elaine R.
Peterchev, Angel V.
Peters, Amy T.
Peters, Bart D.
Peters, Gary L.
Peters, Jamie
Peterson, Bradley
Peterson, Randall
Peterson, Sean
Petit, Isabelle
Petrides, Georgios
Petrie, Eric C.
Petryshen, Tracey
Pfaff, Donald
393
278
235
258
328
276
291
211
283
276
241, 242
259
232
324
327
274
195, 275, 288,
291
271
299
326
250
233
239
303
327
232
281, 310, 316
102, 274, 326
236, 268
281
237, 328
288
323
245, 282
283, 326
326
233
243
273
190
284
263
54, 256
267
327
283
275
325
ACNP 50th Annual Meeting • Final Program
Pfefferbaum, Adolf
Pham, Steve T.C.
Phan, K. Luan
Phillips, Katharine A.
Phillips, Mary
Phillips, Paul
Pi, Bo
Pich, Emilio Merlo
Pickel, James
Pierpaoli, Carlo
Pieschl, Rick
Pikalov, Andrei
Piletz, John
Pilhatsch, Maximilian
Pillai, Anilkumar
Pimentel, Jeff
Pine, Daniel
Pine, Daniel S.
Pineles, Suzanne
Pinna, Graziano
Piomelli, Daniele
Pitman, Roger
Pittenger, Christopher
Pittman, Brian
Pizzagalli, Diego A.
Platt, Brian J.
Platt, Donna M.
Platt, Michael
Pleil, Kristen
Pletnikov, Mikhail
Ploense, Kyle L.
Ploubidis, George
Pocivavsek, Ana
Pogorelov, Vladimir M.
Poh, Ernest
Poldrack, Russell A.
Poline, Jean-Baptiste
Pollack, Mark
Pollock, Bruce
Pollock, Jonathan
Polt, Robin
289
266
61, 231, 234,
241
331
114, 216, 290,
328
33, 46, 47, 117,
138, 262, 266,
267, 312
247
107
264
246
255
229, 250, 251,
252
293
248
226, 333
294
100, 232, 233
231, 232, 241,
243, 320
297
223
318
97, 279, 297
70, 240, 261,
291
240
236
225
258
299, 312
336
266, 268
258, 302
329
264
268
310
312
195
292
287, 299
185
296
Pomara, Nunzio
Pope Jr., Harrison G.
Pope, Harrison G.
Popp, Danielle
Porges, Eric C.
Porges, Stephen
Porreca, Frank
Porrino, Linda
Portella, André
Portland, Kimberly B.
Post-Munson, Debra
Potenza, Marc
Potkin, Steven G.
Potter, William
Potts, Bryan
Powell, Susan B.
Powers, Robyn
Prater, Katherine
Preda, Adrian
Price, Julie C.
Price, Lawrence
Prickaerts, Jos
Prinz, Susanne
Prisciandaro, James
Prokai, Laszlo
Propper, Lukas
Prossin, Alan
Prudic, Joan
Pruessner, Jens C.
Psaros, Christina
Ptáček, Louis J.
Purcell, Shaun
Pushparaj, Abhiram
Qian, Yongxian
Qin, Liya
Qualmann, Krista
Quevedo, Karina
Qui, Haijun
Rabin, Rachel
Rabinak, Christine
Rabinowitz, Jonathan
Radant, Allen D.
Rademacher, Lena
Radua, Joaquim
Raeke, Lisa
Ragland, J. Daniel
394
226
314
307
227, 231, 271,
311
319
214
189
258
79, 280
270, 308
255
288, 307
252
19, 103, 182
318
266, 303
273
231
252
246
240, 279
255
289
233, 249
261
321
115, 121, 122,
298, 326
327
243
308
91, 92, 93
206
303
328
306
299
325
237
252, 274
231
230
238, 239, 282
289
243
129, 271
275
ACNP 50th Annual Meeting • Final Program
Rahman, Zia
Raimondi, Giorgio
Rainnie, Donald
Rajbhandari, Abha K.
Rajji, Tarek
Rajkowska, Grazyna
Ramachandra, Vorani
Ramchandani, Vijay
Ramirez-Restrepo, Manuel
Ramsey, Lissandra C.
Ramsey, Stacy
Rana, Zaker
Randall, Pat
Rane, Pallavi
Rangel, Antonio
Rangeon, Beatriz Molina
Rankin, Katherine
Rao, Jagadeesh S.
Rao, Uma
Rapaport, Mark H.
Rapoport, Judith
Rapoport, Stanley I.
Rascovsky, Simon
Rasenick, Mark
Rasetti, Roberta
Rasgon, Natalie L.
Raskin, Joel
Raskind, Murray A.
Rasmussen, Keith
Rasmussen, Kurt
Rauch, Scott
Rauch, Sheila
Ray, Lara
Ray, Neepa
Ray, Riju
Ray, Russell
Razdan, Varun
Raznahan, Armin
Rebecca, Sripada
Redila, Van A.
Redmon, Sarah N.
Redmond Jr., D. Eugene
Reed, Brian
Reed, Carol
Reed, Malcolm
Reed, Stephanie Collins
Reese, Edmund A.
Reichel, Carmela M.
308, 310
304
113, 191
221
287, 299
226, 227
256
253, 314, 332
281
261
326
278
272
321
179
238
335
226
275, 322
90
31
226
330
103
286
326
328
283, 303
327
263
155, 233, 298
232, 248
254
252, 330
315
210
233, 320
244
248
303
222
328
250
310
331
298
226
290
Reidy, Brooke L.
Reilly-Harrington, Noreen
Reilly, James
Reiman, Eric
Reinares, Maria
Reines, Scott
Reisberg, Barry
Reiss, Allan
Reissner, Kate J.
Reite, Martin
Remington, Gary
Ren, Xinguo
Renshaw, Perry
Resa, Joshua
Resnik, Jack
Ressler, Kerry
Rethorst, Chad
Reti, Irving M.
Retzbach, Edward
Revel, Florent G.
Reyes, Beverly
Reyes, Teresa M.
Reynolds, Charles
Reynolds, David S.
Richard, Jan
Richards, Anne
Richardson, Jarrett W.
Richardson, Patricia
Ring, Robert
Rio, Dan
Ripoll, Luis
Risbrough, Victoria
Risch, Neil
Rissling, Anthony J.
Rissman, Emilie
Rivera, Hilda N.
Rizavi, Hooriyah S.
Roach, Brian J.
Robbins, Trevor
Robert, Duwors
Roberts, Amanda J.
Roberts, Laura
Roberts, Rosalinda
Robertson, Chelsea
395
245, 282
310
234, 275, 285
211
329
271
272
323, 324
142, 146, 147,
257, 300
299
230, 287
240
247, 286, 307
335
255
113, 178, 242,
279, 281
327
327
327
334
46, 262
305
233, 308, 311,
312
258
237
297
307
61, 234
180
253
316
102, 221, 237,
337
213
239
209
274
240
259
215
284
301, 332
101
268
290
ACNP 50th Annual Meeting • Final Program
Robertson, David H.
Robertson, Jonanthan
Robicsek, Odil
Robinson, Delbert
Robinson, Donald
Robinson, Terry E.
Roche, Joy
Rodd, Zachary A.
Rodebaugh, Thomas
Rodriguez, Eric E.
Roe, Katheine
Roffman, Joshua L.
Rogers, Joan C.
Rogers, Joshua T.
Rogers, Robert D.
Rohleder, Cathrin
Rohlfing, Torsten
Rojas, Don C.
Rokosik, Sandra
Role, Lorna
Rollins, Brandi
Rollins, Nancy
Romine, Ann
Roof, Rebecca A.
Rook, Jerri M.
Rosa, Moacyr A.
Rosario, Maria C.
Rose, Jed
Roseboom, Patrick H.
Rosell, Daniel R.
Rosen, Brooke
Rosen, Cherise
Rosen, Julie
Rosenbloom, Margaret J.
Rosenblum, Katherine
Rosenthal, Norman
Rosenzweig-Lipson, S.
Rosoklija, Gorazd B.
Ross, Christopher
Ross, Philip
Ross, Stephen
Ross, Thomas J.
Ross, Zafonte
Rosse, Richard B.
Rosse, Stephanie M.
Rostrup, Egill
222
273
267
230, 252
270, 310
261
268
300, 304
269
290
246
115, 129, 130,
271, 279
233
295
295
318
289
286, 299
295
217
236, 268
245
284
335
335
326
276
289, 318
263
283
233
285
275
289
61, 234
307
231
225
226
307, 310
331
244, 286, 313
283
228, 230
228, 230
284
Roth, Bryan
Roth, Robert
Roth, Thomas
Rothbaum, Barbara
Rothschild, Anthony J.
Rothwell, Alice
Rotrosen, John
Roussos, Panos
Roux, Perrine
Rowe, Brian H.
Rowland, Laura M.
Rowles, Brieana M.
Rowlett, James K.
Rowny, Stefan B.
Roy, Alec
Roy, Kamalika
Roybal, Donna J.
Rubenstein, Liza M.
Ruber, Gregory
Rubin, Eric
Rubinow, David
Rubinow, Marisa J.
Rubinstein, Dani
Rudolph, Richard L.
Rumbaugh, Gavin
Rummans, Teresa
Ruoff, Leslie
Ruparel, Kosha
Rush, John
Rusjan, Pablo
Rusjan, Pablo M.
Russo, Scott J.
Rutenberg, Julia G.
Ruth, Nora
Sabb, Fred W.
Sabbag, Samir
Sabo, Aniko
Saborido, Tommy P.
Saccone, Phillip A.
Sacher, Julia
Sachs, Gary S.
Sackeim, Harold
Sackeim, Harold A.
Sacramento, Arianne D.
Safadi, Ziad
Safren, Steven
396
54, 104, 117,
256, 267
309, 328
271, 272
232
269, 320
240
331
316
331
242
318
309
258
271
281
272
323
278
295
298, 331
83, 192
227
298
293
163, 164, 165,
224, 305
327
297
244, 285
95
319, 324
288, 322
119, 262
320
262
312
291
280
303
331
322
311
323
327
258, 304
294
308
ACNP 50th Annual Meeting • Final Program
Saggu, Shalini
Sahakian, Barbara
Sailor, Kurt A.
Saint Marie, Richard L.
Saito, Atsushi
Saito, Toshikazu
Sakharkar, Amul J.
Saladin, Michael E.
Saleh, Sam A.
Salloum, Ihsan M.
Salman, Nava
Salmeron, Betty Jo
Salter, Michael
Salzman, Carl
Sambunaris, Angelo
Sampath, Hemaltha
Sampson, Shirlene
Samudra, Preeti G.
Samulski, Richard J.
Sanacora, Gerard
337
19, 28, 29, 215
225
255
268
306
306
249
259
293
267
244, 313
99
201
309
286
307
288
328
70, 240, 263,
291
Sanchez, Mar
243, 244
Sanchez, Raymond
294
Sanders, Erica Marie
323
Sanders, Jeff D.
267
Sanders, Stephan J.
280
Sandman, Curt A.
243, 273
Sanford, Benjamin J.
322
Sanghavi, Kunal M.
286
Sano, Mary
187
Santesso, Diane L.
236
Sarchiapone, Marco
281
Sargent, Bruce J.
256
Sarma, Kaushik
229
Sarter, Martin
261
Sassano, Maria
54, 256
Satterthwaite, Theodore D. 244
Saunders, Erika F.H.
297
Savadjiev, Peter
285
Savage, Cary
282
Savitz, Adam
229
Sawa, Akira
99, 104, 266,
268
Sawaf, Nadeem
296
Saxena, Kirti
245
Saxena, Sanjaya
270
Scahill, Laurence
270
Schaefer, Catherine
277
Schafer, Scott
Schank, Jesse
Schatzberg, Alan
Schellenberg, Gerard D.
Scherer, Stephen
Schettler, Pamela J.
Schipper, Jacques
Schlagenhauf, Florian
Schlaggar, Bradley
Schloesser, Robert
Schmidt, Matthew
Schmidt, Peter J.
Schmitt, Lauren
Schneider, Lon S.
Schneiderman, Inna
Schneier, Franklin
Schobel, Scott A.
Schoch, Hannah
Schoemaker, Joep
Schoenbaum, Geoffrey
Schoepp, Darryle
Schofield, Peter
Schooler, Nina
Schork, Nicholas
Schrama, Regina
Schreiber, Stuart L.
Schreiner, Matthew
Schroeder, Mathew
Schuebel, Kornel
Schuh, Kory
Schulteis, Gerhard
Schulz, Kurt
Schulz, S. Charles
Schulze, Thomas
Schumann, Gunter
Schwandt, Melanie
Schwarcz, Robert
Schwartz, Barbara L.
Schweiger, Julia A.
Seager, Matthew A.
Sebat, Jonathan
Sedó, Manuel
See, Ronald E.
Segreti, Anthony
397
242, 322
37, 315
31, 95, 241, 302,
323
280
197
329
231, 271, 334
248
281
248, 265
323, 327
283
134, 273
270
325
269
116, 140, 141,
274, 286
269
231
190
205, 271, 272,
334
278
170, 175, 176,
228, 229, 230
238, 278, 282
245
268
284
287
37, 315
228
303
231
284
218
100, 316
253, 258, 314
264
228, 230
235
256
180, 206
238
290
72, 229
ACNP 50th Annual Meeting • Final Program
Seibt, Julie
Seidman, Larry
Selby, Peter
Self, David
Seltzer, Marsha M.
Selvakuma, Balakrishnan
Sen, Namita
Sen, Srijan
Senashova, Olga
Sengupta, Elizabeth J.
Sequeira, Pedro A.
Serrano, Verenea
Sesack, Susan
Setola, Vincent
Seubert, Janina
Severe, Joanne B.
Sevy, Serge
Sewalia, Kaveish
Shad, Mujeeb
Shafe, Samuel
Shaffer, Scott A.
Shah, Nikunj
Shah, Ravi S.
Shaham, Yavin
Shahid, Mohammed
Shamji, Alykhan
Shampine, Lawrence J.
Shan, Dan
Shanahan, Nancy A.
Shapiro, Danielle
Shatti, Shireen
Shavitt, Roseli G.
Shavkunov, Alexander
Shear, M. Katherine
Shearman, Mark
Sheehan, David V.
Sheehan, John
Sheffler, Douglas J.
Shekhar, Anantha
Shekhtman, Tatyana
Shelton, Richard
Shelton, Steve E.
Shen, Joan H.Q.
Shen, Ling
Shen, Pei-Hong
337
102, 238, 239,
274, 282, 284,
291
324
303
246
268
259
103
326
319
236, 268
329
338
54, 256
285
230
273
336
275
253
320
252
253
112, 190, 265
256, 271
268
295
319
222
326
286
276
226
308, 311
294
269
310
259
103, 136, 278,
298, 304
236
308, 310, 337
263
231
277
322
Shen, Xiaoyun
Shenton, Martha
Sheridan, Steven D
Sherman, Scott
Shi, Yundi
Shields, Angela D.
Shih, Jean C.
Shimamoto, Akiko
Shin, Joshua
Shin, Lisa M.
Shinday, Nina M.
Shinn, Ann K.
Shirasaka, Tomohiro
Shirey-Rice, Jana K.
Shoaff, Jessica
Shoaib, Mohammed
Shofer, Jane B.
Short, Baron E.
Short, E. Baron
Shorter, Daryl
Shorter, Edward
Shreshtha, Sunil
Shuler, Catherine
Shulman, Melanie B.
Shumay, Elena
Shumsky, Jed S.
Shungu, Dikoma C.
Sibley, David R.
Siburian, Richie
Sidor, Michelle M.
Siegel, Steven
Siegle, Greg
Siever, Larry J.
Sikka, Sharad
Sikoglu, Elif M.
Silberman, Yuval
Silva, Robert
Silveira, Patrícia
Silver-Ritter, Amy
Silverman, Bernard
Silverman, Daniel
Silverman, Jeremy M.
Silverstein, Noah J.
Silverstein, Steven
Sim, Minyoung
Simeon, Daphne
Simeon, Laika R.
398
296
283, 284, 285
268
296
244
259
263
302
325
279
258
246
306
222
68, 332
266
326
327
323
254
201
285
270
272
316
221
241
335
279
227
194, 212
312
238, 239, 282,
283, 316
282
320
259
229, 250, 252
79, 280
292
104
326
239
129, 271
194
251
307
225
ACNP 50th Annual Meeting • Final Program
Simmons, Alan N.
Simmons, Alex
Simmons, Rebecca
Simon, Naomi
Simonelli, Doreen
Simpson, Danielle
Simpson, H. Blair
Simpson, Tracy
Simpson, William
Sinacore, James
Singer, Zachary
Singh, Jaskaran
Singh, Manpreet
Sinha, Rajita
Siu, Cynthia
Skaar, Todd
Skol, Andrew
Skolnick, Phil
Skritskaya, Natalia
Skuban, Aleksandar
Sladek, John R.
Slaymaker, Valerie
Sleiman, P.M.
Slifstein, Mark
Sloan, Richard
Small, Scott A.
Smith, Alicia K.
Smith, Ashley
Smith, Edward E.
Smith, Erin
Smith, Gwenn
Smith, Hilary
Smith, Jeffrey
Smith, Karen
Smith, Katie M.
Smith, Laura N.
Smith, Matthew S.
Smith, Michelle R.
Smith, Robert C.
Smith, Yoland
Smoller, Jordan W.
Snaidero, Rachel
Snavely, Duane
Snyder, Ellen
Snyder, Solomon H.
Soares, Claudio
Soares, Giovana Zunta
241, 242
245
41, 222
292, 308, 311
250
41, 222
221
94
276
293
290, 325
229
323, 324
317
229, 230, 251
136, 298
317
107
311
294
328
288
237
287
59, 297
140, 274, 286
279
282
221
232
19
258
46, 262
261
247
301
247
239
252
19, 24, 25
279, 288
237
271
271, 272
266, 268
79, 227, 237,
280
293
Soares, Jair
Sobieraj, Jeff
Sodhi, Monsheel
Sodhi, Simrit
Sokoloff, Greta
Solomon, Marjorie
Song, Hongjun
Song, Wenjie
Song, Youeun
Sonntag, Kai C.
Soosman, Steffan
Soreca, Isabella
Sorenson, Andrea
Sorisio, Denise
Sotnikova, Tatyana D.
Southall, Noel
Sowell, Sharon
Sparks, JonDavid
Specker, Sheila
Speer, Andrew
Speer, Taylor
Spencer, Kevin M.
Spencer, Sade
Spicer, Ken
Spivey, James R.
Sponheim, Scott R.
Spreckelmeyer, Katja N.
Sprock, Joyce
Sripada, Chandra S.
Srivastava, Shefali
Stahl, Edward
Stahl, Stephen
Stahl, Stephen M.
Stains, Jean
Stamm, Thomas
Stangl, Bethany L.
Stanley, Jeffrey A.
Stansbrey, Robert J.
Stanwood, Gregg D.
Starosciak, Amy K.
State, Matthew
Staub, Brittany
Stauffer, Shaun R.
Stauffer, Virginia
Stead, Squire M.
Steffener, Jason
399
245, 275, 293
305
239
277
317
275
225
310
280
335
287
277
301
236
334
296, 335
331
328
288
330
331
276
227
323
321
290
289
239
232
237
254
309
231
312, 328
248
332
286
309
303
298
131, 180, 197,
261, 280, 283
234
335
228, 230
307
312
ACNP 50th Annual Meeting • Final Program
Stein, Elliot A.
Stein, Mark
Stein, Murray
Steinberg, Joel L.
Steiner, Meir
Steinfeld, Sara
Steizhammer, Viktoria
Stephen, Lindell
Stephens, Matthew
Stepien, Iwona
Stern, Emily R.
Stern, Yaakov
Stevens, Beth
Stevens, Hanna E.
Stevens, Michael
Stevens, Mike C.
Stevenson, Glenn
Stewart, Jonathan W.
Stewart, Michelle
Stewart, Scott
Stigler, Kimberly A.
Stindl, Julia
Stockmeier, Craig A.
Stoeckel, Luke E.
Stoppel, Cynthia J.
Storr, Carla L.
Stout, J.N.
Strakowski, Stephen
Straub, Richard
Straubhaar, Juerg
Strauss, John
Strauss, Olaf
Strecker, Robert
Strecker, Robert E.
Strohmaier, Christine
Stroup, T. Scott
Stuart, Barbara K.
Stuber, Garret
Stutz, Sonja J.
Styner, Martin
Sublette, M. Elizabeth
Subramaniam, Karuna
Sudheimer, Keith
Sugar, Catherine A.
Suh, Jesse J.
Suh, Yousin
Sullivan, Edith V.
244, 286, 313,
325
271
102, 241, 242
289, 295
79, 237, 280
274
119, 262
301
317
267
241, 242
312
185
268
288
232
296
330
229
272
270
338
226, 227
249
307
314
286
247, 320
318
235
238
338
39
227
269
252, 330
326
260
296
140, 244, 286
323, 327
276
323
239, 324
290, 324, 325
96
289
Sullivan, Gregory
Sullivan, Maria A.
Sullivan, Regina
Sullivan, Sarah G.
Sulser, Fridolin
Sumerel, Brittany
Summergrad, Paul
Sumner, Jennifer
Sun, Ping
Sun, Wei-Lun
Sunday, Suzanne
Sunderajan, Prabha
Suomi, Stephen
Suomi, Stephen J.
Sur, Cyrille
Suridjan, Ivonne
Sussman, Steve
Sutcliffe, James S.
Sutherland, Matthew T.
Suyenobu, Brandall Y.
Svensson, Torgny H.
Svetnik, Vladimir
Swain, James
Swann, Alan C.
Swanson, James
Swedo, Susan
Sweeney, John
Sweet, Robert A.
Swerdlow, Neal R.
Swift, Robert
Swift, Robert M.
Sylvain, Bouix
Sylvia, Louisa G.
Sypek, Elizabeth
Szabo, Steven T.
Szanto, Katalin
Szegedi, Armin
Szelinger, Szabolcs
Szeszko, Philip
Szucs-Reed, Regina
Szuhany, Kristin L.
Szumlinski, Karen K.
Ta, Karen
Tabas, Linda
400
242, 310, 323
331
222
242
201
290
293
270
253
302
230
309, 331
198
301
334
319
253
280
313
328
254, 256
272
57, 61, 62, 63,
234
295
101
183, 270
100, 134, 234,
273, 275, 285,
291
267
238, 239, 243,
255, 266, 282
68, 332
254, 317
283
310
321
257
312
231, 271, 309
236
243, 323
324
241
258, 302, 304
317, 324
270
ACNP 50th Annual Meeting • Final Program
Taffe, Michael A.
Taheri, Saeid
Takahashi, Nagahide
Talbot, Konrad
Talbot, Peter
Taliaz, Dekel
Talledo, Jo A.
Tallman, Katie R.
Tamashiro, Kellie
Tamm, Leanne
Tamminga, Carol
Tan, Hao Yang
Tan, Seong-Seng
Tandon, Neeraj
Taniguchi, Makoto
Taniguchi, Yu
Tantawy, Mohammed N.
Tanum, Lars
Tao, Ran
Tapocik, Jenica
Tarazi, Frank
Tarobochia, Matthew
Tateno, Masaru
Tatrai-Prokai, Katalin
Tavares, Tonya
Taylor, Charles T.
Taylor, Dawn
Taylor, Stephan F.
Teale, Peter
Tedford, Stephanie
Teegarden, Sarah L.
Tekin, Sibel
Telang, Frank
Tempelman, Linda
Teng, Stephanie
Terrace, Herbert
Terry, Alvin
Teslovic, Theresa
Thaker, Gunvant
Thanam, Susrutha
Thapa-Chhetry, Binod
Thase, Michael
Thayer, Julian
Theresa, Lafavor
Theriault, Kraig
336
290
291
99
287
333
243
336
327
245
110, 119, 192,
262, 275, 285,
291, 318, 322
299
181
286
301
268
335
272
318
33, 37, 38, 315
271, 296
283
306
261
68, 332
242
324
228, 241, 242
299
295
224
269
290
254
249
234
333
48, 277
275, 286, 291
243
323
192, 234, 294,
307, 310
322
325
268
Thermenos, Heidi W.
Thoma, Robert J.
Thompson, Britta
Thompson, Judy L.
Thompson, Paul
Thompson, Tyler L.
Thomson, Fiona
Thorp, Steven R.
Thorsell, Annika
Thurmond, Linda M.
Tian, Hui
Tian, Qingjun
Tidey, Jennifer W.
Tighe, Alex
Tillisch, Kirsten
Timpano, Kiara C.
Tischfield, Jay A.
Titus, Steven A.
Tiwari, Arun K.
Todtenkopf, Mark
Toga, Arthur
Tohen, Mauricio
Toki, Shigeru
Tolliver, Bryan
Tolliver, Teresa J.
Tomasi, Dardo
Tompa, Tamas
Toney, Glenn M.
Tong, Junchao
Tong, My-Linh
Tootell, Roger B.H.
Torossian, Carol
Tost, Heike
Toups, Marisa S.P.
Tourian, Karen A.
Tourino, Clara M.
Toy, William A.
Tracey, Irene
Trantham-Davidson, H.
Travis, Michael J.
Tregellas, Jason R.
Treistman, Steven N.
Trestman, Robert
Tristan, Amanda
Trivedi, Madhukar
Truitt, William A.
Truong, Amy
401
284
319
335
274, 287
322
292
334
241
258
247
294
264
253
249
312, 328
278
280
296
239
34, 52, 53, 335
242
310, 329
324
233
263
246, 289
259
332
324
311
298
272
286
327
292
232
303
189
313
290
286
301
309
298
307, 309, 322,
327
300, 304
264
ACNP 50th Annual Meeting • Final Program
Trzepacz, Paula
Tsai, Guochuan Emil
Tsai, Hsing-Chen
Tsareva, Alina
Tseng, Kuei
Tseng, Wen-Yih I.
Tsuang, Debby W.
Tsuang, Ming
Tsutsui-Kimura, Iku
Tucker, Adrienne
Tuinstra, Dan
Tuma, Ivan
Tumuluru, Rameshwari
Turck, Chris W.
Turecki, Gustavo
Turek, Fred W.
Turetsky, Bruce
Turner, Cortney A.
Turner, Jessica
Turner, Jill R.
270
294
43, 337
297
109
245
238, 239, 282
239, 274
338
312
129, 271
250
246
334
177
316
275, 276, 285
241
319
155, 159, 160,
315
Tye, Kay
227, 263
Tyrka, Audrey
240, 279
Ueckermann, Jennifer
240
Uhl, George R.
318
Uhlhaas, Peter
181
Ukai, Wataru
306
Underwood, Mark D.
225
Urban, Nina B.
287
Uslaner, Jason
334
Vaccarino, Flora M.
268
Vaidya, Punit
327
Vaidya, Vidita
224
Vaillancourt, David
134, 273
Vakkalanka, Radhakrishna 286, 318
Valentino, Rita
209
Valentino, Rita J.
222
Vallender, Eric
306
Van Ameringen, Michael 276
van Berckel, Bart N.
287
Van Bockstaele, Elisabeth 46, 262
Van den Burg, Erwin H.
338
van der Doef, Thalia
287
van Engeland, Herman
245
van Enkhuizen, Jordy
125, 266
van Goethem, Nick
255
van Haren, Neeltje E.
287
Van Meter, Anna
277, 329
van Zessen, Ruud
Vanderschuren, Louk
Vaquero-Lorenzo, C.
Vardigan, Josh
Varney, Mark
Varney, Seth A.
Vasudev, Akshya
Vasudev, Kamini
Vatsalya, Vatsalya
Vawter, Marquis P.
Vederman, Aaron
Veenstra-VanderWeele, J.
Veer, Ashlee Van’t
Veijolai, Juha
Velasquez, Natalie D.
Velazquez-Sanchez, Clara
Velez, Lady P.
Vellios, Evan
Verbiest, RyAnna
Vergne, Derick
Vermetten, Eric
Vernaleken, Ingo
Vgontzas, Alexandros
Vialou, Vincent
Vialou, Vincent F.
Vieira-Brock, Paula L.
Vieta, Eduard
Villafuerte, Sandra
Vinje, Morten
Vinogradov, Sophia
Vitaterna, Martha H.
Vitek, Michael
Vitela, Melissa
Vitiello, Ben
Vogt, Brent
Voineskos, Aristotle
Volk, David W.
Volkow, Nora
Volkow, Nora D.
Voronin, Konstantin
Vosburg, Suzanne
Vranjkovic, Oliver
Wadhwa, Pathik D.
Wager, Tor
Wager, Travis T.
Wahl, Troy A.
402
260
336
281
334
266
225
311
311
332
236, 268
322
19, 20, 21
221, 261
316
290
334
222
324
259
272
178
289
297
43, 148, 149,
150, 223, 262,
333
337
257
329
288
272
276, 326, 335
316
187
332
270
203
287, 299
319
30, 246, 290
289, 316
272
250, 253, 331
302
273
242, 322
316
336
ACNP 50th Annual Meeting • Final Program
Wakai, Sara
Walker, Christopher
Walker, Elaine
Walker, Lindsay
Walker, Sara
Wallace, Nathan M.
Wallace, Tanya
Walley, Annie
Wallis, Jon
Walsh, Jessica
Walsh, Paul L.
Walsh, Tim
Walters, Oakland C.
Walther, Donna
Waltz, James
Wanat, Matthew
Wand, Gary S.
Wang, Gene-Jack
Wang, Heng
Wang, Jennifer
Wang, Kuan H.
Wang, Lei
Wang, Liqin
Wang, Nulang
Wang, Pengwei
Wang, Po W
Wang, Si-Wei
Wang, Yang
Wang, Yanhong
Wang, Yuanjia
Wang, Yun
Wang, Ze
Wang, Zhishun
Ward, Susan
Wardecker, Britney
Warden, Melissa
Wardle, Margaret
Warnock, Kaitlin T.
Warren, Kenneth
Warrington, Lewis E.
Wass, Caroline E.
Wassink, Tom
Waterhouse, Barry D.
Waterhouse, Rikki
Waters, Alexa M.
Watkins, Linda
Watson Jr., Stanley J.
Watson, Adam
309
259
102, 274
246
64, 321
222
334
245
179
43, 337
223
306
279
318
228
46, 262
289
112, 290, 316
64, 321
268
123, 273
287
318
226
284
237
258
245
305
308, 311
264
290, 324, 325
284
74, 251
326
196, 227, 263
317, 331, 332
249
30
231
252
237, 281
221, 223
205
294
338
41, 222, 241
337
Watson, Annie
Watson, Jeannette
Watson, Stuart
Watteron, Lucas R.
Watts, Emily L.
Waylink, Suzanne
Webb, Sierra M.
Weber, Wade
Webhofer, Christian
Wee, Sunmee
Wegbreit, Ezra
Wei, I-Hua
Wei, Shau-Ming
Wei, Wei
Weickert, Cynthia
Weickert, Thomas W.
Weiden, Peter J.
Weiland, Barbara J.
Weinberger, Daniel
Weinshenker, David
Weisenbach, Sara
Weiss, Brett
Weissman, Myrna
Weissman, Myrna M.
Weisstaub, Noelia V.
Wells, Audrey
Welsh, Robert
Wemmie, John
Wendland, Jens R.
Werbeloff, Nomi
Westmoreland, Susan
Westphal, Ryan
Wetsel, William
Wettstein, Joseph G.
Whitfield-Gabrieli, Susan
Whitford, Thomas
Whybrow, Peter C.
Wiessner, Christoph
Wightman, R. Mark
Wilcox, Charles S.
Wilcox, Marsha
Wileyto, E. Paul
Wilhelmsen, Kirk C.
403
304
273
311
336
262
240
258, 304
320
334
304, 305
232
294
283
302
181, 240
240
285
288
111, 123, 191,
198, 238, 240,
246, 264, 265,
268, 273, 278,
286, 299, 318
94
322
237
31
242
304
257
232, 241, 242,
322
223
278
230
306
255
54, 256
334
249
276, 285
248
294
223
311
237
315, 317
317
ACNP 50th Annual Meeting • Final Program
Wilkins, Patricia P.
Wilkinson, Charles W.
Williams, Alison
Williams, Janet
Williams, Kyle
Williams, Leanne
Williams, Wendol
Williamson, Douglas
Willis, Brian
Willuhn, Ingo
Wilson, Alan
Wilson, Daniel R.
Wilson, Lisa B.
Wilson, Mark
Wilson, Steven P.
Wiltfang, Jens
Winder, Danny G.
Windhorst, Bert D.
Winetraub, Yonatan
Wing, Victoria C.
Winseck, Adam
Winsky, Lois
Winstanley, Catharine A.
Witkin, Jeffrey M.
Witten, Ilana
Woicik, Patricia A.
Wolf, Daniel
Wolfe, Barbara E.
Wong, Christopher
Wong, Dean
Wong, Erik H.F.
Woods, Scott
Woodward, John J.
Woolley, Josh
Woolwine, Bobbi
Worthington, John
Wright, M. Jerry
Wroolie, Tonita
Wroten, Melissa
Wu, Gang
Wu, Hui-Qiu
Wu, James
Wu, Qiang
Wu, Richard
Wulff, Sanne
274
326
255
227, 231, 271,
311
261
95, 278
242, 283
279
270
116, 138, 139,
266
288, 319, 322,
324
230
286
243
333
240
259
287
311
252
230
196
295
263, 308
263
289, 316
244, 275, 285
292
290
205
119, 262
274
259
276, 335
321
311
336
326
258
232
264
309
333
230
284
Wynn, Jonathan K.
Xie, Xiaohu
Xu, Jane
Xu, Xiaoyan
Yadav, Prem
Yamashita, Hiroshi
Yamawaki, Shigeto
Yan, Mingjin
Yang, Alex
Yang, Feng
Yang, Jianmin
Yang, Lingfeng
Yang, Shaolin
Yang, Wendy
Yang, Yihong
Yaqub, Maqsood
Yarnykh, Vasily
Ye, Tianzhang
Yehuda, Rachel
Yilmaz, Zeynep
Yingling, Michael D.
Yoon, Jin H.
Yoon, Jong
Yoon, Peter
Yoshida, Keiko
Yoshida, Takayuki
Yoshimura, Shinpei
Yoshinaga, Toshihiro
Yoshioka, Mitsuhiro
Yost, Julianne
Young, Allan H.
Young, Erica
Young, Jared
Young, Kimberly A.
Young, Kymberly D.
Young, L. Trevor
Youngstrom, Eric
Youngstrom, Jennifer
Yu, Dongzi
Yu, Lei
Yu, Qinghui
Yuan, Peixiong
Yuan, Qiaoping
Yue, Xiaomin
Yurgelun-Todd, Deborah
Yuskaitis, Christopher J.
404
234, 276
257
229, 252
287
54, 256
248
324
269
255
264
48, 277
292
320
325
286
287
283
268, 318
178, 279
280
235
253
275, 287, 319
268
248
338
324
306
338
54, 256
311
305
115, 125, 126,
188, 265, 266
324
234
208
277, 309, 329
277, 329
294
185
223
264
37, 238, 315,
316, 317
298
246, 313
295
ACNP 50th Annual Meeting • Final Program
Zacek, Robert
Zack, Martin
Zaczek, Robert
Zagoory-Sharon, Orna
Zai, Clement
Zaidi, Eram
Zakzanis, Konstantine K.
Zalcman, Steven
Zaman, Samir
Zamanyan, Alex
Zametkin, Molly
Zangen, Abraham
Zarate Jr., Carlos
Zarcone, Jennifer
Zeeuw, Patrick de
Zepf, Florian Daniel
Zetterberg, Henrik
Zhan, Liang
Zhang, Aifeng
Zhang, Fengyu
Zhang, Grace
Zhang, Haihong
Zhang, Han-Ting
Zhang, Huaibo
Zhang, Hui
Zhang, Jian-Ping
Zhang, Jianping
Zhang, Lingjiao
Zhang, Lishu
Zhang, Peter
Zhang, Wanli
Zhang, Xiadong
Zhang, Xiaodong
Zhang, Zhongqi
Zhao, Fangyi
Zhao, Jun
Zhao, Xin
255
324
255, 256
325
238, 280
289
274
121, 298
226
242
332
333
196, 293, 320,
330
282
245
221, 240
226
287, 322
272, 320, 322
238, 240
264
249
333
306
225
243
252
331
253
294
309
244
243
303
239
309
271
Zhao, Yonggang
Zhao, Yudong
Zheng, Minq-Qiang
Zhihua, Xie
Zhou, Lili
Zhou, Wenjun
Zhou, Xianjin
Zhou, Y. Sandra
Zhou, Zhifeng
Zhu, Chong-Bin
Zhu, Xun
Ziebell, Steven
Zimmermann, Marco
Zink, Caroline
Zipursky, Robert B.
Zirlin, Benjamin C.
Zisook, Sidney
Zivkov, Milana
Zohar, Joseph
Zook, Michelle
Zorick, Todd
Zorrilla, Eric P.
Zorumsk, Charles
Zou, Jian
Zou, Wei
Zoubovsky, Sandra P.
Zubieta, Jon-Kar
Zucker, Robert A.
Zukin, Suzanne
Zuo, Yantao
Zwart, Fenny
Zweifel, Larry
Zwir, Igor
Zywiak, William
405
231
249
247, 283
306
316
262
237
335
37, 315, 316,
317
225
290
237
240
115, 123, 124,
273
230
301
308, 311
229
178
258
290
303
196
306
236
268
64, 121, 288,
298, 321, 322,
325
288
110
289
245
204
238
68, 317, 332