High School Scientific Session. Learning Center ..... High School Science Initiative Day. Learning ...... Voon Wee Yong, Manoj Mishra, Khalil Rawji, Michael Keough ...... Seton Hall University, Department of Biological Sciences, South. Orange ...
AMERICAN SOCIETY FOR N E U R O C H E M I S T RY
MEETING PROGRAM 46TH ANNUAL ASN MEETING
AT L A N TA , G E O R G I A MARCH 14 — 18, 2015
WELCOME
ASNeurochem.or g
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
Instructions and Hints To use this all new animated Program and Abstract Book we have some hints for you to ease navigation. This should not stop you from exploring this feature rich application on your own.
Instructions & Hints
Different ways of navigation • Table of Contents: Click the very left icon on the button tool bar. Click "Expand All/Collapse All" at the button to see the list of all items in each book. • Thumbnail view: Use the second icon on the button tool bar to quickly browse through the pages. • Arrows: Use the single arrows in the button tool bar on the right to switch between neighboring double pages. • Click and Zoom: Clicking in the middle of a page will zoom the page. You can adjust the zoom level with the slider in the button bar of the page. Dragging the page with the mouse will move the visible section. • Search: Use the search field with the spyglass icon in the top tool bar on the right to search through the program and abstract book. The result list will show on the left side. • Bookmarks: To create your own bookmarks please use the third icon on the button tool bar. We hope you enjoy the new animated Program and Abstract Book.
Sincerely, The Program Committee
Manipulate receptor and enzyme Manipulate function with inhibitor inhibitors, s, activ activators, ators, agonists and antagonists M
G1
G2
S
Low compromise to protein-protein interactions
Range of se lectivity selectivity
for Rapid effects for temporal control
Cell-permeable Cell-per meable and b bloodloodbrain bar barrier rier per permeable meable options
long-ter m R Reversible eversible and long-term effects
Easy to use
JJoin oin peer re reviewed viewed researc researchers hers wor worldwide ldwide and benefit from: - Quality yyou ou can tr trust ust – cita citations tions in n numerous umerous high impact factor jour journals nals -R Resources esources to he help lp yyour our researc research h - Biological vvalidation alidation Our customer customers s are sa saying... ying...
“T “Their heir outstanding quality shows their commitment to fur furthering thering researc research”. h”. Prof David Jane, University of Bristol
abcam.com/Ad abcam.com/Advantages vantages
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
Etty (Tika) Benveniste President
Vlad Parpura Secretary
Jean Harry Treasurer
Steven W. Levison Past President
Babette Fuss President-Elect
COUNCIL: Steven Barger
Dear Participants of the 46th ASN Annual Meeting: On behalf of the ASN Council, Program and Host Committees, we would like to welcome you to Atlanta, Georgia and the 46th Annual Meeting of the American Society for Neurochemistry. The scientific program has been designed to maximize your ability to benefit scientifically from this meeting, with four days of symposia and colloquia that address the four major themes of our Society: -Building the Nervous System -Glial Cell Biology -Molecular and Cell Biology of the Nervous System -Neurodegeneration and Disease
Erhard Bieberich Ernesto Bongarzone Cheryl Dreyfus James Hewett
Special efforts have been taken to encourage Young Investigator participation with a session on funding opportunities, lunch with the plenary speaker each day, travel awards and over 12 abstracts chosen for oral sessions, to provide another avenue of recognition. To maximize scientific interactions
Dianna Hynds
and discussions, posters will be up for 2 consecutive days, with a two hour
Eric Murphy
session for authors to be present. Thank you to all the volunteers, session
Michael Nichols
chairs and speakers for your contribution to the success of this meeting.
Juana Maria Pasquini Arturo Ortega
We look forward to seeing you next year in Denver, Colorado at the 47th ASN meeting.
Seema Tiwari-Woodruff Emma Wilson SECRETARY-ELECT: Sandra Hewett ALTERNATE COUNCIL: Colin Combs
Tika Benveniste
Wilma Friedman
Malú Tansey
President
Scientific Program Chair
Host Committee Chair
!
Michael Fox
1
®
Basic Ba asic N Neurochemistry, eurochem mistryy, Eighth Edition Edittion Principles P rin nciples l off M Molecular, olecular l l , Cellular C Cellular, ll l , and dM Medical ed dicall N Neurobiology eurobiology b
THE CLASSIC TEXT FOR O OVER VER 40 YEARS! This outstanding book continues to be the global standard for information This on the biochemistry off the nervous system. It is the standard reference for neuroscience,, bringing basic and clinical exploring the translational nslational nature off neuroscience neurosc QHXURVFLHQFHWRJHWKHULQRQHDXWKRULWDWLYHYROXPH7KHVXEWLWOHUHȈHFWVWKH Q HXURVFLHQFHWRJHWKHULQRQHDXWKRULWDWLYHYROXPH7KHVXEWLWOHUHȈHFWVWKH expanded attention to the links between neurochemistry and neurologic G GLVHDVH$FRPSDQLRQVLWHZLWKGRZQORDGDEOHȇOHVRIȇJXUHVER[HVDQG LVHDVH$FRPSDQLRQVLWHZLWKGRZQORDGDEOHȇOHVRI ȇJXUHVER[HVDQG references are available ailable to purchasers off the book. Dec. 2011, Hardback, 1,120 pages ISBN: 9780123749475 Print P rint price: $125.00 Member ASN M ember price: $93.75 available.. ebook format also available
Edited by Scott T T.. Brady Brady,, George JJ.. Siegel, R. W Wayne ayne Albers Albers,, and Donald L. P Price rice © American Society for N Neurochemistry eurochemistry
ISN and ASN members save 25% Order online at store store.elsevier.com/9780123749475 .elsevier.com/9780123749475
Enter promotional code NEURO215 at checkout to receive receivve your discount. 20150119
Plan Now to Attend
47th Annual Meeting American Society for Neurochemistry
Denver, Colorado, March 19 – 23, 2016 Join us for four days of symposia, colloquia and workshops designed to address the four major themes of our Society: •
Building the Nervous System
•
Molecular and Cell Biology of the Nervous System
•
Glial Cell Biology
•
Neurodegeneration and Disease
On-line Abstract Submissions: Sept. 15 – Nov. 30, 2015 www.ASNeurochem.org
ASN 2016 – Grand Hyatt Denver Babette Fuss, ASN President
Wendy Macklin, Host Committee Chair Jony Kipnis, Scientific Program Chair
Make Your Plans Today! © 2014 American Society for Neurochemistry Background Photo: Michelle Olsen
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
Hyatt Regency Map
Lobby Level
2
Ballroom Level (LL1)
Exhibit Level (LL2)
2015 Agenda-At-A-Glance All functions are held at the Hyatt Regency Atlanta Friday, March 13 3:00 pm – 7:00 pm
ASN Registration Desk Open
Regency Foyer
Saturday, March 14 7:00 am – 6:00 pm ASN Registration Desk Open 8:00 am – 11:30 am Pre-Meeting Workshop Optogenetics Tools, Techniques and Applications in Neuroscience and Neurochemistry 12:00 pm – 5:00 pm ASN Council Meeting I 4:00 pm – 6:00 pm ASN Public Forum The Social Brain: From the Chemistry of Relationships to Autism Spectrum Disorder 6:00 pm – 7:30 pm ASN President’s Welcome Reception
Regency Terrace/Regency V
Sunday, March 15 7:00 am – 6:00 pm 8:00 am – 9:15 am 9:15 am - 10:00 am 10:00 am - 12:00 pm 12:00 pm – 1:00 pm 12:00 am – 1:00 pm 1:00 pm - 5:30 pm 5:30 pm – 7:00 pm
Regency Foyer Regency VI Regency VII Regency V, VI, Hanover C, D Chicago C, D TBD Regency V, VI, Hanover C, D Regency VII
Chicago C, D Hanover C, D
Monday, March 16 7:00 am – 5:00 pm ASN Registration Desk Open 8:00 am – 9:15 am ASN Plenary Session - Jay Giedd, MD 9:00 am – 2:00 pm High School Scientific Session 9:15 am - 10:00 am Poster Viewing/Coffee Break 10:00 am - 12:00 pm ASN Scientific Sessions 12:00 pm – 1:00 pm Women in Neurochemistry (WIN) Luncheon - Ticket Required 12:00 pm – 1:00 pm Lunch with Plenary Speaker - Dr. Giedd - Ticket Required 1:00 pm - 5:30 pm ASN Scientific Sessions 5:45 pm - 8:00 pm Young Investigators Funding Opportunities & Mingle 5:45 pm – 7:00 pm ASN Business Meeting — All Members Invited to Attend
Regency Foyer Regency VI Learning Center Regency VII Regency V, VI, Hanover C, D Hanover A, B Chicago C, D Regency V, VI, Hanover C, D Regency V Learning Center
Tuesday, March 17 7:00 am – 5:00 pm ASN Registration Desk Open 8:00 am – 9:15 am ASN Plenary Session - Barbara Hempstead, PhD 9:15 am - 10:00 am Poster Viewing/Coffee Break 10:00 am - 12:00 pm ASN Scientific Sessions 12:00 pm – 1:00 pm Lunch with Plenary Speaker - Dr. Hempstead - Ticket Required 12:00 pm – 1:00 pm Metabolic Brain Disease Editorial Board Lunch 1:00 pm - 5:30 pm ASN Scientific Sessions 5:30 pm - 7:00 pm Poster Viewing/Wine & Cheese
Regency Foyer Regency VI Regency VII Regency V, VI, Hanover C, D Chicago C, D TBD Regency V, VI, Hanover C, D Regency VII
Wednesday, March 7:00 am – 5:00 pm 8:00 am – 9:15 am 9:15 am - 10:00 am 10:00 am - 12:00 pm 12:00 pm – 3:00 pm 12:00 pm – 1:00 pm 1:00 pm – 3:30 pm 1:00 pm - 5:30 pm 7:30 pm – 11:00 pm
Regency Foyer Regency VI Regency VII Regency V, VI, Hanover C, D Hanover A, B Chicago C, D Regency VII Regency V, VI, Hanover C, D Regency VII
18 ASN Registration Desk Open ASN Plenary Session - Scott Brady, PhD Poster Viewing/Coffee Break ASN Scientific Sessions ASN Council Meeting II Lunch with Plenary Speaker - Dr. Brady- Ticket Required Poster Viewing - Posters Must be Removed by 3:30 ASN Scientific Sessions ASN Closing Reception/Dinner - Ticket Required
2 015 Agenda-At-A-Glance
ASN Registration Desk Open ASN Plenary Session - Dr. Patrizia Casaccia Poster Viewing/Coffee Break ASN Scientific Sessions Lunch with Plenary Speaker - Dr. Casaccia - Ticket Required Neurochemical Research Editorial Board Lunch ASN Scientific Sessions Poster Viewing/Wine & Cheese
Regency Foyer Learning Center
3
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
S a t u r d a y, M a r c h 1 4 , 2 01 5 7:00 am – 6:00 pm
ASN Registration Desk Open
Regency Foyer
Pre-Meeting Workshop
Optogenetics Tools, Techniques & Applications in Neuroscience & Neurochemistry 8:00 am – 11:30 am (Pre-Registration Required)
Learning Center
Session Chair: Ling Wei (Emory University) Co-Chair: Robert Gross (Emory University) 8:00 – 8:20 am Ken Berglund (Emory) Development of Optogenetics Tools and Techniques 8:20 - 8:40 am Isaac Clements (Axion Biosystems) Multiwell optogenetic stimulation for high-throughput disease modeling and drug discovery 8:40 - 9:00 am Michael Mohammadi (Andor) Precision light patterning for microscopy-based optogenetics
Saturday
9:00 - 9:20 am Arthur English (Emory) Application of Optogenetics in Neuroscience and Neurochemistry Research 9:20 - 9:40 am Robert Gross (Emory) Optogenetics and Epilepsy Treatment 9:40 - 10:00 am Ling Wei (Emory) Stem Cells and Optogenetics: Novel Approaches for the Treatment of Focal Ischemic Stroke 10:00 - 10:20 am Adriana Galvan (Emory) In vivo Optogenetic Control of Striatal and Thalamic Neurons in Non-human Primates 10:20 - 10:40 am Teresa Sanders (Emory) Optogenetic Modulation of Basal Ganglia Circuit Activity in Rodents 11:00 am Bus to Emory 11:30 to 12:30 Lab demonstration at Emory University 12:30 pm to 1:30 Bus to Yerkes Field Station (box lunch on the bus) 2:00 - 3:30 pm Site visit at Yerkes Field Station 3:30 - 4:30 pm Bus to Hyatt Regence Hotel Atlanta Downtown Sponsored by: Andor, Axion BioSystems, Thor Labs, Atlanta Clinical & Translational Science Institute 12:00 pm – 5:00 pm
ASN Council Meeting I
Chicago C, D
ASN Public Forum
The Social Brain: From the Chemistry of Relationships to Autism Spectrum Disorder 4:00 pm – 6:00 pm
Hanover C, D
Chair: Anne Murphy Speakers & Panelists Include: Ami Klin, PhD, Director, Marcus Autism Center and NIH Autism Center of Excellence in Atlanta Celine Saulnier, PhD, Clinical Director, Research, Division of Autism & Related Disorders, Emory Larry Young, PhD, Director, Emory Center for Translational Social Neuroscience Greg Abowd, PhD, President, Atlanta Autism Consortium Sponsored by: Atlanta Clinical & Translational Science Institute and Atlanta Science Festival
6:00 pm – 7:30 pm
4
ASN President’s Welcome Reception Sponsored by: University of Alabama at Birmingham (UAB), Dean’s Office
Regency Terrace/Regency V
46 th Annual Meeting Atlanta, Georgia – 2015
S u n d a y, M a r c h 1 5 , 2 01 5 7:00 am – 6:00 pm
ASN Registration Desk Open
Regency Foyer
8:00 am – 8:15 am
General Session Welcome - Tika Benveniste, ASN President • New Member Recognition • Travel Awardees Recognition
Regency VI
8:15 am – 9:15 am
PL01-Plenary Speaker - Dr. Patrizia Casaccia Looking at the Epigenomic Landscape of Developing Oligodendrocytes
Regency VI
9:15 am – 10:00 am
Poster Session I/Coffee Break
Regency VII
10:00 am – 12:00 pm
ASN Scientific Sessions
Regency V microRNAs (miRNAs) in Alzheimer's disease (AD) and age-related macular degeneration (AMD) Chair: Walter J. Lukiw Co-Chair: Debomoy K. Lahiri Introduction to microRNA in AD and AMD – Walter J. Lukiw S01-01 Debomoy Lahiri ROLE OF MicroRNA-346 IN IRON HOMEOSTASIS AND ALZHEIMER’S DISEASE S01-02 Peng Jin MicroRNA-650 MODULATES THE PATHOGENESIS OF ALZHEIMER’S DISEASE THROUGH REGULATING CDK5 S01-03 Sebastien Hebert MicroRNA THERAPY FOR ALZHEIMER'S DISEASE S01-04 Walter J. Lukiw MicroRNA: SPECIATION & COMPLEXITY, SEQUENCE & STABILITY, VIROID-LIKE PROPERTIES & DISEASE PROPAGATION WITHIN THE HUMAN CNS
12:00 pm – 1:00 pm
Colloquium CO1
Proinflammatory Mechanisms in Neurodegeneration Chair: Michael Nichols C01-01 Linda Van Eldik TARGETING SIGNALING PATHWAYS THAT LEAD TO INFLAMMATORY CYTOKINE DYSREGULATION IN NEURODEGENERATIVE DISORDERS C01-02 Colin Combs THE CONTRIBUTION OF APP TO MICROGLIAL ACTIVATION IN ALZHEIMER’S DISEASE C01-03 Andrea Tenner EFFECTS OF COMPLEMENT ACTIVATION FRAGMENT C5A IN ALZHEIMER'S DISEASE MOUSE MODELS C01-04 Michael Nichols INFLUENCE OF AMYLOID-BETA AGGREGATION STATE ON INNATE IMMUNE PATHWAYS
Hanover C, D Symposium SO2 Glial Vesicular, Metabolic and Morphologic Function in Health and Disease Chair: Robert Zorec Co-Chair: Vladimir Parpura S02-01 Nina Vardjan ADRENERGIC CONTROL OF ASTROCYTE MORPHOLOGY, EXCITABILITY AND METABOLISM
Sunday
Symposium SO1
Regency VI
S02-02 Michela Matteoli DUAL ROLE OF MICROGLIA IN NEURODEGENERATIVE DISEASES S02-03 Vedrana Montana METABOLIC REGULATION OF VESICULAR GLUTAMATE RELEASE FROM CULTURED ASTROCYTES S02-04 Maiken Nedergaard THE TRIPARTITE SYNAPSE A DEVELOPMENTAL RESTRICTED PHENOMENON?
Lunch with Plenary Speaker - Dr. Casaccia Chicago C, D Sponsored by: International Society for Neurochemistry Ticket Required
2 015 Sanofi-Genzyme Travel Award The Sanofi-Genzyme travel award has been established to support an outstanding young scholar’s participation at the ASN Meeting.
Javier Palazuelos
Stony Brook University, State University of New York
5
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
S u n d a y, M a r c h 1 5 , 2 01 5 1:00 pm – 3:00 pm
ASN Scientific Sessions
Regency V
Symposium SO4
Symposium SO3
Sunday
Hanover C, D
Regency VI
RNA Toxicity in Neurodegeneration Chair: Rita Sattler Co-Chair: Christopher Donnelly
Frontiers of Neurolipids Chair: Stefka Spassieva Co-Chair: Glyn Dawson
S03-01 Rita Sattler ROLE OF SYNAPTIC DYSFUNCTION IN C9ORF72-MEDIATED PATHOGENESIS IN PATIENT-DERIVED IPS NEURONS AND IN VIVO ANIMAL MODELS
S04-01 Robert Ledeen DEFICIENCY OF GM1 AS CAUSE OF IDIOPATHIC PARKINSON’S DISEASE: RESTORATION OF GM1-GDNF INTERACTION AS CURE IN PD MOUSE
S03-02 Christopher Donnelly NUCLEAR TRANSPORT DEFECT UNDERLIES C9ORF72 ALS/FTD NEURONAL INJURY
S04-02 Lihong Zhao TOXICITY OF LONG CHAIN BASES: ACCUMULATION OF LONG CHAIN BASES UNDERLIES NEURODEGENERATION CAUSED BY CERS1 DEFICIENCY
S03-03 Tania Gendron RAN TRANSLATION IN C9ORF72 FRONTOTEMPORAL DEMENTIA AND AMYOTROPHIC LATERAL SCLEROSIS
S04-03 Glyn Dawson CERAMIDE HOMEOSTASIS IN THE BRAIN S04-04 Erhard Bieberich CERAMIDE-ENRICHED COMPARTMENTS IN NEURAL STEM CELL DIFFERENTIATION AND NEURODEGENERATION
S03-04 Auinash Kalsotra CUG RNA TOXICITY DISRUPTS THE DEVELOPMENTALLY REGULATED MICRORNA PROGRAM IN MYOTONIC DYSTROPHY S03-05 Aaron Gitler YEAST GENETIC SCREENS REVEAL NEW INSIGHTS INTO NEURODEGENERATIVE DISEASE MECHANISMS: TDP-43, FUS/TLS, C9ORF72, AND BEYOND S03-06 Daniela Zarnescu TDP-43 DEPENDENT TRANSLATION DYSREGULATION IN AMYOTROPHIC LATERAL SCLEROSIS
S04-05 Narayan Bhat CELL DEATH SPREADING BY EXOSOME SHEDDING: THE ROLE OF CERAMIDEENRICHED SECRETED MICROVESICLES IN SYNERGISTIC CYTOKINE TOXICITY
Symposium SO5
Novel Strategies in Stem Cell Therapy and Clinical Potentials Chair: Michael Chopp Co-Chair: Shan Ping Yu S05-01 Koji Abe NEUROPROTECTION AND GENE-STEM CELL THERAPY FOR ISCHEMIC STROKE S05-02 Xiao-Ming Xu CO-TRANSPLANTATION OF GLIAL RESTRICTED PROGENITOR AND SCHWANN CELLS IN PROMOTING REPAIR AFTER SPINAL CORD INJURY S05-03 Michael Chopp MICRO RNA AND SIGNALING PATHWAYS IN STEM CELL THERAPY AFTER ISCHEMIC STROKE S05-04 Ling Wei IPS CELL TRANSPLANTATION AND COMBINATIONAL THERAPY FOR THE REGENERATIVE TREATMENT AFTER ISCHEMIC STROKE
S04-06 Stefka Spassieva PACLITAXEL TREATMENT GENERATES NEUROTOXIC DEOXYSPHINGOLIPIDS
S03-07 Robin Reed ALS-CAUSING MUTATIONS IN THE FUS PRION-LIKE DOMAIN BLOCK TRANSCRIPTION-COUPLED PRE-MRNA SPLICING
2 015 ASN NEURO Award The ASN NEURO travel award has been established to support an outstanding young scholar’s participation at the ASN Meeting.
Justin Brooks
University at Buffalo School of Medicine
6
46 th Annual Meeting Atlanta, Georgia – 2015
S u n d a y, M a r c h 1 5 , 2 01 5 3:30 pm – 5:30 pm
Regency V Symposium SO6
Autophagy in Neurodegeneration: Role in Disease and Therapy Target Chair: Kenneth Hensley Co-Chair: Marni Harris-White S06-01 Kenneth Hensley CELL-PENETRATING LANTHIONINE KETIMINE DERIVATIVE ACTIVATES AUTOPHAGY THROUGH A NOVEL MECHANISM INVOLVING CRMP2 AND MTORC1
S06-03 Paul Salvaterra AUTOPHAGY-ENDOSOMAL-LYSOSOMAL PATHWAY DYSFUNCTION IN A!(1-42) DEPENDENT NEURODEGENERATION S06-04 Sandra Maday AUTOPHAGOSOME BIOGENESIS IN PRIMARY NEURONS FOLLOWS AN ORDERED AND SPATIALLY REGULATED PATHWAY
Hanover C, D
Regency VI Symposium SO7 Central and Peripheral Inflammatory and Immune Responses in Parkinson's Disease Chair: Malú Tansey Co-Chair: Etty (Tika) Benveniste S07-01 Howard Gendelman DESTRUCTIVE OR PROTECTIVE IMMUNITY IN PARKINSON'S DISEASE S07-02 Matt LaVoie ROLES FOR MICROGLIAL LRRK2 EXPRESSION AND FUNCTION IN PARKINSON’S DISEASE S07-03 Kathleen Shannon CRITICAL GASTROINTESTINAL FUNCTIONS AND DISEASE MARKERS IN PARKINSON'S DISEASE S07-04 Malú Tansey A COMMON GENETIC VARIANT IS ASSOCIATED WITH ALTERED MHC-II EXPRESSION AND SYNERGIZES WITH PYRETHROID EXPOSURE TO INCREASE PD RISK
Symposium SO8
Wiring of the Nervous System: From Molecular Mechanisms to Complex Behaviors Chair: Tracy Tran S08-01 Anthony Barnes A MOLECULAR PATHWAY REGULATING AXOGENESIS S08-02 Wenquin Luo COMBINATORY CIS AND TRANS RET SIGNALING CONTROLS THE SURVIVAL AND CENTRAL PROJECTION GROWTH OF RAPIDLY ADAPTING MECHANORECEPTORS S08-03 Tracey Tran ROLES FOR NEUROPILIN 2 SIGNALING IN HIPPOCAMPALAND CORTICO-STRIATAL-DEPENDENT LEARNING AND MEMORY S08-04 Christopher Deppmann EXTRINSIC MECHANISMS GOVERNING INJURY-INDUCED AXON DEGENERATION
Sunday
S06-02 Mami Harris-White THE ELUSIVE ROLE OF AUTOPHAGY IN TRAUMATIC BRAIN INJURY
ASN Scientific Sessions
S07-05 Etty Benveniste INHIBITION OF THE JAK/STAT PATHWAY PROTECTS AGAINST ALPHA-SYNUCLEIN-INDUCED NEUROINFLAMMATION AND DOPAMINERGIC NEURODEGENERATION
5:30 pm – 7:00 pm
P oster Session I – Author’s Present – Wine & Cheese
Regency VII
7:00 pm
Neurochemical Research Editorial Board Dinner
TBA
7
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
M o n d a y, M a r c h 1 6 , 2 01 5 7:00 am – 5:00 pm
ASN Registration Desk Open
Regency Foyer
8:00 am – 8:15 am
General Session • ASN NEURO Award Presented by Monica Carson
Regency VI
8:15 am – 9:15 am
PL02-Plenary Speaker Jay Giedd, MD The Teen Brain: Insights from Neuroimaging
Regency VI
9:00 am – 2:00 pm
High School Science Initiative Day Learning Center Chair: Kyle Frantz, Sponsored by Atlanta Science Festival
9:15 am – 10:00 am
Poster Session I/ Coffee Break
10:00 am – 12:00 pm
ASN Scientific Sessions
Regency V
Regency VI
Monday
Symposium SO9
Symposium S10
Astrocytes as Obligatory Partners in Purinergic and Glutamatergic Neurotransmission Chair: Arne Schousboe Co-Chair: Vladimir Parpura
Neuropsychiatric Disorders: New Mechanisms and Models Chair: Cristina Ghiani Co-Chair: Antonieta Lavin S10-01 Dawn Loh CIRCADIAN RHYTHM DISRUPTION IN A MOUSE MODEL OF RETT SYNDROME
S09-01 Alex Verkhratsky ASTROCYTES AS OBLIGATORY PARTNERS IN PURINERGIC AND GLUTAMATERGIC NEUROTRANSMISSION S09-02 Vlad Parpura ON VESICULAR FUSIONS IN ASTROCYTES: SINGLE VESICLE/MOLECULE APPROACHES S09-03 Michael Robinson ASTROGLIAL GLUTAMATE TRANSPORT: AT THE INTERSECTION OF EXCITATORY SIGNALING AND BRAIN ENERGETICS S09-04 Mary McKenna GLUTAMATE AS A KEY ENERGY SUBSTRATE FOR SUPPORTING ASTROCYTIC GLUTAMATE TRANSPORT AND NEURONAL-GLIAL INTERACTIONS
S10-02 Atsushi Kamlya A NATURAL PRODUCT AS A TOOL TO EXPLORE NOVEL THERAPEUTIC TARGETS FOR STRESS-INDUCED IMMUNE CHANGES AND DEPRESSIVE BEHAVIORS S10-03 Cristina Ghiani GENETIC RISK FACTORS AND BRAIN MALDEVELOPMENT S10-04 Victor Faundez MOLECULAR AND GENETIC STUDIES OF A SCHIZOPHRENIA SUSCEPTIBILITY INTERACTOME
Regency VII
Hanover C, D Symposium S11 Dissecting the Molecular Mechanisms of Acute Neurologic Injury Chair: Carol Troy S11-01 Ulrich Hengst INTRA-AXONAL PROTEIN SYNTHESIS AS A MEDIATOR OF LONG-RANGE NEURODEGENERATIVE SIGNALS S11-02 Russell Nicholls THE ROLE OF BETA AMYLOID AND TAU IN THE RESPONSE TO TRAUMATIC BRAIN INJURY S11-03 Wilma Friedman NEUROTROPHIN REGULATION OF NEURONAL DEATH AFTER INJURY S11-04 Carol Troy MECHANISMS OF AND THERAPIES FOR BRAIN INJURY AND NEURODEGENERATION
S10-05 Shannon Gourley DURABLE CONSEQUENCES OF SOCIAL ADVERSITY IN ADOLESCENCE: NEUROBIOLOGICAL AND BEHAVIORAL OUTCOMES FOLLOWING ISOLATION S10-06 Antonieta Lavin EFFECTS OF FINGOLIMOD ADMINISTRATION IN DYSBINDIN DEFICIENT MICE: A GENETIC MODEL FOR COGNITIVE DEFICITS
8
12:00 pm – 1:00 pm
Lunch with Plenary Speaker - Dr. Giedd Chicago C, D Sponsored by: International Society for Neurochemistry Ticket Required
12:00 pm – 1:00 pm
Women in Neurochemistry (WIN) Hanover A, B Luncheon & Discussion Sponsored by UAB Department of Cell, Developent & Integrative Biology Ticket Required
46 th Annual Meeting Atlanta, Georgia – 2015
M o n d a y, M a r c h 1 6 , 2 01 5 1:00 pm – 3:00 pm
ASN Scientific Sessions
Regency V Symposium S12
Signaling Pathways Regulating Remyelination Chair: Teresa Wood S12-01 Li-Jin Chew SOX17 IN WHITE MATTER REPAIR: VENTURING BEYOND WNT S12-02 Sharyl Fyffe-Maricich ERK2 MAP KINASE REGULATES TIMELY REMYELINATION IN THE ADULT BRAIN
S12-04 Seema Tiwari-Woodruff THERAPEUTIC INTERVENTION TO INDUCE OLIGODENDROCYTE SURVIVAL AND DIFFERENTIATION IN MOUSE MODELS OF DEMYELINATION
Colloquium C02
Post-translational Prenylation of GTPases in Neurodegeneration and Repair Chair: Evan Stubbs Co-Chair: DiAnna Hynds C02-01 Ling Li PROTEIN PRENYLATION AND SYNAPTIC PLASTICITY: IMPLICATIONS FOR ALZHEIMER’S DISEASE C02-02 MariVi Tejada-Simon TRICKING THE CHOLESTEROL PATHWAY TO TARGET NEURODEVELOPMENTAL DISORDERS: ISOPRENOID DYSFUNCTION IN AN ANIMAL MODEL OF AUTISM C02-03 Jairus Reddy FUNCTIONAL ROLE OF NATIVE, NON-PRENYLATABLE GTPASES IN NEURON MORPHOLOGY AND NEURITE OUTGROWTH C02-04 Kelly Langert CDC42 AND RALA GTPASES AS NOVEL THERAPEUTIC TARGETS IN INFLAMMATORY AUTOIMMUNE NEUROPATHY C02-05 Cynthia Pervan PRENYLATION OF RHO GTPASES: A NOVEL MECHANISM REGULATING GENE EXPRESSION AND PROTEIN STABILITY
3:00 pm – 3:30 pm
Coffee Break
Hanover C, D Symposium S13
Mechanisms of Cell Death and Diseases Chairs: Elias Aizenman Co-Chair: Shan Ping Yu S13-01 Elias Aizenman CONVERGENT ZINC AND CALCIUM SIGNALING CASCADES IN KV2.1-ENABLED NEURONAL CELL DEATH S13-02 Durga Mohapatra THE SOMATODENDRITIC K+ CHANNEL KV2.1 IS A CRITICAL REGULATOR OF NEURONAL SURVIVAL AND DEATH S13-03 Roger Simon MITOCHONDRIAL BASE EXCISION REPAIR ATTENUATES REPERFUSION INJURY IN ISCHEMIC STROKE S13-04 Marc Simard THE Sur1-Trpm4 CHANNEL IN CNS INJURY S13-05 Dandan Sun WNK3-SPAK/OSR1-NKCC1 SIGNALING PATHWAY IN ISCHEMIC BRAIN INJURY
Monday
S12-03 Cheryl Dreyfus GLIA-DERIVED BDNF AFFECTS RECOVERY FROM A DEMYELINATING INJURY
Regency VI
S13-06 Shan Ping Yu DYSFUNCTION OF MEMBRANE TRANSPORTER/EXCHANGER AND HYBRID CELL DEATH IN NEURONAL AND CANCER CELLS
Terrace/Regency Foyer
2 015 Young Investigator Educational Enhancement Awards A special thank you to the International Sociey for Neurochemistry for their support and generous contribution to the YIEE Awards. David Braun, University of Illinois @ Chicago Isis Carletti, Rutgers Biomedical and Health Sciences April Cox, Medical University of South Carolina Evan Goldstein, The Ohio State University Corey Heffernan, Rutgers, The State University of New Jersey Emily Mathews, University of Colorado Jarius Reddy, Texas Woman’s University ShiPing Zou, Virginia Commonwealth University
9
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
M o n d a y, M a r c h 1 6 , 2 01 5 3:30 pm – 5:30 pm
Regency V Symposium S14
Monday
Retromer and Neurodegenerative Diseases Chair: Wen-Chen Xiong
ASN Scientific Sessions
Symposium S15
Endoplasmic Reticulum Stress in Myelin Disorders Chair: Wensheng Lin
S14-01 Dagmar Ringe CHEMICAL CHAPERONES AS AN APPROACH TO TREATMENT OF ALZHEIMER’S DISEASE
S15-01 Wensheng Lin IMPAIRED EIF2B ACTIVITY IN OLIGODENDROCYTES CONTRIBUTES TO VANISHING WHITE MATTER DISEASE PATHOGENESIS
S14-02 Samuel Gandy VPS10-FAMILY RECEPTORS IN NEURODEGENERATION AND INSULIN RESISTANCE
S15-02 Sharon Way OLIGODENDROCYTE PROTECTION BY THE INTEGRATED STRESS RESPONSE: A POTENTIAL PHARMACOLOGICAL TARGET
S14-03 Wen-Cheng Xiong VPS35-DEFICIENCY IS A RISK FACTOR FOR THE PATHOGENESIS OF BOTH ALZHEIMER’S DISEASE AND PARKINSON’S DISEASE S14-04 Xiongwei Zhu PARKINSON DISEASE-ASSOCIATED MUTANT VPS35 CAUSES ABNORMAL MITOCHONDRIAL DYNAMICS AND MITOCHONDRIAL DYSFUNCTION
Hanover C, D
Regency VI
Symposium S16 Wiring Axons by mRNA Regulation in Neuronal Health and Disease Chair: Jeffery Twiss Co-Chair: Gary Bassell S16-01 Nora Perrone-Bizzozero OPPOSING ROLES OF HUD AND KSRP IN SHARED TARGET MRNA STABILITY AND AXONAL GROWTH S16-02 Daniela Zarnescu FUTSCH/MAP1B IS A TRANSLATIONAL TARGET OF TDP-43 AND IS NEUROPROTECTIVE IN A DROSOPHILA MODEL OF ALS
S15-03 Lawrence Wrabetz ENDOPLASMIC RETICULUM STRESS IN CHARCOT MARIE TOOTH NEUROPATHY
S16-03 Jeffery Twiss RNA PROTEIN INTERACTIONS FOR AXONALLY TRANSPORTED MRNAS
S15-04 Scott Whittemore ER STRESS AND SPINAL CORD INJURY
S16-04 Gary Bassell IMPAIRED MRNP COMPLEX ASSEMBLY AND AXONAL MRNA LOCALIZATION IN A MOUSE MODEL OF SPINAL MUSCULAR ATROPHY S16-05 Ben Szaro COORDINATING SYNTHESIS OF AXONAL CYTOSKELETAL PROTEINS WITH CELL SIGNALING PATHWAYS THROUGH RNA REGULATION BY HNRNP K
5:45 pm – 7:30 pm
ASN Business Meeting - All Members Welcome!
Learning Center
5:45 pm – 8:00 pm
Young Investigators Funding Opportunities & Mingle Regency V Chair: Sanoj K. Suneja, PhD, National Institute on Aging/NIH/DHHS Presentation followed by Pizza & Beverages
T-Shirt $15 S-M-L-XL
Now on Sale
ASN 2015 T-Shirts and Mugs Designed by the Host Committee and Student Volunteers
Mug $10
10
Purchase Yours at the ASN Registration Desk
Poster Session I NO PHOTOGRAPHY ALLOWED OF POSTERS
Authors Present Sunday: 5:30—7:00pm / Monday: 9:30—10:00am
Sunday - Monday PSM01 Development, Differentiation and Disorders PSM01-01 Abha Chauhan Impaired mitochondrial biogenesis in the cerebellum of autistic subjects: Reduced levels of PGC1" and NRF1
PSM01-03 Asem Singh TPH1 gene involvement in serotonergic abnormality of autism spectrumdisorder: a genetic and genotype-phenotype correlation study PSM01-04 Alexandra Wright Impact of Site-directed Mutagenesis of Actin related protein 3 on Interface Dynamics PSM01-05 Emily Mathews Investigating the mechanistic basis of cholesterol-mediated myelination PSM01-06 Aboozar Monavarfeshani Lrrtm1 is necessary for the development of corticogeniculate inputs in the mouse dorsal lateral geniculate nucleus PSM01-07 JinXiang Dai Olig1 function is required for oligodendrocyte differentiation in the mouse brain PSM01-08 Juana Pasquini Oligodendrocyte maturation through gestational iron deprivation: the road not taken PSM01-09 Juan Zanin Role of p75NTR In Cell Cycle Regulation during Cerebellar Development PSM01-10 Kathryn Bercury Post-mitotic neuronal ablation of Raptor and Rictor impacts developmental oligodendrocyte differentiation and myelination
PSM01-15 Rashad Hussain Integrin linked kinase (ILK) deletion reduces commitment to the oligodendrocyte lineage
PSM03-06 Douglas Feinstein Liver kinase B1 deficiency increases astrocyte proliferation and activation PSM03-07 Paul Drew PPAR-# Agonist Pioglitazone Suppresses Ethanol-Induced Neuroinflammation in a Mouse Model of Fetal Alcohol Spectrum Disorder
PSM01-17 Jianmin Su Matricryptins derived from collagen XIX trigger inhibitory synapse formation
PSM03-08 Elma Frias Investigating the role of aquaporin-4 in the influx of leukocytes during Toxoplasma gondii infection
PSM01-18 Tatyana Budylin Sex-specific Neonatal Hippocampal Neurogenesis and Mood Disorders: Possible Cooperation between Serotonin 1A receptor and GPR30
PSM03-09 Emma Wilson Toxoplasma gondii infection disrupts CNS glutamate homeostasis and neuronal connectivity: implications for behavioral abnormality
PSM02 Drugs of Abuse: Alcohol, Cocaine, Methamphetamine
PSM03-10 Charu Garg Pharmacological inhibition of Pannexin channels attenuates ATP release and migration in activated microglial cells
PSM02-01 Kristen Stout Chronic methamphetamine alters dopamine release in the ventral pallidum PSM02-02 Lauren DePoy Mechanisms and reversal of adolescent cocaine-induced habits
PSM03 Neuroinflammation PSM03-01 Andrew Mendiola Fractalkine signaling in microglia is neuroprotective in the diabetic retina following lipopolysaccharide-induced activation PSM03-02 Narayan Bhat Myeloid cell-specific p38 MAP kinase deficiency suppresses LPS-induced amyloid deposition in a mouse model of AD PSM03-03 Zhihong Chen The Potential Role of Activated Microglia in Epilepsy
PSM01-11 Lauren Shapiro The ROCKII inhibitor fasudil activates TrkB-mediated signaling in the PFC and has antidepressant-like efficacy in adolescent mice
PSM03-04 Jamie Church Altered immune signaling impairs oligodendrocyte lineage cell responses and functional recovery after spinal cord injury
PSM01-12 Maria Sanchez Sonic Hedgehog Responsive Cells Contribute to Postnatal Myelination Patterns in the Telencephalon
PSM03-05 April Cox Nanoparticle Estrogen Elicits Rapid Modulation of Inflammation in Spinal Cord Injury
PSM03-12 George Kannarkat Loss of Regulator of G-protein Signaling-10 (RGS10) predisposes to neuroinflammation through alteration of immune cell chemotaxis PSM03-13 Gregory Konat Peripheral challenge with a viral mimic upregulates hippocampal expression of the complement genes
Poster Session I
PSM01-02 Alexandra Marsillo Rescuing Hippocampal Development and Behavioral Deficits in Fmr1 (-/-) Mice by Activating PKC Epsilon
PSM01-14 Pablo Paez Golli myelin basic proteins modulate voltage-operated Ca++ uptake and development in cortical and hippocampal neurons
PSM03-14 Evan Goldstein Intraspinal TLR4 activation promotes iron storage and rescues iron-mediated neuron death PSM03-15 Heping Zhou Effects of free fatty acids on microglial cells PSM03-16 James Stoll Lipocalin-2 induction following pilocarpine-induced seizures and in a cell culture neuroinflammatory model PSM03-17 Kirsten Evonuk System xc- transporter modulates CNS infiltration of immune cells and reactive gliosis in autoimmune inflammatory disease PSM03-18 Courtney Rella Specialized pro-resolving mediators promote recovery from neuroinflammation and improve behavioral outcome resulting from TBI
11
Poster Session I NO PHOTOGRAPHY ALLOWED OF POSTERS
Authors Present Sunday: 5:30—7:00pm / Monday: 9:30—10:00am
Sunday - Monday PSM03-19 Sanjib Karki Aggregation-dependent activation of innate immune pathways by the amyloid-! protein
PSM04-06 Miguel Escalante Lopez Glutamate-dependent translational control in HepG2 cells: Involvement of glutamate transporters
PSM07-04 Steven Barger Toward a Better a Model of the Energy Metabolism Derangements in Alzheimer’s Disease
PSM03-20 Grace Sun Botanicals regulating redox and inflammatory homeostasis in microglial cells
PSM04-07 Ming-Shuo Chen Nectin-like 2 (Necl-2) cell adhesion molecule is a negative regulator of Schwann cell proliferation and myelination
PSM07-05 Bethany Wilson Development of a high throughput pharmacological and toxicological assay for vesicular dopamine transport
PSM04-08 Nina Vardjan Morphological changes and the dynamics of !-Adrenergic/cAMP signaling in astrocytes in culture
PSM07-06 Carina Weissmann A FRET-based approach suggests allosteric activation of Mixed Lineage Kinases by mutant huntingtin
PSM04-09 Robert Zorec Astrocytic Vesicles: Diameter-dependent mode of exocytosis is independent of functional integrity of SNARE proteins
PSM07-07 Ching-Chieh Chou TDP-43-associated protein modulates TDP-43 toxicity
Poster Session I
PSM03-21 Seema Yousuf Progesterone is Neuroprotective in Stroke-Prone Spontaneously Hypertensive Rats PSM03-22 Veronica Cheli Participation of L-type calcium channels in astrocytes reactivity PSM03-23 Yu-Yo Sun Blood-borne monocytes convert to microglia in rodent neonates only after brain injury PSM03-24 Christopher McPherson Association of C1q and microglia activation phenotype with loss of hippocampal mossy fiber synapses PSM03-25 Gordon Meares PERK modulates ER Stress-induced Neuroinflammation PSM03-26 Yelena Grinberg A Th1 biased autoimmune response against a non-functional astrocyte antigen causes progressive epilepsy
PSM04 Neuron-Glial Interactions 1: Metabolism, Signal Transduction and Axon Biology PSM04-01 Corey Heffernan Nectin-like 4 (Necl-4) cell adhesion molecule regulates transmembrane choline transport through Choline Transporter-Like 1 (CTL1) PSM04-02 Shinghua Ding Imaging of mitochondrial Ca2+ dynamics in astrocytes using cell-specific mitochondria-targeted GCaMP5G/6s PSM04-03 Hyosung Kim Wnt/PCP pathway regulates myelination of the PNS PSM04-04 Akihiro Ishii Role of ERK1/2 MAPK Signaling in the Maintenance of Myelin and Axonal Integrity in the Adult CNS PSM04-05 Alain Guillem Del Angel Methylphenidate regulates glutamate uptake in Bergmann Glial Cells
12
PSM04-10 Robert Zorec Sphingolipids modulate calcium homeostasis in cultured rat astrocytes PSM04-11 Dan Song Chronic treatment with anti-bipolar drugs suppresses glutamate release from astrocytes PSM04-12 Thad Rosenberger Acetate treatment modulates adenosinergic enzyme and receptor levels in microglia and astrocyte cultures
PSM06 Axon Biology and Pathobiology PSM06-01 Minsu Kang Fast axonal transport deficits induced by mutant Huntingtin involve activation of a specific MAPK pathway PSM06-02 Nichole Mesnard-Hoaglin Axonal outgrowth rates of primary neurons using custom mutlicompartment microfluidic chamber system
PSM07 Neurodegeneration 1: AD, PD, HD, ALS
PSM07-08 David Braun Neurotrophins, Noradrenaline and Neuroinflammation PSM07-09 Darcie Cook The role of LRRK2 in inflammaging and Parkinson’s Disease PSM07-10 David Hartmann The Influence of Type 2 Diabetes on the Cerebral Microvasculature of Transgenic Alzheimer’s Disease Mice PSM07-11 Isaac Clements Multiwell optogenetic stimulation for high-throughput disease modeling and drug discovery PSM07-12 Angela Jeong Prenyltransferases as potential therapeutic targets for Alzheimer’s Disease PSM07-13 Jeremy Herskowitz Drug Inhibition of ROCK2 Suppresses Amyloid-! Production in an Alzheimer’s Disease Mouse Model PSM07-14 Kathryn MacPherson Inhibition of soluble TNF signaling with XPro1595 modulates CNS and peripheral immune cell populations in Alzheimer’s mouse model
PSM07-01 Amarallys Cintron Cargo Transport from Periphery to Brain by Circulating Monocytes
PSM07-15 Laura Butkovich Characterization of a transgenic mouse model with Parkinson’s disease-like "-synuclein aggregation in noradrenergic regions
PSM07-02 Amy Dunn The role of the synaptic vesicle glycoprotein 2C (SV2C) in Parkinson's disease
PSM07-16 Lee-Way Jin TDP-43 forms neurotoxic oligomers in frontotemporal lobar degeneration-TDP
PSM07-03 Asgar Zaheer Absence of glia maturation factor protects dopaminergic neurons and improves motor behavior in mouse model of Parkinsonism
PSM07-19 Nicholas Seyfried Prion-like aggregation properties of the small nuclear ribonucleoprotein U1-70K in Alzheimer's Disease Continued On Next Page
Poster Session I NO PHOTOGRAPHY ALLOWED OF POSTERS
Authors Present Sunday: 5:30—7:00pm / Monday: 9:30—10:00am
Sunday - Monday PSM07-20 Paul Donlin-Asp SMA motor neurons show impaired mRNP complex assembly PSM07-21 Rawand Chabayta Small Rho GTPases and Alzheimer’s Disease Proteins: Amyloid Precursor Protein (APP), Beta Amyloid (A!), and Tau
PSM07-22 Michael Dinkins Immunization against ceramide increases amyloid plaque burden in the 5XFAD mouse model of Alzheimer’s disease
PSM07-25 Ved Chauhan Effect of Trichostatin A-induced gelsolin in the brain of transgenic mouse model of Alzheimer's disease
PSM07-24 Thomas Westergard Arginine-rich sense and antisense dipeptides in post-mortem human tissues of C9ORF72-ALS/FTD patients
Abcam Andor Association Book Exhibit Bio-Techne / Tocris & Systems R&D BioLegend Biomedical Solutions Inc.
Poster Session I
ASN Welcomes the Following Exhibitors To the 46 th Annual Meeting:
BioRad International Society for Neurochemistry LC Sciences MBF Bioscience Mesoscale Miltenyi Biotec Nikon Instruments Noldus Information Technology ThorLabs Inc University of Georgia Wiley Wilkins Parkinson’s Foundation
13
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
Tu e s d a y, M a r c h 1 7 , 2 01 5 7:00 am – 5:00 pm 8:00 am – 8:15 am
ASN Registration Desk Open General Session • Marian Kies Award Winner: Joshua Burda Presented by Seema Tiwari-Woodruff PL03-Plenary Speaker Barbara Hempstead, PhD BDNF Proteins: Three Forms, Many Functions
Regency Foyer Regency VI
9:15 am – 10:00 am
Poster Session II/Coffee Break
Regency VII
10:00 am – 12:00 pm
ASN Scientific Sessions
8:15 am – 9:15 am
Regency V Symposium S17
Tuesday
Stress Organelles and Neurodegenerative Diseases Chair: Zixu Mao Co-Chair: Michael Lee S17-01 Leonard Petrucelli INHIBITING HISTONE DEACETYLASE 6 AS A THERAPEUTIC STRATEGY FOR TAUOPATHIES S17-02 Michael Lee ROLE OF C-ABL AND P53 IN AUTOPHAGIC DEFECTS ASSOCIATED WITH ALPHA-SYNUCLEINOPATHY IN VIVO S17-03 Zixu Mao REGULATION AND FUNCTION OF NEURONAL SURVIVAL FACTOR BY MULTIPLE STRESS ORGANELLES IN PARKINSON’S DISEASE S17-04 Charleen Chu MITOCHONDRIAL HOMEOSTASIS AND DENDRITIC STABILIZATION: BEYOND MITOPHAGY
12:00 pm – 1:00 pm
Regency VI Symposium S18
Cys-Loop Receptors in Neurotransmission: Structural Aspects of Regulation by Exogenous and Endogenous Compounds Chair: Victor Tsetlin Co-Chair: Jerrel Yakel S18-01 Margot Ernst GABA-A RECEPTOR SUBTYPES - RECENT DEVELOPMENTS IN PHARMACOLOGY S18-02 Victor Tsetlin INTERACTION OF NICOTINIC RECEPTORS WITH THREE-FINGER NEUROTOXINS AND LY6 PROTEINS S18-03 Jerrel Yakel PRESYNAPTIC "7 NICOTINIC ACH RECEPTORS ENHANCE HIPPOCAMPAL MOSSY FIBER GLUTAMATERGIC TRANSMISSION VIA PKA ACTIVATION
Hanover C, D Symposium S19
Mechanisms of Cell Death in Inflammatory Processes Chair: Malú Tansey S19-01 Malú Tansey NEUROPROTECTIVE EFFECTS VIA PERIPHERAL ADMINISTRATION OF XPRO1595 TO BLOCK SOLUBLE TNF SIGNALING IN A RAT MODEL OF PD S19-02 William Kaiser CELL DEATH PATHWAYS IN HOST DEFENSE, INFLAMMATION, AND DEVELOPMENTAL FAILURE S19-03 Christopher Norris TARGETING ASTROCYTE SIGNALING PATHWAYS PROTECTS HIPPOCAMPAL SYNAPTIC FUNCTION FOLLOWING TRAUMATIC BRAIN INJURY
S18-04 Pierre-Jean Corringer BACTERIAL AND BACTERIAL-HUMAN CHIMERAS AS MODELS TO STUDY THE MOLECULAR MECHANISMS GOVERNING PENTAMERIC LIGAND-GATED ION CHANNELS
Lunch with Plenary Speaker - Dr. Hempstead Chicago C, D Sponsored by: International Society for Neurochemistry Ticket Required
2 015 CAENLA Young Latin American Scholarships This Award is co-sponsored by the ISN and ASN to promising young neuroscientists from Latin America to attend the ASN Meeting and to visit a U.S. Laboratory for one week.
14
Anabela Palandri Instituto de Investigación Médica Mercedes y Martin Ferreyra
46 th Annual Meeting Atlanta, Georgia – 2015
Tu e s d a y, M a r c h 1 7 , 2 01 5 1:00 pm – 3:00 pm
ASN Scientific Sessions
Regency V Symposium S20
Neuron-Oligodendrocyte Interactions in Development and Disease Chair: Vittorio Gallo Co-Chair: David Pleasure S20-01 Bruce Appel ACTIVITY-BIASED SELECTION OF AXONS FOR MYELINATION IN VIVO
S20-03 David Pleasure IMMUNE-MEDIATED DEMYELINATION AND NEURONOPATHY IN AN AUTOIMMUNE MULTIPLE SCLEROSIS MODEL S20-04 Jeff Rothstein OLIGODENDROCYTE SUPPORT OF NEURONS AS A BASIS FOR NEURODEGENERATION INITIATION
Symposium S21
Glia Amino Acid Transporters in Health and Disease Chair: Arturo Ortega S21-01 Georgi Gegelashvili FUNCTIONAL CROSS-TALK BETWEEN DIFFERENT SYSTEMS OF GLUTAMATE TRANSPORT AND METABOLISM IN THE SPINAL CORD S21-02 Farrukh Chaudry THE SLC38 FAMILY OF GLUTAMINE TRANSPORTERS AND THEIR CONTRIBUTION TO THE GLUTAMATE/GABA-GLUTAMINE CYCLE
S22-02 Yoshiyuki Tamada ISCHEMIA ACTIVATES CALPAINS IN EXPERIMENTAL RETINAL NEUROPATHIES
S21-04 Michael Aschner GPR30 REGULATES GLUTAMATE TRANSPORTER GLT-1 EXPRESSION IN RAT PRIMARY ASTROCYTESITLE
S22-04 Isaac Donkor CALPAIN INHIBITORS: A SURVEY OF COMPOUNDS IN THE PATENT AND SCIENTIFIC LITERATURE
3:30 pm – 5:30 pm
ASN Scientific Sessions
Stroke and Mitochondria Chair: Selva Baltan Co-Chair: Richard Morrison
S22-01 Kathryn Saatman EFFECTS OF CALPAIN INHIBITION IN TRAUMATIC BRAIN INJURY AND AXONAL DEGENERATION
S22-03 Supriti Samantaray CHRONIC INTERMITTENT ETHANOLINDUCED AXON AND MYELIN DEGENERATION IS ATTENUATED BY CALPAIN INHIBITION
Coffee Break
Colloquium C03
Calpain Inhibitors in Preclinical Models of Neurodegenertion and Neurotrauma Chair: Naren Banik Co-Chair: Supriti Samantaray
S21-03 Arturo Ortega GLAST-DEPENDENT CONTROL OF THE GLUTAMATE/GLUTAMINE SHUTTLE: MOLECULAR TARGETS OF POLLUTANTS
3:00 pm – 3:30 pm
Regency V
Symposium S22
Terrace/Regency Foyer
Hanover C, D
Regency VI Symposium S23
Inflammation and Myelination: A Match Made in Heaven? Chair: Dana McTigue
Symposium S24
C03-01 Richard Morrison LOSS OF BAX-INTERACTING FACTOR-1 INCREASES NEURONAL SENSITIVITY TO ISCHEMIC INJURY
S23-01 Margot Mayer-Proschel LATENT HERPESVIRUS 6 INFECTION IMPAIRS HUMAN GLIAL PROGENITOR CELL FUNCTIONS IN VITRO AND IN VIVO
R. Wayne Albers Memorial Symposium Modulaton of ABC Transporters and APOE Levels as Therapeutic Targets for Alzheimers Chair: Danny Michaelson Co-Chair: Mary Jo LaDu
C03-02 Ludmila Voloboueva ROLE OF MITOCHONDRIA IN POST STROKE NEUROGENESIS AND INFLAMMATION
S23-02 Voon Wee Yong HARNESSING THE BENEFITS OF INFLAMMATION FOR REMYELINATION
S24-01 John Parks KEY FUNCTIONS OF ABCA1 IN MULTIPLE TISSUES
C03-03 Selva Baltan MITOCHONDRIAL DYNAMICS IN WHITE MATTER STROKE
S23-03 Jianrong Li GLIAL INTERACTIONS IN INFLAMMATORY DEMYELINATION AND MYELIN REPAIR: ROLE OF GALECTIN-9
S24-02 Cheryl Wellington THE THERAPEUTIC POTENTIAL OF ABCA1 FOR ALZHEIMER DISEASE
C03-04 Shinghua Ding ROLE OF PBEF IN MITOCHONDRIAL FUNCTION AND BIOGENESIS AFTER ISCHEMIA
S23-04 Dana McTigue LEVERAGING MACROPHAGE ACTIVATION TO PROMOTE OLIGODENDROCYTE GENESIS
5:30 pm – 7:00 pm
Tuesday
S20-02 Vittorio Gallo NEONATAL BRAIN INJURY CAUSES ABNORMALITIES IN NEURON-NG2 CELL SYNAPTIC COMMUNICATION
Hanover C, D
Regency VI
S24-03 Mary Jo LaDu APOLIPOPROTEIN E LIPIDATION AS A MECHANISTIC THERAPEUTIC TARGET FOR LOWERING SOLUBLE AMYLOID-! LEVELS IN ALZHEIMER’S DISEASE S24-04 Daniel Michaelson ABCA1 DIRECTED TREATMENT OF APOE4 DRIVEN NEURODEGENERATION IN ALZHEIMER’S DISEASE
Poster Session II - Authors Present/Wine & Cheese
Regency VII
15
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
We d n e s d a y, M a r c h 1 8 , 2 01 5 7:00 am – 5:00 pm
ASN Registration Desk Open
Regency Foyer
8:00 am – 9:15 am
General Session PL04-Basic Neurochemistry Textbook Lecture Scott Brady, PhD Life in the Fast Lane: Regulation of Fast Axonal Transport and Neurodegeneration
Regency VI
9:30 am – 10:00 am 10:00 am – 12:00 pm
Poster Session II/Coffee Break ASN Scientific Sessions
Regency VII
Regency V
Regency VI Oral Presentation 01
Wednesday
Symposium S25
Chair: Jeff Twiss
Neurodegeneration and Oxidative Stress Chair: Mohammed Akbar Co-Chair: Byoung-Joon Song
OP01-01 - Ji Na Kong REGULATION OF PRIMARY CILIA AND NEURONAL PROCESSES BY C24:1 CERAMIDE
S25-01 Rukhsana Sultana SYNAPSE LOSS: AN INITIAL EVENT IN THE PROGRESSION AND PATHOGENESIS OF ALZHEIMERS DISEASE
OP01-02 - Sparkle Williams INOSITOL PROTECTS ZEBRAFISH IN MODEL OF FETAL ALCOHOL SYNDROME
S25-02 Mohammed Akbar AN OVERVIEW ON THE MECHANISMS OF MITOCHONDRIAL DYSFUNCTION AND TISSUE INJURY IN NEURODEGENERATIVE DISEASES
OP01-03 - Megan Allen SELECTIVE REGULATION OF CDK5 ACTIVATORS BY HUD CONTROLS THE DEVELOPMENT AND FUNCTION OF HIPPOCAMPAL NEURAL CIRCUITRY
S25-03 Mohamed-Essa Musthafa DIETARY SUPPLEMENTATION OF FRUITS ALLEVIATE OXIDATIVE STRESS AND INFLAMMATION IN ALZHEIMER'S DISEASE TRANSGENIC MICE MODEL
OP01-04 - Muralidhar Hegde ROLE OF RNA/DNA BINDING PROTEINS, TDP-43 AND FUS IN DNA DAMAGE RESPONSE: ETIOLOGICAL LINKAGE TO AMYOTROPHIC LATERAL SCLEROSIS
S25-04 James O'Callaghan EARLY ACTIVATION OF STAT3 REGULATES REACTIVE ASTROGLIOSIS INDUCED BY DIVERSE FORMS OF NEUROTOXICITY S25-05 Bonghee Lee INDUCTION OF NEURONAL DEATH BY MICROGLIAL AGE-ALBUMIN: COMMON IMPLICATIONS FOR NEURODEGENERATIVE DISEASES
16
Hanover C, D Symposium S26 CDK5 Signaling: A Double-edged Sword in Normal and Diseased Brain Chair: Yue Feng S26-01 Ashok Kulkami IMPORTANT ROLE OF CDK5 IN PAIN MODULATION S26-02 James Bibb INTEGRATION OF EXCITATORY AND METABOTROPIC SIGNAL TRANSDUCTION S26-03 Yue Feng CONTROLLING CDK5 FUNCTION IN NEURONAL AND GLIAL LINEAGES S26-04 Dianbo Qu PHYSIOLOGICAL AND PATHOLOGICAL CONDITIONS ALTER THE ROLE OF PHOSPHORYLATED JIP1 BY CDK5
OP01-05 - Anabela Palandri MYELIN-ASSOCIATED GLYCOPROTEIN MODULATES PROGRAMED CELL DEATH OF MOTONEURONS VIA NGR/P75NTR RECEPTOR-MEDIATED RHOA ACTIVATION OP01-06 - Rodolfo Gatto ADDRESSING THE IN VIVO CONTRIBUTION OF JNK3 TO HUNTINGTON’S DISEASE PATHOGENESIS
12:00 pm – 1:00 pm
Lunch with Plenary Speaker - Dr. Brady Chicago C, D Sponsored by: International Society for Neurochemistry Ticket Required
12:00 pm – 3:00 pm
ASN Council Meeting II
Hanover A, B
46 th Annual Meeting Atlanta, Georgia – 2015
We d n e s d a y, M a r c h 1 8 , 2 01 5 1:00 pm – 3:00 pm
ASN Scientific Sessions
Regency V
Regency VI Oral Presentation 02
Symposium S27
Chair: Bruce Carter
Sex-Specific Effects of Adolescent Drugs and Stressors on Neural, Metabolic, Immune and Behavioral Outcomes Chair: Kyle Frantz Co-Chair: Gretchen Neigh
3:00 pm – 3:30 pm
Poster Session II/Coffee Break
3:30 pm – 5:30 pm
ASN Scientific Sessions
Regency V
Regency VI Symposium S30
Symposium S29
Pharmacologically Induced Hypothermia and Clinical Implications Chair: Shan Ping Yu Co-Chair: Thomas A. Dix
Neuroinflammation and Immune Responses in Autoimmune Demyelination Diseases Chair: Jae Kyung Lee Co-Chair: Kareem Graham S29-01 Katerina Akassoglou ACTIVATION OF INNATE IMMUNITY BY BLOOD-CLOTTING FACTORS: MECHANISMS, IMAGING, THERAPEUTICS S29-02 Kareem Graham REGULATION OF CNS INFLAMMATION BY A NORMAL LIPID-METABOLIZING ENZYME S29-03 Jae-Kyung Lee THE NOVEL FUNCTION OF RGS10 IN EFFECTOR T LYMPHOCYTES TO AUGMENT MOUSE EAE S29-04 Brian Evavold FREQUENCY AND AFFINITY OF T CELLS IN THE CNS DETERMINES EXTENT OF NEUROINFLAMMATION
7:30 pm – 11:00 pm
S30-01 Shan Yu NEUROTENSIN RECEPTOR 1 AGONISTS INDUCED HYPOTHERMIA FOR THE TREATMENT OF STROKE AND TBI S30-02 Kelly Drew TRANSLATING DRUG-INDUCED HIBERNATION TO THERAPEUTIC HIBERNATION S30-03 Sean Marrelli DO CHILI PEPPERS PROVIDE THE SPICE OF LIFE? HOW TRPV1 AGONISM PROMOTES REVERSIBLE HYPOTHERMIA AND NEUROPROTECTION FOLLOWING STROKE S30-04 Flemming Fryd Johansen MULTI-TARGET TREATMENT IN ACUTE ISCHEMIC STROKE
ASN Closing Party
Symposium S28 Contribution of Environmental Chemicals to Neurodegenerative Disease Chair: Mike Caudle S28-01 Michael Aschner SLC30A10 IS A CELL SURFACE-LOCALIZED MANGANESE EFFLUX TRANSPORTER S28-02 Paul Cox THE BMAA/CYANOBACTERIAL THEORY AND ALS/PDC IN GUAM S28-03 Jason Richardson MECHANISMS OF GENE-ENVIRONMENT INTERACTIONS IN ALZHEIMER DISEASE S28-04 William Caudle THE STRIATAL SYNAPSE AS A TARGET FOR DAMAGE BY FLAME RETARDANT COMPOUNDS
Wednesday
S27-01 Bradley Cooke SEX DIFFERENCES IN THE AMYGDALA AS A RISK FACTOR FOR MOOD DISORDERS S27-02 Shannon Gourley INCUBATION AND REVERSAL OF ADOLESCENT COCAINE-INDUCED HABITS S27-03 Gretchen Neigh STRESS AND PUBERTY: THE PERFECT STORM OR A CHANGE OF COURSE? S27-04 Kyle Frantz ADOLESCENT (IN)VULNERABILITY TO ENDURING EFFECTS OF DRUGS OF ABUSE
OP02-01 - Allan MacKenzie-Graham VISUALIZING THE NEUROPATHOLOGY UNDERLYING GRAY MATTER ATROPHY IN EAE USING CLARITY OP02-02- Elizabeth Gould MYELIN DISRUPTION LEADS TO TARGETED BEHAVIORAL DEFICITS OP02-03 - Grace Hobson INFLAMMATION AND MYELIN DEGENERATION IN THE PLP1DUP MOUSE MODEL OF PELIZAEUS-MERZBACHER DISEASE OP02-04 - Whitney Lee UNIQUE FUNCTIONS OF MYELIN PROTEOLIPID PROTEIN IN MYELIN AND IN MITOCHONDRIA DEFINED WITH A TRANSGENIC MOUSE OP02-05 - Michael Klingener ADAM10 IN THE DEMYELINATED CENTRAL NERVOUS SYSTEM OP02-06 - Tong Luo ROLE OF GLUN2C-CONTAINING NMDA RECEPTORS IN OLIGODENDROCYTE MATURATION AND MYELINATION
Hanover C, D
Regency VII
Hanover C, D Symposium S31
Revisit Mitochondria in Neurodegeneration Chair: Wenzhen Duan Co-Chair: Xin Qi S31-01 Hiroko Yano IMPAIRED MITOCHONDRIAL PROTEIN IMPORT IN NEURODEGENERATIVE DISEASES S31-02 Richard Morrison LOSS OF BAX-INTERACTING FACTOR-1 EXACERBATES ALZHEIMER’S DISEASE PATHOLOGY S31-03 Xin Qi REGULATION OF MITOCHONDRIAL FISSION IN HUNTINGTON’S DISEASE S31-04 Wenzhen Duan MITOCHONDRIAL PROTEIN DEACETYLATION AND NEURODEGENERATION
Regency VII
17
Poster Session I I NO PHOTOGRAPHY ALLOWED OF POSTERS
Authors Present Tuesday: 5:30—7:00pm / Wednesday: 9:30—10:00am
Tu e s d a y - We d n e s d a y PTW01 Neuron-Glial Interactions 2: Development and Disease PTW01-01 Abdullah Madany TREM2 deficient microglia and macrophages display decreased phagocytosis without altering TAM receptor expression
Poster Session I I
PTW01-02 Bruno López Bayghen PKC Activation Leads to Notch Pathway Induced Differentiation in Bergmann Glial Cells: A Role for Glutamate? PTW01-03 Kareem Clark Axon Initial Segment Disruption in Multiple Sclerosis and EAE PTW01-04 Esther Lopez-Bayghen Glutamate sensitive-expression of adhesion molecules Class-1 MHCrestricted T-cell associated molecule (CRTAM) and Nectin-like 2 PTW01-06 Edna Suárez Glutamatergic disorders associated to fluoride or particulate matter PM10 and PM2.5 exposure PTW01-07 Isis Carletti mTOR and BMP Pathway interaction during oligodendrocyte differentiation PTW01-08 Juhi Singh Role of Rho associated protein kinase in the Platelet derived growth factor-A induced Oligodendrocyte progenitor cell migration PTW01-09 Lee-Way Jin Bioenergetic mechanisms of microglial dysfunction in Rett syndrome
PTW01-14 Catherine Fressinaud Neurofilaments penetrate into oligodendrocytes via clathrin-dependent endocytosis to promote their growth and survival in vitro
PTW02 Neurodegeneration 2: Ischemia, Trauma and Other PTW02-01 Zeynep Akgoc The restricted ketogenic diet reduces brain invasion and metastasis of the VM-M3 murine glioblastoma PTW02-02 Alan Hazell Treatment of rats with Fedratinib does not lead to experimental Wernicke's encephalopathy PTW02-03 Alicia Hawthorne DRGs haploinsufficient for mRNA binding protein IMP1 show increased regenerative properties and changes in the PTEN/mTOR pathway PTW02-04 Andrew Campbell Loss of ATM does not alter oxidative state but results in increased neuronal survival following DNA damage PTW02-05 Narendra Banik Calpain as a Therapeutic Target in EAE PTW02-06 Justin Brooks Altered inhibitory circuitry leads to seizure susceptibility in mice chronically infected by Type II Toxoplasma gondii PTW02-07 Debapriya Garabadu Risperidone and paroxetine attenuates anxiety and apoptosis in an animal model of post-traumatic stress disorder (PTSD)
PTW01-10 Melanie Pleiss Proteolysis of calcineurin in human brain PTW02-08 Geoffrey Pronovost tissue appears in astrocytes surrounding Reduction in gliosis and brain hemispheric swelling in TREM2-deficient mice amyloid deposits and microinfarcts following traumatic brain injury PTW01-11 Pradoldej Sompol
18
Inhibition of the astrocytic calcineurin/NFAT pathway quells glutamatergic hyperactivity in a mouse model of Alzheimer’s disease
PTW02-09 Haley Titus-Mitchell Nf1 loss in Oligodendrocytes Induces Myelin Decompaction through Notch Signaling and Nitric Oxide
PTW01-12 Richa Hanamsagar Sex differences in developmental gene expression in hippocampal microglia of mice: Relevance for neurodevelopmental disorders
PTW02-10 Steven Levison LIF Haplodeficiency Desynchronizes Glial Reactivity Prolonging Damage and Functional Deficits After a Concussive Brain Injury
PTW01-13 Yiting Liu The effect of antibody and complementdependent cytotoxicity on neuronal-glial network in neuromyelitis optica
PTW02-11 Saurav Bhowmick Neuroprotective role of neuroglobin via targeting nitric oxide signaling pathway
PTW02-12 David Calderon Role of oseltamivir in novel dyskinesia young model: to use or not to use PTW02-13 Tibor Kristian Ischemia-induced cell-type specific alterations in brain mitochondrial homeostasis PTW02-14 Rachel Bennett Assessment of Mass Spectrometry Imaging by DESI and MALDI to Visualize Changes in Lipid Patterns Due to Traumatic Brain Injury
PTW03 Neuroprotection and Repair PTW03-03 Christopher Theisen SIRT3/NAMPT dependence on neuronal cell survival PTW03-04 Guanghu Wang Low dose Hsp90 inhibitor 17AAG protects neural progenitor cells from ischemia induced death PTW03-05 Justin Siebert Pattern of Oligodendrocyte Progenitor Cell Migration Following a Spinal Contusion Injury PTW03-06 James Walsh TGF! potentiates retinal ganglion cell death after optic nerve injury through action on glial cells PTW03-07 Kelly Lohr Increased vesicular function improves synaptic dopamine handling and opposes neurotoxicity PTW03-08 Mauricio Cunha Creatine affords protection against glutamate-induced nitrosative and oxidative stress in the neuroblastoma SH-SY5Y cell line PTW03-09 Poonam Jaiswal-Sharma DREADD mediated increases in excitability of targeted sciatic neurons in vivo PTW03-10 Evgeniia Pushchina Neural regeneration in salmon model of adult brain injury
PTW04 Demyelination: Pathology, Protection and Repair PTW04-02 Aminat Saliu Mammalian Target of Rapamycin (mTOR) - mediated mechanisms underlying Oligodendrocyte process extension
Poster Session I I NO PHOTOGRAPHY ALLOWED OF POSTERS
Authors Present Tuesday: 5:30—7:00pm / Wednesday: 9:30—10:00am
Tu e s d a y - We d n e s d a y PTW04-03 Bernard Zalc Remyelination by resident oligodendrocyte precursor cells in a Xenopus laevis inducible model of demyelination PTW04-04 Douglas Feinstein Lanthionine ketimine ester (LKE) reduces clinical signs and neuronal degeneration in EAE
PTW04-06 Judith Grinspan Antiretroviral Compounds Differentially Alter Oligodendrocyte Maturation PTW04-07 Megan Vignos Cerebral white matter lesions absent in Myelocortical Multiple Sclerosis PTW04-08 Hongwei Qin Preferential Recruitment of Neutrophils into the Cerebellum and Brainstem Contributes to the Atypical EAE Phenotype PTW04-09 Javier Palazuelos A role for TACE/ADAM17 during CNS myelination and remyelination PTW04-10 Joyce Benjamins Signaling Pathways Utilized by ACTH1-39 to Protect Oligodendroglia Vary Depending on the Type of Insult PTW04-11 Juan Palavicini Sulfatide depletion leads to a dramatic age-dependent reduction of myelin lipid content PTW04-12 Lauren McLane Functions of TSC/mTOR signaling in oligodendrocyte differentiation and remyelination in a focal demyelinated lesion PTW04-14 ShiPing Zou HIV-1 Tat affects the viability of immature and mature oligodendrocytes via Ca2+ dysregulation and GSK3! activation
PTW05 Gene Expression/Regulation PTW05-01 Esther Lopez-Bayghen Neuronal Growth Factor regulates Brain Specific Kinase 1 expression by modulating promoter methylation and SP1 recruitment PTW05-02 Cynthia Gomes Neuritin/Cpg15 and Gap-43 mRNA Compete for Axonal Localization
PTW07-03 Matthew Kanzler Regulation of Seizure Threshold by Sex and L-12/15 Lipoxygenase in the Pentylenetetrazol Model of Temporal Lobe Epilepsy
PTW05-04 Jason Hinman Transcriptional profiling of regionally distinct oligodendrocyte populations reveals unique biology in cortical oligodendrocytes
PTW07-04 Scott Hogan High Resolution Rat Serum Lipidomics for the Detection of Mild Traumatic Brain Injury
PTW05-05 Kathryn Williams Repression of GAP-43 expression by hnRNP-Q1: uncovering a potential local mechanism to regulate neuronal development PTW05-06 Mandakh Bekhbat The influences of ovarian hormones on co-chaperones of the glucocorticoid receptor PTW05-07 Patricia Wight Control of Human PLP1 Expression through Transcriptional Regulatory Elements and Alternatively Spliced Exons in Intron 1 PTW05-08 Sandra Hewett Regulation of Constitutive Neuronal Cyclooxygenase-2 Expression
PTW06 The Synapse: Signals and Plasticity PTW06-01 Amelia Burch The Dysbindin Schizophrenia Susceptibility Network Interacts with the Actin Polymerization Machinery PTW06-02 Bart Anderson FMRP-Regulated Association of Specific miRNAs with RISC PTW06-03 Georgina Rodriguez de Lores Arnaiz Sodium-potassium atpase changes after administration of levocabastine, an antagonist for nts2 neurotensin receptor PTW06-04 Sharon Swanger GluN2D-specific NMDA receptor control of the subthalamic nucleus
PTW07 Lipids: Biology and Pathobiology PTW07-01 Erika Calvo-Ochoa Saturated fatty acids promotes insulin signaling alterations and mitochondrial dysfunction in the rat hippocampus and in vitro
PTW07-05 YUTAKA ITOKAZU Ganglioside expression and neural stem cell proliferation in an AD mouse model
PTW08 Late Breaking Abstracts PTW08-01 Iryna Ethell The role of ephrin-B1 signaling in astrocyte-mediated synapse pruning PTW08-02 Akshata Almad Elucidating the contribution of Cx43 astroglial gap junction protein and hemichannels in Amyotrophic Lateral Sclerosis PTW08-03 Nancy Ratner RAS signaling and no in oligodendrocytes modulate permeability of the blood brain barrier PTW08-04 Ki Ma Polycomb protein EED is required for the long term maintenance of peripheral nerve myelin PTW08-05 Frank Lee Evidence for a role of BLOC-1 in mouse brain development
Poster Session I I
PTW04-05 Ali Fatemi Activated Microglial Supernatant Inhibits Oligodendroglial Process Outgrowth and Myelin synthesis
PTW05-03 Joshua Burda Resolving disease-specific forms of reactive astrogliosis in vivo by cell-type specific analysis of actively translating mRNA
PTW08-06 Ella Doron-Mandel RNA transport and local translation in retrograde injury signaling in peripheral Vs. central axons of dorsal root ganglia neurons PTW08-07 Giordano Santos Thiol repletion therapy in animal and human studies of Parkinsons disease PTW08-08 Giordano Santos Guanosine exerts neuroprotector effect in a preclinical model of episodic hyperammonemia PTW08-09 Giordano Santos Neuronal markers, behavior and electrophysiological brain alterations in a model of partial hepatectomy in rats PTW08-10 Joyce Bilinang Morphine enhances HIV-1 effect on the proliferation of primary human NPCs; role of $-opioid receptor splice variants
19
ASN Officers & Council
ASN Officers & Council
Elected Officers
20
Steve Levison
Babette Fuss
Treasurer
Past President
President Elect
Erhard Bieberich
Ernesto Bongarzone
Cheryl Dreyfus
Jim Hewett
Eric Murphy
Michael Nichols
Juana Pasquini
Arturo Soto Ortega
Etty (Tika) Benveniste
Vlad Parpura
President
Secretary
Steve Barger
Dianna Hynds
Jean Harry
Council
Alternate Council
Colin Combs
Seema Tiwari-Woodruff
Emma Wilson
Secretary-Elect
Appointments
Sandra Hewett
Douglas Feinstein
George DeVries
Intersociety Liaison
Historian
Michael Fox ASN Business Manager
Sheilah Jewart
ASN 2015 Program & Host Committees Scientific Program Committee
Erhard Bieberich
Bruce Carter
Cheryl Dreyfus
Steve Levison
Wendy Macklin
Patrice Maurel
Mary McKenna
Helen Scharfman
Seema Tiwari-Woodruff
Jeff Twiss
Emma Wilson
Chair
Yue Feng
Kyle J. Frantz
Shelley Hooks
Jae-Kyung Lee
Anne Murphy
Gretchen Neigh
Michelle Olsen
Shan Ping Yu
Atlanta Host Committee
Malú Tansey
ASN Program & Host Committees
Chair
Wilma Friedman
21
ASN Committees Basic Neurochemistry - Editorial Board
Jordi Folch-Pi Award Committee
Scott T. Brady, Editor-in-Chief George Siegel R. Wayne Albers Donald Price Joyce Benjamins Stephen Fisher Nicolas Bazan Alison Hall Sangram Sisodia Joseph Coyle
Eric Murphy (Chair); 2008-2015 Xialin Han; 2010-2014 Gerardo Morfini; 2012-2016 Matthew Rasband; 2013-2015 Tom Seyfried; 2010-2014
ASN Committees
Bernard Haber Award Committee Nicolas Bazan (Chair) 2004-2016 Beatrice Capputo; 2004-2016 H. David Shine; 2008-2016
Committee for the Advancement & Encouragement of Neurochemistry in Latin America (CAENLA) Juana Pasquini, Argentina (Chair); 2007-2015 Ernesto Bongarzone; 2010-2014 Gustavo Pigino; 2010-2014 Francisco Nualart; 2010-2014 Carmen Sato-Bigbee; 2010-2014
Electronic Communication Committee Vlad Parpura (Chair); 2013-2015 Babette Fuss Jean Harry Sandra Hewett
ASN Journal Oversight and Publisher Liason Committee Steve Levison; 2013-2015 Etty (Tika) Benveniste; 2011-2017 Babette Fuss; 2013-2019 Monica Carson; (ASN NEURO Editor-in-Chief)
Finance Committee Jean Harry (Treasurer) Etty (Tika) Benveniste Steve Levison Babette Fuss Karen Chandross Vlad Parpura Sandra Hewett
22
Marian Kies Award Committee Seema Tiwari-Woodruff and Emma Wilson (Co-Chairs); 2008-2018 Astrid Cardona; 2014-2018 Dianna Hynds; 2010-2014 Jonathan Kipnis; 2013-2017
Membership Committee Shinghua Ding (Chair); 2013-2017 José Julio Rodriguez Arellano; 2012-2016 Selva Baltan; 2012-2016 Brian Gulbransen; 2014-2018 Alexej Verkhratsky; 2012-2016
Nominating Committee Douglas Feinstein (Chair); 2014-2015
Public Policy and Education Committee Michael Nichols (Chair); 2012-2015 Bonnie Dittel; 2012-2016 Tajie Harris; 2014-2018 Mark Kindy; 2012-2016 Thad Rosenberger; 2014-2018
Young Investigator Education Enhancement Committee Michael Fox (Chair); 2009-2015 Ben Deneen; 2014-2018 Debra Mayes; 2014-2018 Donna Osterhout; 2011-2015 Andre Phillips; 2013-2017
2016 Denver Scientific Program Committee Jony Kipnis (Chair)
2016 Denver Host Committee Wendy Macklin (Chair)
2 015 ASN Meeting Sponsors The American Society for Neurochemistry Acknowledges the Generous Support from the Following Sponsors MBF Bioscience
ACTSI
Mesoscale
Andor
Miltenyi Biotec, Inc.
ASN NEURO
NeuroSci Association
Assn Book Exhibit
NIH / NINDS
Atlanta Science Festival
Nikon
Avanti Lipids
National MS Society
Axion BioSystems
Noldus Information Technology
Bio-Techne / Tocris & R&D Systems
The Ohio State University Center for Brain & Spinal Cord Repair
BioLegend Biomedical Solutions Inc. BioRad Bioselec
Parkinson’s Association of Alabama Parkinsons Disease Coalition of the South
Center for Behavioral Neuroscience (GSU)
South Carolina SmartState Center for Childhood Neurotherapeutics
Cytoskeleton
ThermoFischer
Elsevier – Basic Neurochemistry Textbook
Thor Labs
Emory College Center for Science Education University of Alabama at Birmingham (UAB), Dean's Office HHV6 Society UAB Dept of Cell, Developmental & International Society for Neurochemistry Integrative Biology JT Pharmaceuticals University of Georgia Karger Publications UGA Vice-President for Research LC Sciences Wiley Leica
2015 ASN Meeting Sponsors
Abcam
Funding for this conference was made possible in part by Grant #1R13NS093218-01 from NIH/NINDS. The views expressed in written conference materials or publications and by speakers and moderators do not necessarily reflect the official policies of the NIH; nor does mention by trade names, commercial practices, or organizations imply endorsement by the U.S. Government.
23
AMERICAN SOCIETY FOR NEUROCHEMISTRY
Notes
The Latest in Molecular and Cellular Neurobiology
24
Front Cover B/W Photo: Michelle Olsen “Making Connections” Best of Show 2014
©2015 American Society for Neurochemistry
500
Basic Ba asic N Neurochemistry, eurochem mistryy, Eighth Edition Edittion Principles P rin nciples l off M Molecular, olecular l l , Cellular C Cellular, ll l , and dM Medical ed dicall N Neurobiology eurobiology b
THE CLASSIC TEXT FOR O OVER VER 40 YEARS! This outstanding book continues to be the global standard for information This on the biochemistry off the nervous system. It is the standard reference for neuroscience,, bringing basic and clinical exploring the translational nslational nature off neuroscience neurosc QHXURVFLHQFHWRJHWKHULQRQHDXWKRULWDWLYHYROXPH7KHVXEWLWOHUHȈHFWVWKH Q HXURVFLHQFHWRJHWKHULQRQHDXWKRULWDWLYHYROXPH7KHVXEWLWOHUHȈHFWVWKH expanded attention to the links between neurochemistry and neurologic G GLVHDVH$FRPSDQLRQVLWHZLWKGRZQORDGDEOHȇOHVRIȇJXUHVER[HVDQG LVHDVH$FRPSDQLRQVLWHZLWKGRZQORDGDEOHȇOHVRI ȇJXUHVER[HVDQG references are available ailable to purchasers off the book. Dec. 2011, Hardback, 1,120 pages ISBN: 9780123749475 Print P rint price: $125.00 Member ASN M ember price: $93.75 available.. ebook format also available
Edited by Scott T T.. Brady Brady,, George JJ.. Siegel, R. W Wayne ayne Albers Albers,, and Donald L. P Price rice © American Society for N Neurochemistry eurochemistry
ISN and ASN members save 25% Order online at store store.elsevier.com/9780123749475 .elsevier.com/9780123749475
Enter promotional code NEURO215 at checkout to receive receivve your discount. 20150119
Plan Now to Attend
47th Annual Meeting American Society for Neurochemistry
Denver, Colorado, March 19 – 23, 2016 Join us for four days of symposia, colloquia and workshops designed to address the four major themes of our Society: •
Building the Nervous System
•
Molecular and Cell Biology of the Nervous System
•
Glial Cell Biology
•
Neurodegeneration and Disease
On-line Abstract Submissions: Sept. 15 – Nov. 30, 2015 www.ASNeurochem.org
ASN 2016 – Grand Hyatt Denver Babette Fuss, ASN President
Wendy Macklin, Host Committee Chair Jony Kipnis, Scientific Program Chair
Make Your Plans Today! © 2014 American Society for Neurochemistry Background Photo: Michelle Olsen
STAY UPDATED WITH THE LATEST ADVANCES IN THE CELLULAR AND MOLECULAR NEUROSCIENCES!
IMPACT FACTOR: 4.436 Source: 2013 Journal Citation Reports® (Thomson Reuters, 2014)
EDITOR-IN-CHIEF: Monica Carson University of California, Riverside
Submit by June 30th to receive a conference discount!
$1950 $1550
asn.sagepub.com
ASN Neuro is an open access, peer-reviewed journal uniquely positioned to provide investigators with the most recent advances across the breadth of the cellular and molecular neurosciences. Article Processing Charge: $1550 with your submission by June 30th Use “ASN2015” at checkout Or become a member to receive the low rate of $999
Submit today at mc.manuscriptcentral.com/asnneuro
AMERICAN SOCIETY FOR N E U R O C H E M I S T RY
TRANSACTIONS OF THE AMERICAN SOCIETY FOR NEUROCHEMISTRY 46TH ANNUAL ASN MEETING
AT L A N TA , G E O R G I A MARCH 14 — 18, 2015
WELCOME
ASNeurochem.or g
AMERICAN SOCIETY FOR NEUROCHEMISTRY The Latest in Molecular and Cellular Neurobiology
Etty (Tika) Benveniste President
Vlad Parpura Secretary
Jean Harry Treasurer
Steven W. Levison Past President
Babette Fuss President-Elect
COUNCIL: Steven Barger
Dear Participants of the 46th ASN Annual Meeting: On behalf of the ASN Council, Program and Host Committees, we would like to welcome you to Atlanta, Georgia and the 46th Annual Meeting of the American Society for Neurochemistry. The scientific program has been designed to maximize your ability to benefit scientifically from this meeting, with four days of symposia and colloquia that address the four major themes of our Society: -Building the Nervous System -Glial Cell Biology -Molecular and Cell Biology of the Nervous System -Neurodegeneration and Disease
Erhard Bieberich Ernesto Bongarzone Cheryl Dreyfus James Hewett
Special efforts have been taken to encourage Young Investigator participation with a session on funding opportunities, lunch with the plenary speaker each day, travel awards and over 12 abstracts chosen for oral sessions, to provide another avenue of recognition. To maximize scientific interactions
Dianna Hynds
and discussions, posters will be up for 2 consecutive days, with a two hour
Eric Murphy
session for authors to be present. Thank you to all the volunteers, session
Michael Nichols
chairs and speakers for your contribution to the success of this meeting.
Juana Maria Pasquini Arturo Ortega
We look forward to seeing you next year in Denver, Colorado at the 47th ASN meeting.
Seema Tiwari-Woodruff Emma Wilson SECRETARY-ELECT: Sandra Hewett ALTERNATE COUNCIL: Colin Combs Michael Fox
Tika Benveniste
Wilma Friedman
Malú Tansey
President
Scientific Program Chair
Host Committee Chair
!
®
Council of the American Society for Neurochemistry The Latest in Molecular and Cellular Neurobiology
Elected Officers: Etty (Tika) Benveniste President
Vlad Parpura
Jean Harry
Steve Levison
Babette Fuss
Secretary
Treasurer
Past President
President Elect
Council: Steve Barger
Cheryl Dreyfus
Eric Murphy
Arturo Soto Ortega
Erhard Bieberich
Jim Hewett
Michael Nichols
Seema Tiwari-Woodruff
Ernesto Bongarzone
Dianna Hynds
Juana Pasquini
Emma Wilson
Alternate Council: Colin Combs
Secretary-Elect Sandra Hewett
Scientific Program Committee: Wilma Friedman, Chair Cheryl Dreyfus
Patrice Maurel
Seema Tiwari-Woodruff
Erhard Bieberich
Steve Levison
Mary McKenna
Jeff Twiss
Bruce Carter
Wendy Macklin
Helen Scharfman
Michael Fox Appointments: Intersociety Liaison: Douglas Feinstein
Historian: George DeVries
Atlanta Host Committee: Yue Feng Malu Tansey, Chair
Jae-Kyung Lee
Michelle Olsen
Kyle J. Frantz
Anne Murphy
Shan Ping Yu
Shelley Hooks
Gretchen Neigh
ASN Business Manager Sheilah Jewart American Society For Neurochemistry 9037 Ron Den Lane, Windermere, FL 34786 407-909-9064 Fax: 407-876-0750 www.ASNeurochem.org Cover B/W Photo: Michelle Olsen “Making Connections” Best of Show 2014
Contents Saturday, March 14th, 2015 08:30 am – 05.00 pm
Pre-Meeting Workshop PMW01
P Page a Optogenetics Tools, Techniques and Applications in Neuroscience 75 g and Neurochemistry (Chair: Ling Wei, Robert Gross) es Sunday, March 15th, 2015
08:15 am - 09:15 am
Plenary Lecture PL01
Looking at the Epigenomic Landscape of Developing Oligodendrocytes (Plenary Speaker: Patrizia Casaccia)
1
10:00 am - 12:00 pm
Symposium S01
microRNAs (miRNAs) in Alzheimer's disease (AD) and age-related macular degeneration (AMD) (Chair: Walter Lukiw, Debomoy Lahiri)
5
Colloquium C01
Proinflammatory Mechanisms in Neurodegeneration (Chair: Michael Nichols)
Symposium S02
Glial Vesicular, Metabolic and Morphologic Function in Health and Disease (Chair: Robert Zorec, Vladimir Parpura)
7
Symposium S03
RNA Toxicity in Neurodegeneration (Chair: Rita Sattler, Christopher Donnelly)
9
Symposium S04
Frontiers of Neurolipids (Chair: Stefka Spassieva, Glyn Dawson)
12
Symposium S05
Novel Strategies in Stem Cell Therapy and Clinical Potentials (Chair: Michael Chopp, Shan Ping Yu)
15
Symposium S06
Autophagy in Neurodegeneration: Role in Disease and Therapy Target (Chair: Kenneth Hensley, Marni Harris-White)
17
Symposium S07
Central and Peripheral Inflammatory and Immune Responses in Parkinson's Disease (Chair: Malú Tansey, Etty (Tika) Benveniste)
19
Symposium S08
Wiring of the Nervous System: From Molecular Mechanisms to Complex Behaviors (Chair: Tracy Tran)
21
01:00 pm – 03:00 pm
03:30 pm - 05:30 pm
68
Sunday/Monday Poster Sessions
05:30 pm - 07:00 pm
Poster Session PSM01 Poster Session PSM02 Poster Session PSM03 Poster Session PSM04 Poster Session PSM06 Poster Session PSM07
Development, Differentiation and Disorders Drugs of Abuse: Alcohol, Cocaine, Methamphetamine Neuroinflammation Neuron-Glial Interactions 1: Metabolism, Signal Transduction and Axon Biology Axon Biology and Pathobiology Neurodegeneration 1: AD, PD, HD, ALS
2015 Transactions of the American Society for Neurochemistry ®
83 89 90 100 105 106
I
Contents
Monday, March 16th, 2015
Page
08:15 am - 09:15 am
Plenary Lecture PL02
The Teen Brain: Insights from Neuroimaging (Plenary Speaker: Jay Giedd)
09:30 am - 10:00 am
Poster Sessions PSM01 to PSM07
Sunday/Monday Poster Sessions
83
10:00 am - 12:00 pm
Symposium S09
Astrocytes as Obligatory Partners in Purinergic and Glutamatergic Neurotransmission (Chair: Arne Schousboe, Vladimir Parpura)
23
Symposium S10
Neuropsychiatric Disorders: New Mechanisms and Models (Chair: Cristina Ghiani, Antonieta Lavin)
25
Symposium S11
Dissecting the Molecular Mechanisms of Acute Neurologic Injury (Chair: Teresa Wood)
27
Symposium S12
Signaling Pathways Regulating Remyelination (Chair: Carol Troy)
29
Colloquium C02
Post-translational Prenylation of GTPases in Neurodegeneration and Repair (Chair: Evan Stubbs, DiAnna Hynds)
70
Symposium S13
Mechanisms of Cell Death and Diseases (Chair: Elias Aizenman, Shan Ping Yu)
31
Symposium S14
Retromer and Neurodegenerative Diseases (Chair: Wen-Chen Xiong)
33
Symposium S15
Endoplasmic Reticulum Stress in Myelin Disorders (Chair: Wensheng Lin)
35
Symposium S16
Wiring Axons by mRNA Regulation in Neuronal Health and Disease (Chair: Jeffery Twiss, Gary Bassell)
37
01:00 pm - 03:00 pm
03:30 pm - 05:30 pm
2015 Transactions of the American Society for Neurochemistry ®
2
II
Contents
Tuesday, March 17th, 2015
Page
08:00 am - 09:15 am
Plenary Lecture PL03
BDNF Proteins: Three Forms, Many Functions (Plenary Speaker: Barbara Hempstead)
10:00 am - 12:00 pm
Symposium S17
Stress Organelles and Neurodegenerative Diseases (Chair: Zixu Mao, Michael Lee)
39
Symposium S18
Cys -loop Receptors in Neurotransmission: Structural Aspects of Regulation by Exogenous and Endogenous Compounds (Chair: Victor Tsetlin, Jerrel Yakel)
41
Symposium S19
Mechanisms of Cell Death in Inflammatory Processes (Chair: Malú Tansey)
43
Symposium S20
Neuron-Oligodendrocyte Interactions in Development and Disease (Chair: Vittorio Gallo, David Pleasure)
44
Symposium S21
Glia Amino Acid Transporters in Health and Disease (Chair: Arturo Ortega)
46
Symposium S22
Calpain Inhibitors in Preclinical Models of Neurodegenertion and Neurotrauma (Chair: Naren Banik, Supriti Samantaray)
48
Colloquium C03
Stroke and Mitochondria (Chair: Selva Baltan, Richard Morrison)
73
Symposium S23
Inflammation and Myelination: A Match Made in Heaven? (Chair: Dana McTigue)
50
Symposium S24
Modulaton of ABC Transporters and APOE Levels as Therapeutic Targets for Alzheimers (Chair: Danny Michaelson, Mary Jo LaDu)
52
01:00 pm - 03:00 pm
03:30 pm - 05:30 pm
3
Tuesday/Wednesday Poster Sessions
05:30 pm - 07:00 pm
Poster Session PTW01 Poster Session PTW02 Poster Session PTW03 Poster Session PTW04 Poster Session PTW05 Poster Session PTW06 Poster Session PTW07 Poster Session PTW08
Neuron-Glial Interactions 2: Development and Disease Neurodegeneration 2: Ischemia, Trauma and Other Neuroprotection and Repair Demyelination: Pathology, Protection and Repair Gene Expression/Regulation The Synapse: Signals and Plasticity Lipids: Biology and Pathobiology Late Breaking Abstracts
2015 Transactions of the American Society for Neurochemistry ®
114 119 124 127 132 136 138 140
III
Contents
Wednesday, March 18th, 2015
Page
08:00 am - 09:15 am
Plenary Lecture PL04
Basic Neurochemistry Textbook Lecture – Life in the Fast Lane: Regulation of Fast Axonal Transport and Neurodegeneration (Plenary Speaker: Scott Brady)
09:30 am - 10:00 am
Poster Sessions PTW01 to PTW08
Tuesday/Wednesday Poster Sessions
10:00 am - 12:00 pm
Symposium S25
Neurodegeneration and Oxidative Stress (Chair: Mohammed Akbar, Byoung-Joon Song)
54
Oral Presentation OP01
Post Doc Neurochemistry Symposium (Chair: Jeff Twiss)
77
Symposium S26
CDK5 Signaling: A Double-edged Sword in Normal and Diseased Brain (Chair: Yue Feng)
56
Symposium S27
Sex-Specific Effects of Adolescent Drugs and Stressors on Neural, Metabolic, Immune and Behavioral Outcomes (Chair: Kyle Frantz, Gretchen Neigh)
58
Oral Presentation OP02
Graduate Student Neurochemistry Symposium (Chair: Bruce Carter)
80
Symposium S28
Contribution of Environmental Chemicals to Neurodegenerative Disease (Chair: Mike Caudle)
60
Symposium S29
Neuroinflammation and Immune Responses in Autoimmune Demyelination Diseases (Chair: Jae Kyung Lee, Kareem Graham)
62
Symposium S30
Pharmacologically Induced Hypothermia and Clinical Implications (Chair: Shan Ping Yu, Thomas A. Dix)
64
Symposium S31
Revisit Mitochondria in Neurodegeneration (Chair: Wenzhen Duan, Xin Qi)
66
01:00 pm - 03:00 pm
03:30 pm - 05:30 pm
4
114
Author Index
144
Keyword Index
149
2015 Transactions of the American Society for Neurochemistry ®
IV
Contents In Abstract Order Plenary Lectures PL01 PL02 PL03 PL04
Page
Looking at the Epigenomic Landscape of Developing Oligodendrocytes The Teen Brain BDNF Proteins: Three Forms, Many Functions Life in the Fast Lane: Regulation of Fast Axonal Transport and Neurodegeneration
1 2 3 4
Symposia S01 S02 S03 S04 S05 S06 S07 S08 S09 S10 S11 S12 S13 S14 S15 S16 S17 S18 S19 S20 S21 S22 S23 S24 S25 S26 S27 S28 S29 S30 S31
microRNAs (miRNAs) in Alzheimer's disease (AD) and Age-related Macular Degeneration (AMD) Glial Vesicular, Metabolic and Morphologic Function in Health and Disease RNA Toxicity in Neurodegeneration Frontiers of Neurolipids Novel Strategies in Stem Cell Therapy and Clinical Potentials Autophagy in Neurodegeneration: Role in Disease and Therapy Target Central and Peripheral Inflammatory and Immune Responses in Parkinson's Disease Wiring of the Nervous System: From Molecular Mechanisms to Complex Behaviors Astrocytes as Obligatory Partners in Purinergic and Glutamatergic Neurotransmission Neuropsychiatric Disorders: New Mechanisms and Models Dissecting the Molecular Mechanisms of Acute Neurologic Injury Signaling Pathways Regulating Remyelination Mechanisms of Cell Death and Diseases Retromer and Neurodegenerative Diseases Endoplasmic Reticulum Stress in Myelin Disorders Wiring Axons by mRNA Regulation in Neuronal Health and Disease Stress Organelles and Neurodegenerative Diseases Cys-loop Receptors in Neurotransmission: Structural Aspects of Regulation by Exogenous and Endogenous Compounds Mechanisms of Cell Death in Inflammatory Processes Neuron-Oligodendrocyte Interactions in Development and Disease Glia Amino Acid Transporters in Health and Disease Calpain Inhibitors in Preclinical Models of Neurodegenertion and Neurotrauma Inflammation and Myelination: A Match Made in Heaven? Modulaton of ABC Transporters and APOE Levels as Therapeutic Targets for Alzheimers Neurodegeneration and Oxidative Stress CDK5 Signaling: A Double-edged Sword in Normal and Diseased Brain Sex-Specific Effects of Adolescent Drugs and Stressors on Neural, Metabolic, Immune and Behavioral Outcomes Contribution of Environmental Chemicals to Neurodegenerative Disease Neuroinflammation and Immune Responses in Autoimmune Demyelination Diseases Pharmacologically Induced Hypothermia and Clinical Implications Revisit Mitochondria in Neurodegeneration
5 7 9 12 15 17 19 21 23 25 27 29 31 33 35 37 39 41 43 44 46 48 50 52 54 56 58 60 62 64 66
Colloquia C01 C02 C03
Proinflammatory Mechanisms in Neurodegeneration Post-translational Prenylation of GTPases in Neurodegeneration and Repair Stroke and Mitochondria
2015 Transactions of the American Society for Neurochemistry ®
68 70 73
V
Contents In Abstract Order
Workshops
Page
PMW01 Pre-Meeting Workshop: Optogenetics Tools, Techniques and Applications in Neuroscience and Neurochemistry
75
Oral Sessions OP01 OP02
Oral Presentation I: Post Doc Neurochemistry Symposium Oral Presentation II: Graduate Student Neurochemistry Symposium
77 80
Poster Sessions I – Sunday/Monday PSM01 PSM02 PSM03 PSM04 PSM06 PSM07
Development, Differentiation and Disorders Drugs of Abuse: Alcohol, Cocaine, Methamphetamine Neuroinflammation Neuron-Glial Interactions 1: Metabolism, Signal Transduction and Axon Biology Axon Biology and Pathobiology Neurodegeneration 1: AD, PD, HD, ALS
83 89 90 100 105 106
Poster Sessions II – Sunday/Monday PTW01 PTW02 PTW03 PTW04 PTW05 PTW06 PTW07 PTW08
Neuron-Glial Interactions 2: Development and Disease Neurodegeneration 2: Ischemia, Trauma and Other Neuroprotection and Repair Demyelination: Pathology, Protection and Repair Gene Expression/Regulation The Synapse: Signals and Plasticity Lipids: Biology and Pathobiology Late Breaking Abstracts
114 119 124 127 132 136 138 140
Author Index
144
Keyword Index
149
2015 Transactions of the American Society for Neurochemistry ®
VI
Plenary Lectures PL01 Plenary Lecture of Patrizia Casaccia PL01 LOOKING AT THE EPIGENOMIC LANDSCAPE OF DEVELOPING OLIGODENDROCYTES Patrizia Casaccia-Bonnefil Mount Sinai School of Medicine, Neuroscience and Genetics& Genomics, New York, USA Recent advances have suggested an important role of myelin in plasticity and axonal survival, thereby highlighting the critical importance of oligodendrocyte differentiation for functional activity of the central nervous system. Several studies identified transcription factors and histone modifications regulating the progression of __________________
2015 Transactions of the American Society for Neurochemistry ®
progenitors into oligodendrocytes. This lecture will discuss oligodendrocyte lineage progression as the integration of transcriptional networks, nuclear structure and epigenetic modifiers, including histone-specific enzymatic activities, microRNA and DNA modifications. We suggest that progenitors are directly responsive to signals modulating transcription factors, due to predominantly euchromatic nuclei. Pre-myelinating and myelinating cells, in contrast are characterized by the formation of heterochromatin, which modifies the association of DNA with nucleosomal histones and renders the access of transcription factor dependent on the activity of epigenetic modulators.
1
PL02 Plenary Lecture of Jay Giedd PL02 THE TEEN BRAIN: INSIGHTS FROM NEUROIMAGING Jay Giedd University of California, San Diego, Department of Psychiatry, La Jolla, USA Jay N. Giedd, M.D. is a Professor in the Department of Psychiatry at the University of California, San Diego, and Director of the Division of Child and Adolescent Psychiatry at the Rady Children’s Hospital-San Diego. In addition, he is an adjunct Professor at Johns Hopkins School of Public Health in the Department of Family and Reproductive Medicine. Over the past 23 years Dr. Giedd has combined brain imaging, genetics, and behavioral analysis to explore the path and influences of brain development in health and illness. As one of the most highly cited neuroscientists of his generation, his __________________
2015 Transactions of the American Society for Neurochemistry ®
over 200 scientific publications have had a transformative impact on medicine, psychology, education, judicial, and public policy. Dr. Giedd’s recent work has focused on how new insights from pediatric neuroscience can be used to optimize the environment for healthy brain development, particularly regarding education and the use of digital technologies that have transformed the way youth learn, play, and interact with each other. In addition to his numerous academic awards, his work has been prominently featured in the general media with cover stories in Time, National Geographic, and national newspapers as well as over 30 television documentaries. For his outreach to students of all ages and frequent talks to parents, teachers, mental health workers, legislators, and the general public, Dr. Giedd was honored as co-recipient of the 2012 Society for Neuroscience’s Science Educator Award.
2
PL03 Plenary Lecture of Barbara Hempstead PL03 BDNF PROTEINS: THREE FORMS, MANY FUNCTIONS Barbara Hempstead Weill Cornell Medical College, Medicine, New York, USA Structural and biochemical alterations in brain circuitry during childhood and adolescence can affect learning, memory, and functional circuitry later in life, although the molecular mechanisms that regulate these processes are incompletely understood. Mature brainderived neurotrophic factor (BDNF) plays crucial roles in modulating neuronal structure and synaptic plasticity, by activation of the TrkB receptor tyrosine kinase to promote dendritic arborization and enhance LTP. Like most growth factors, BDNF is initially synthesized as a precursor form, proBDNF, which can be cleaved to a N-terminal prodomain and C-terminal mature BDNF. While mature BDNF is the most abundant form in adulthood, significant levels of intact proBDNF are detected in neonatal and adolescent rodent brains, and proBDNF can be released from neurons in an activity dependent manner. In vivo, proBDNF negatively regulates hippocampal dendritic complexity and spine density utilizing p75, and hippocampal slices from proBDNF-expressing __________________
2015 Transactions of the American Society for Neurochemistry ®
mice exhibit depressed synaptic transmission, impaired LTP and enhanced LTD. These results suggest that proBDNF acts in vivo as a biologically active factor to regulate hippocampal structure and plasticity, effects that are distinct from mature BDNF. We have also evaluated the biological activities of the prodomain of BDNF. In humans, a common single nucleotide polymorphism (SNP) in the BDNF prodomain that leads to valine-to-methionine substitution at codon 66 (Val66Met) has provided insights into the role of BDNF in altered learning and memory, especially in the realm of fearrelated processes. We have observed that the prodomain is released from neurons in an activity dependent manner. While the Val and Met prodomains are intrinsically disordered, the Val66Met substitution induces structural changes that lead to differential engagement with the SorCS2 receptor. This results in acute growth cone retraction of hippocampal neurons following exposure to Met prodomain, but not Val prodomain, an effect that requires expression of SorCS2 and p75. These results identify the Met prodomain as a new active ligand that acutely modulates neuronal morphology. Thus, in humans, the BDNF gene encodes three distinct ligands that can alter neuronal function.
3
PL04 Plenary Lecture of Scott Brady PL04 LIFE IN THE FAST LANE: REGULATION OF FAST AXONAL TRANSPORT AND NEURODEGENERATION Scott Brady Univ. of Illinois/Chicago, Dept. of Anatomy & Cell Biology, Chicago, USA Alzheimer’s, and Huntington’s disease as well as other adult-onset neurodegenerative diseases like Amyotrophic Lateral Sclerosis present as dying back neuropathies. This pattern of neurodegeneration can result from changes in fast axonal transport (FAT) and pathogenic changes in FAT are a hallmark of these diseases. In each case, disease-related inhibition of FAT can be related to dysregulation of signaling pathways that may normally regulate the targeting of FAT cargoes. Critical for an understanding of these diseases is character__________________
2015 Transactions of the American Society for Neurochemistry ®
ization of the molecular basis relating alteration of neuronal signaling and the observed inhibition of FAT to the presence of pathogenic proteins. For example, the selective reductions in FAT seen in Huntington’s and Alzheimer’s disease result from activation of specific neuronal kinases that phosphorylate molecular motors like kinesin and dynein. However, these different neurodegenerative diseases involve different kinase activities, leading to disease-specific effects on FAT. A theme has now emerged indicating that misfolded proteins become pathogenic when biologically active motifs are exposed. The characteristic phenotype of each disease can be related to the specific motif(s) involved and the kinases activated as a result. These diseases represent a distinctive class of neuropathology closely linked to the regulation of FAT in normal and pathological states.
4
Symposia S01 microRNAs (miRNAs) in Alzheimer's Disease (AD), Age-related Macular Degeneration (AMD) S01-01
S01-02
ROLE OF MICRORNA-346 IN IRON HOMEOSTASIS AND ALZHEIMER’S DISEASE Debomoy Lahiri, Justn Long Indiana University School of Medicine,, Dept. of Psychiatry,, Indianapolis, USA
MICRORNA-650 MODULATES THE PATHOGENESIS OF ALZHEIMER’S DISEASE THROUGH REGULATING CDK5 Peng Jin Emory University School of Medicine, Human Genetics, Atlanta, USA
A major hallmark of Alzheimer’s disease (AD) is the presence of excessive amyloid-! (A!) peptide as neuritic plaques. The ratelimiting step in A! production is the proteolytic cleavage of the A! precursor protein (APP) by !-secretase or BACE1. Dysregulation of proteins involved in A! production, such as APP and BACE1, may contribute to excess A! deposition. We study how these gene products are regulated by microRNAs, which are an abundant class of small RNAs that mediate potent regulatory effects on global gene expression. Using bioinformatics and functional studies in human cultures and brain specimens, we recently reported specific microRNA species regulate APP levels (Long et al, 2012). We also identified a novel BACE1-specific miRNA (Long et al, 2014). APP is regulated post-transcriptionally with changes in iron homeostasis. Without iron, IRP1 binds to an iron response element (IRE) in the APP 5’-UTR and inhibits translation. MiRNAs post-transcriptionally regulate APP expression via the APP 3’-UTR. MiRNAs occasionally mediate regulatory effects via 5’-UTR and coding sequence, no such regulatory interactions are known for APP. We report that miR-346 stimulates APP expression via APP 5’-UTR at a site that overlaps with the IRE. We demonstrate a novel regulatory microRNA interaction controlling APP expression that may be targeted therapeutically in AD. This interaction is unique in that miR-346, stimulates APP expression by targeting the APP 5’-UTR. Most microRNA interactions are inhibitory and act via the 3’-UTR. The mechanism involves blockade of an IRE by miR-346. The inhibition of miR-346 should then synergize with iron chelation strategies as an AD treatment. APP 5’-UTR also contains a site for the acute box element and is also subjected to inflammatory pathway. During mucosal inflammation, TNF-" induces miR-346, which downregulates epithelial Vitamin D Receptor (VDR). miR346 also controls TNF-" synthesis (Semaan et al., 2011). Taken together, miR-346 plays an important role in iron homeostasis and anti-inflammation, and its dysregulation may lead to AD. Thus, our reporting of miRNA-346 activates levels of endogenous human APP and maintains cellular iron balance is significant.
Alzheimer’s disease (AD) is pathogenically featured with the progressive neurodegeneration, amyloid-! (A!) plaques and neurofibrillary tangles. Ample evidence has indicated the involvement of epigenetic pathways in AD pathogenesis. Here, we show that microRNA 650 (miR-650) showed altered expression in AD brain. Meanwhile, the processing from primary miR-650 to mature miR650 is also altered. Bioinformatic analysis predicted CyclinDependent Kinase 5 (CDK5) is a potential target of miR-650. The overexpression of miR-650 using lentiviral vector could significantly decrease CDK5 level and increase microtubule-associated protein 2 positive neurons in the hippocampus of APP-PSEN1 transgenic mice. Our results indicate that miR-650 is involved in the AD pathogenesis through modulating CDK5.
2015 Transactions of the American Society for Neurochemistry ®
S01-03 MICRORNA THERAPY FOR ALZHEIMER'S DISEASE Sebastien Hebert 1, 2 1 Université Laval, Psychiatry and Neurosciences, Quebec, Canada 2 Centre de recherche du CHU de Quebec, Neurosciences, Quebec, Canada Failure at different clinical trials emphasizes the need for developing new therapeutics for Alzheimer disease (AD), a fatal neurodegenerative disorder and the most common form of dementia. The naturally occurring microRNAs (miRNAs) have attracted much attention in recent years for their potential therapeutic applications in humans. These small regulatory RNAs function as master regulators of the genome, and are important for brain homeostasis. We and others have identified a number of miRNAs that are misregulated in AD patients. Among those miRNAs, the highly conserved miR-16/15 family is of high interest. In functional assays in vitro and in cells, we could show that specific miR-16/15 members could regulate key AD-related genes, including APP, BACE1, and Tau. Accordingly, candidate miRNAs significantly reduced amyloid-! and Tau-related pathologies, the main hallmarks of AD. Validation studies in mice demonstrated that miRNA delivery to the brain is safe and well tolerated by the organism. Finally, accumulating studies suggest that selected miR-16/15 family members could function as powerful biomarkers for AD. These results will help to further investigate the potential therapeutic and diagnostic use of miRNAs in neurodegenerative disorders.
5
S01 microRNAs (miRNAs) in Alzheimer's Disease (AD), Age-related Macular Degeneration (AMD)
S01-04 MICRORNA: SPECIATION & COMPLEXITY, SEQUENCE & STABILITY, VIROID-LIKE PROPERTIES & DISEASE PROPAGATION WITHIN THE HUMAN CNS Walter Lukiw 1, 3, Aileen Pogue 2, Surjyadipta Bhattacharjee 1 Prerna Dua 4, Brandon Jones 1, Yuhai Zhao 1, Evgeny Rogaev 5 James Hill 6 1 Louisiana State University, Neurosci. Ctr., New Orleans, LA, USA 2 Alchem Biotech, Neurobiology, Toronto, ON, Canada 3 Louisiana State University, Ophthalmology, New Orleans, USA 4 Louisiana State University, Information Management, Ruston, LA, USA 5 University of Massachusetts, Genetics, Worcester, MA, USA 6 Louisiana State University, Department of Microbiology and Pharmacology, New Orleans, LA, USA MicroRNAs-(miRNAs) represent a relatively recently-discovered class of small non-coding single-stranded RNAs (ssRNAs) that have gained considerable attention in the molecular-genetic mechanisms that contribute to human-health, aging and-disease. As highly-soluble and mobile-entities, emerging-evidence indicates that miRNAs possess a highly-selected ribonucleotide-sequencestructure, are part of an evolutionary-ancient genetic-signaling system, resemble plant-ssRNA pathogens known as viroids in their structure-and-function, and are very-abundant in the physiologicalfluids that surround the-cells-and-tissues of the human-CNS. Persistence and altered abundance of miRNAs in the extracellular__________________
2015 Transactions of the American Society for Neurochemistry ®
fluid (ECF) or cerebrospinal-fluid (CSF) may play a role in the intercellular spreading of genetic signals, including pathogeneticsignals, systemically throughout functionally-linked cellular and tissue-systems such as those encountered within the CNS. As such, miRNAs represent attractive diagnostic-disease-markers and pharmaceutical-targets for novel-drug-development. Human-brain and retinal-miRNAs appear to utilize novel-biological signalingsystems, such as circular-RNAs (circRNAs) and extracellularvesicles containing miRNAs, to carry out and propagate both homeostatic-and pathogenic-miRNA functions. miRNA-abundance and complexity appear to exhibit-variation between different individuals and human-populations in accordance with the concept of ‘human genetic individuality’. This paper will review some of the more intriguing features of these highly structured ssRNAs with emphasis on their presence and function in the human CNS, with particular reference to Alzheimer’s disease and prion disease wherever possible. Acknowledgements/Support: Research on neurotoxic-metals, small non-coding RNA, microRNA, the innate-immune response, amyloidogenesis, neuroinflammation and possible vira-contribution to the Alzheimer’s disease (AD) process was supported through the Alzheimer Association, an unrestricted grant from Research to Prevent Blindness (RPB), the Louisiana Biotechnology Research Network (LBRN) and NIH grants from the National Eye Institute (NEI) and National Institute of Aging (NIA).
6
S02 Glial Vesicular, Metabolic and Morphologic Function in Health and Disease S02-01 ADRENERGIC CONTROL OF ASTROCYTE MORPHOLOGY, EXCITABILITY AND METABOLISM Nina Vardjan 1, 2 1 Celica BIOMEDICAL, Laboratory of Cell Engineering, Ljubljana, Slovenia 2 Faculty of Medicine, University of Ljubljana, Institute of Pathophysiology, Laboratory of Neuroendocrinology-Molecular Cell Physiology, Ljubljana, Slovenia Astrocytes are an abundant type of glial cells in the brain. They form tight connections with synapses (tripartite synapse), contain glycogen and provide for homeostasis in the CNS. It was shown recently that extracellular space is changing diurnally, which likely involves noradrenergic signaling. Extracellular space regulation may in part be due to adrenergic control of astrocyte shrinkage, as observed during adrenergic activation of cycling AMP, a second messenger, monitored by FRET nanosensor measurements. Astrocyte shape changes may also be modulated by vesicles, which play a role in cell-to-cell communication as in astrocytic antigen presentation in neuroinflammatory states. During this process astrocytes become reactive. They overexpress intermediate filaments, which augment the cytoplasmic traffic of vesicles to the plasma membrane, enhancing the probability of fusion of these vesicles with the plasma membrane. In this lecture the results of how adrenergic activation of astrocytes modulates cytoplasmic calcium excitability, and how this relates to release of gliotransmitters, astrocyte volume changes, and energy provision in the CNS will be presented. Hence optical measurements of gliotransmitter release, astrocyte morphology changes, and changes in second messengers (cAMP, calcium) and metabolites (glucose, lactate) in response to adrenergic activation were conducted in single astrocytes. Adrenergic receptor regulation of astrocytes may have profound effects on neural network activity in health and neurologic disorders, including trauma, neurodegeneration, neuroinflammation, and neurodevelopment disorders.
S02-02 DUAL ROLE OF MICROGLIA IN NEURODEGENERATIVE DISEASES Michela Matteoli 1, 2, Sonia Mazzitelli 2 1 National Research Council, Institute of Neuroscience, Milano, Italy 2 Humanitas Research Hospital, Neuroscience, Rozzano, Italy Astroglial cells are housekeepers of the central nervous system. Astrocytes and microglia support CNS development and control synaptogenesis, provide for maintenance of the extracellular environment and form multiple lines of neural tissue defense, also contributing to cognitive functions through multi-directional communication with cells residing in the brain. The cell-to-cell communication between astrocytes, microglia and neurons is mediated, to a large extent, by the secretion of signaling molecules through conventional and unconventional forms of exocytosis. Importantly, __________________ 2015 Transactions of the American Society for Neurochemistry ®
glial cells are the major producers of inflammatory mediators and glia dysfunctions have been widely demonstrated in different neurodegenerative diseases. Although the inflammatory response during most chronic neurodegenerative disease is known to be dominated by microglia, the mechanisms by which these cells lead to neuronal damage and dysfunction is still to be defined. This is especially true in the case of Alzheimer’s Disease, where microglia cells are recruited to beta amyloid plaques, with microglial activation increasing linearly throughout the disease and specifically correlating with neurodegeneration. I will report data showing that activated microglia directly control the formation of beta amyloid aggregates in Alzheimer’s Disease, through both extracellular and intracellular metabolic processes. These data underline the pathogenic role of microglia and inflammation in the initiation and progression of Alzheimer’s Disease and support the possibility that a microgliatargeted therapy may be potentially beneficial for the treatment of the pathology.
S02-03 METABOLIC REGULATION OF VESICULAR GLUTAMATE RELEASE FROM CULTURED ASTROCYTES Vedrana Montana 1, Daniel Flint 1, Landon Wilson 2 Helle Waagepetersen 4, Arne Schousboe 4, Vladimir Parpura 1 1 University of Alabama, Neurobiology, Birmingham, USA 2 University of Alabama, Pharmacology, Birmingham, USA 3 University of Copenhagen, Drug Design and Pharmacology, Copenhagen, Denmark 4 University of Rijeka, Biotechnology, Rijeka, Croatia Astrocytes have a prominent role in brain physiology and pathophysiology. In addition to maintaining blood flow, metabolic and ionic homeostasis, they have capability of signaling to adjacent neurons by releasing glutamate via process of regulated exocytosis. Astrocytes synthesize glutamate de novo owing to pyruvate entry to the citric acid cycle via pyruvate carboxylase. Pyruvate is sourced from the utilization of two metabolic fuels, glucose and lactate. Glucose can be polymerized to glycogen and stored as fuel within astrocytes and/or lysed to pyruvate, while lactate can be converted to pyruvate. To that end, we investigated the role of the above energy sources, glycogen, glucose and lactate, in exocytotic glutamate release from astrocytes. We used purified primary astrocyte cultures acutely incubated (1 hour) in glucose and/or lactatecontaining media. We used mechanical stimulation, known to increase intracellular calcium levels and cause exocytotic glutamate release. Using single cell fluorescence microscopy, we monitored stimulus-induced intracellular calcium responses as well as glutamate release to the extracellular space. Our data indicate that glucose, either taken-up from media or mobilized from the glycogen storage, sustained glutamate release, while the availability of lactate significantly reduced the release of glutamate from astrocytes. Based on further pharmacological manipulation, it appears that lactate caused metabolic changes consistent with an increased synthesis of fatty acids. The above metabolic and functional changes were corroborated by tandem mass spectrometry proteomics analysis __________________ 7
S02 Glial Vesicular, Metabolic and Morphologic Function in Health and Disease which confirmed appropriate altered protein expression. These findings support the notion that the availability of energy sources and metabolic milieu play a role in glial-neuronal interactions and modulation of synaptic activity in health and disease.
S02-04 THE TRIPARTITE SYNAPSE - A DEVELOPMENTAL RESTRICTED PHENOMENON? Maiken Nedergaard Univ of Rochester, Center for Translational Neuromedicine, Rochester, USA
decade ago, when astrocytes were shown to propagate Ca2+ signals that was linked to glutamate release in cultured preparations. However, several independent lines of observations have in recent years questioned whether our current model for neuroglia signaling conceptualized as the tripartite synapse is valid in the adult mammalian brain. For example, astrocytes in adult behaving mice do not exhibit Ca2+ increases in response to local glutamatergic transmission, but display widespread increases in Ca2+ in response to release of the neuromodulator, norepinephrine. Moreover, no evidence for Ca2+ mediated exocytotic release of gliotransmitter release have been collected in vivo questioning the relevance of existing literature based largely on ex vivo preparations. Thus, a critical re-evaluation of the basic pathways of both neuron-to-glia, but also glia-to-neuron signaling in the adult nervous system is warrented.
Astrocytes are electrically none excitable cells that play important homeostatic roles. Much excitement was generated more than a __________________
2015 Transactions of the American Society for Neurochemistry ®
8
S03 RNA Toxicity in Neurodegeneration S03-01 ROLE OF SYNAPTIC DYSFUNCTION IN C9ORF72-MEDIATED PATHOGENESIS IN PATIENT-DERIVED IPS NEURONS AND IN VIVO ANIMAL MODELS Rita Sattler 1 1 Johns Hopkins University, Neurology, Baltimore, USA 2 Johns Hopkins University, Brain Science Insititute, Baltimore, USA The newly discovered gene mutation in C9orf72 represents the most common genetic abnormality in frontotemporal dementia (FTD; 10-30%) and Amyotrophic Lateral Sclerosis (ALS; 20-50%). This mutation is characterized by an expanded GGGGCC (G4C2) hexanucleotide repeat in the non-coding region of the C9orf72 gene on chromosome 9p21. Previous studies in our laboratory using patient derived adult pluripotent stem cells differentiated into neurons (iPSNs) support the presence of RNA toxicity as a mechanism of disease pathogenesis for C9orf72 carriers. We hypothesize that this RNA toxicity leads to aberrant RNA processing of synaptic proteins, which in turn results in synaptic dysfunction and consequently in cognitive impairment as well as increased susceptibility to cellular stressors, including excitotoxicity. To examine neuronal dendrite and spine morphology, we transduced patient derived (ALS and FTD) adult pluripotent stem cells (iPSCs) differentiated into mixed neurons with a lentivirus overexpressing eGFP. Confocal microscopy and 3D image analysis revealed decreased dendritic branching (as quantified by Sholl Analysis) and decreased spine density in C9orf72 iPSNs. In addition, we performed immunohistochemistry on major synaptic proteins (per-and postsynaptic marker proteins) and revealed altered expression patterns showing a loss of puncta appearance in C9orf72 iPSNs compared to control, non-diseased iPSNs. Ongoing experiments are aimed to confirm these in vitro findings in whole animals by injecting AAV viral constructs overexpressing (GGGGCC) repeats of different lengths. In addition, we will examine neuronal morphology and spine structures in a novel BAC C9 animal model. In conclusion, our data strongly suggest that synaptic dysfunction plays a role in C9ORF72 pathogenesis and may explain increased susceptibility to cellular stressors as well as cognitive impairment, as observed in FTD patients as well as a large number of C9ORF72 ALS patients.
S03-02 NUCLEAR TRANSPORT DEFECT UNDERLIES C9ORF72 ALS/FTD NEURONAL INJURY Christopher Donnelly Johns Hopkins University School of Medicine, Department of Neurology/Neuromuscular Division, Baltimore, USA A hexanucleotide repeat expansion in the C9ORF72 gene has been identified as the most common known genetic cause of familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Recent evidence employing iPSneurons from C9ORF72 ALS patients suggests that the GGGGCC RNA products transcribed from the repeat expansion form complex G-quartet RNA structures and are neurotoxic. Similar to other __________________ 2015 Transactions of the American Society for Neurochemistry ®
repeat expansion disorders (e.g., myotonic dystrophy type I and II), the C9ORF72 GGGGCCexp RNA can sequester nuclear factors, including RNA binding proteins, and we hypothesize that these aberrant interactions are the primary cause of C9ORF72 neurotoxicity. Previous work from our laboratory has indicated that the GGGGCC RNA interacts with RanGAP1, a regulator of Ranmediated cyto-nuclear trafficking. We have also found that RanGAP1 is a robust suppressor of neurotoxicity in a Drosophila model system that overexpresses GGGGCC RNA. Consistent with a RanGAP1 loss-of-function model, we have found that the G4C2 Drosophila model show reduced nuclear localization of NLScontaining reporters and iPS-neurons from C9ORF72 ALS patients exhibit perturbed Ran protein gradients and reduced nuclear import rates of classical NLS-containing reporters. These nuclear transport deficits are rescued by treating the Drosophila and C9ORF72patient derived iPS-neurons with antisense oligonucleotides that target GGGGCC repeat-containing RNAs or small molecules that bind G-quartet RNA structures to prevent any interaction with endogenous proteins. Moreover, we have observed pathological RanGAP1 intra- and peri-nuclear inclusions in Drosophila cells that express G4C2 RNA, C9ORF72 ALS iPS neurons, and human motor cortex, which also colocalize with nucleoporins of the nuclear pore complex (NPC). Taken together, these data suggest that deficits in nucleocytoplasmic trafficking due to loss of RanGAP1 or NPC function underlie G4C2 RNA-mediated neurotoxicity.
S03-03 RAN TRANSLATION IN C9ORF72 FRONTOTEMPORAL DEMENTIA AND AMYOTROPHIC LATERAL SCLEROSIS Tania Gendron Mayo Clinic, Neuroscience, Jacksonville, USA A hexanucleotide repeat expansion within the C9orf72 gene is the most common known mutation causative of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), two devastating neurodegenerative diseases that share genetic, neuropathological and clinical overlap. Elucidating how the bidirectionally transcribed G4C2!G2C4 expanded repeat causes “c9FTD/ALS” has become an important goal of the field. Likely pathogenic mechanisms include toxicity induced by G4C2 and G2C4 repeat-containing RNAs. For instance, transcripts of the expanded repeat interact with various RNA-binding proteins and cause their sequestration in discrete nuclear structures, termed RNA foci. In addition, these transcripts undergo repeat associated non-ATG (RAN) translation, a form of translation that occurs in the absence of an ATG start codon and despite the fact that the repeat is located within a non-coding region of C9orf72. This unconventional mode of translation results in the production of poly(GA), poly(GP), poly(GR), poly(PR) and poly(PA) dipeptide repeat proteins, collectively referred to here as c9RAN proteins. c9RAN proteins form neuronal inclusions throughout the central nervous system of c9FTD/ALS patients, implicating RAN translation as a mechanism of disease. This presentation will focus on the putative contribution of c9RAN proteins to disease pathogenesis and their influence on clinical phenotype, as well as their potential use as diagnostic and pharmacodynamic biomarkers. 9
S03 RNA Toxicity in Neurodegeneration
S03-04 CUG RNA TOXICITY DISRUPTS THE DEVELOPMENTALLY REGULATED MICRORNA PROGRAM IN MYOTONIC DYSTROPHY Auinash Kalsotra 1, Thomas Cooper 2 1 University of Illinois, Biochemistry, Urbana, USA 2 Baylor College of Medicine, Pathology and Immunology, Houston, USA Myotonic dystrophy type 1 (DM1) is a neuromuscular disease caused by an expanded CTG repeat in the last exon of the dystrophia myotonica-protein kinase (DMPK) gene. Pathogenesis is caused primarily by the mRNA containing expanded CUG repeats (CUGexp RNA) that is expressed from the mutated allele. Cardiac dysfunction is the second leading cause of death in myotonic dystrophy type 1 (DM1) primarily due to arrhythmias and cardiac conduction defects. A screen of more than 500 miRNAs in a DM1 mouse model identified 54 that were differentially expressed in heart. More than 80% exhibited down regulation towards the embryonic expression pattern and showed a DM1-specific response. Twenty of 22 miRNAs tested were also significantly down regulated in human DM1 heart tissue. We demonstrate that many of these miRNAs are direct MEF2 transcriptional targets including miRNAs for which depletion is associated with arrhythmias or fibrosis. MEF2 protein is significantly reduced in both DM1 and mouse model heart samples and exogenous MEF2C restores normal levels of MEF2 target miRNAs and mRNAs in a DM1 cardiac cell culture model. We conclude that loss of MEF2 in DM1 heart causes pathogenic features through aberrant expression of both miRNA and mRNA targets.
S03-05 YEAST GENETIC SCREENS REVEAL NEW INSIGHTS INTO NEURODEGENERATIVE DISEASE MECHANISMS: TDP-43, FUS/TLS, C9ORF72, AND BEYOND Aaron Gitler Stanford University School of Medicine, Department of Genetics, Stanford, USA Several human neurodegenerative diseases, which include Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis (ALS) share a common feature in protein misfolding and aggregation. We use the baker’s yeast, Saccharomyces cerevisiae, as a simple, yet powerful, model system to study the cell biology underpinning protein-misfolding diseases. We have focused on the ALS disease proteins TDP-43 and FUS/TLS and have used yeast models to define novel disease mechanisms and have extended our findings into animal models and even recently into human patients. We recently discovered mutations in one of the human homologs of a hit from our yeast TDP-43 modifier screen in ALS patients. Mutations in this gene are relatively common (~5% of cases) making it one of the most common genetic risk factors for ALS discovered to date. This underscores the power of such simple model systems to help reveal novel insight into human disease. These screens are also providing new and completely unexpected potential drug targets – another hit from one the TDP-43 modifier screens is a __________________
2015 Transactions of the American Society for Neurochemistry ®
gene encoding lariat debranching enzyme and we discovered that inhibiting this enzyme potently suppresses TDP-43 toxicity in yeast and in mammalian neurons. Launching from these studies in yeast to test known ALS disease genes, we have also been using yeast as a discovery platform to predict novel ALS disease genes based on functional properties (for example, the presence of a prion-like domain) and to combine this approach with human genetics and next generation sequencing to further define the complex genetic landscape of ALS. We anticipate that our novel approach will be broadly applicable to other human disease situations, many of which are deeply rooted in basic biology. We will present new data on using two new yeast models and genetic screens to uncover mechanisms by which the new ALS/FTD gene C9orf72 contributes to disease and also unexpected new facets to Parkinson’s disease pathogenesis.
S03-06 TDP-43 DEPENDENT TRANSLATION DYSREGULATION IN AMYOTROPHIC LATERAL SCLEROSIS Daniela Zarnescu, Scott Daniel, Alyssa Coyne Bhavani Bagevalu Siddegowda, Shizuka Yamada University of Arizona, Molecular and Cellular Biology, Tucson, USA RNA dysregulation has recently been demonstrated to be a critical contributor to the pathophysiology of amyotrophic lateral sclerosis (ALS) and related neurodegenerative diseases. Several RNA binding proteins have been identified in pathologic aggregates and have also been shown to harbor mutations causative of ALS. Among these, TDP-43 is linked to the vast majority of ALS cases, has been implicated in several aspects of RNA metabolism including splicing, transport, storage in stress granules and translation. These findings suggest an intimate link between TDP-43, RNA stress granules, translation and disease. Although recent studies have provided insights into the relationship between TDP-43 and RNA stress granules in ALS, our current knowledge of TDP43’s role in translation, the identity of its translational mRNA targets and their contribution to disease remain poorly understood. Using a combined proteomic and genetic approach we identified several RNA binding proteins including translation initiation factors as functional partners of TDP-43. Among these, Fragile X protein (FMRP), a well established ribosome associated translational regulator is neuroprotective by remodeling TDP-43 containing complexes and restoring translation of specific mRNA targets. In addition, using ribosome profiling we have identified several candidate translational targets. Gene ontology term analyses pinpoint to several putative targets involved in synaptic function including synaptic vesicle, cytoskeletal associated proteins and molecular chaperones. Using a combined molecular and genetic approach in a Drosophila model of ALS we find that Hsc70, a ubiquitous molecular chaperone is undertranslated at the neuromuscular synapse and that restoring its expression rescues locomotor dysfunction and improves lifespan in a disease variant dependent manner.
10
S03 RNA Toxicity in Neurodegeneration
S03-07 ALS-CAUSING MUTATIONS IN THE FUS PRION-LIKE DOMAIN BLOCK TRANSCRIPTION-COUPLED PRE-MRNA SPLICING Robin Reed, Yong Yu Harvard Medical School, Cell Biology, Boston, USA Pre-mRNA splicing is coupled to RNA polymerase II (RNAP II) transcription. Previously, we established an in vitro system for this coupled reaction1. Analysis of U1 snRNP in this system revealed that it is abundantly associated with both RNAP II2 and the ALScausative protein FUS3. In addition, FUS associates with RNAP II3,4. These observations suggested FUS as a candidate for mediating interactions between RNAP II and U1 snRNP, and thereby coupling transcription to splicing. To investigate the association of FUS with U1 snRNP and RNAP II, we used a U1 antisense morpholino oligo (AMO) that base pairs to U1 snRNA. We found that the U1 AMO abolishes the association between U1 snRNP and both FUS and RNAP II. In contrast, FUS remains associated with RNAP II. To investigate whether FUS mediates U1 snRNP-RNAP __________________
2015 Transactions of the American Society for Neurochemistry ®
II interactions, we prepared FUS knockdown nuclear extracts5. Significantly, we found that U1 snRNP cannot interact with RNAP II in the FUS knockdown extracts. Moreover, in these extracts, transcription is normal but splicing is inhibited. Together, our data indicate that FUS plays a critical role in splicing by coupling transcription to splicing via mediating an interaction between RNAP II and U1 snRNP. To determine whether there is a link between ALS and the function of FUS in transcription/splicing, we examined the effect of ALS-causing mutations in FUS. Strikingly, this analysis revealed that mutations in the prion-like domain of FUS, but not in the NLS, specifically inhibit splicing in the coupled system, and the inhibition is dose-dependent. Together, our data indicate that ALS disease mechanisms caused by FUS mutations involve disruption of transcription-coupled splicing. 1. Das et al., G&D, 2006, 20: 1100 2. Das et al., Mol. Cell, 2007, 26: 867 3. Yamazaki et al., Cell Reports 2012, 2: 799 4. Schwartz et al., G&D, 2012, 26: 2690 5. Folco et al., JoVE, 2012, 64: e4140
11
S04 Frontiers of Neurolipids S04-01
S04-02
DEFICIENCY OF GM1 AS CAUSE OF IDIOPATHIC PARKINSON’S DISEASE: RESTORATION OF GM1-GDNF INTERACTION AS CURE IN PD MOUSE. Robert Ledeen 1, Gusheng Wu 1, Piotr Hadaczek 2, Zi-Hua Lu 1 Michael Higgins 1, Neil Majmundar 1, Peter Sharoupim 1 Yoon Ho Park 1, Adam Yu 1, Victoria Nguyen 1, John Forsayeth 2 1 Rutgers, New Jersey Medical School, Neurology & Neurosciences, Newark, USA 2 University of California San Francisco, Neurological Surgery, San Francisco, USA
TOXICITY OF LONG CHAIN BASES: ACCUMULATION OF LONG CHAIN BASES UNDERLIES NEURODEGENERATION CAUSED BY CERS1 DEFICIENCY Lihong Zhao 1, Ye Liu 1, Jacek Bielawski 2, Stefka Spassieva 3 1 The Jackson Laboratory, Research, Bar Harbor, ME, USA 2 Medical University of South Carolina, Biochemistry and Molecular Biology, Charleston, SC, USA 3 Medical University of South Carolina, Medicine, Charleston, SC, USA
Several studies have employed GM1 ganglioside to treat animal models of Parkinson’s disease (PD), and a recent randomized, controlled, delayed start trial showed that PD patients treated with GM1 had lower UPDRS motor scores than at baseline after 120 weeks (Schneider et al. 2013). Our studies of mice with disrupted B4galnt1 gene (Wu et al. 2011; 2012) demonstrated pathogenesis of PD symptoms due to deficiency of ganglio-series gangliosides. Both knockout and heterozygous (HT) mice showed depletion of striatal dopamine (DA), loss of TH+ nigral neurons, and aggregation of alpha-synuclein. These manifestations of parkinsonism were largely alleviated by LIGA20, a membrane permeable analog of GM1, and also by GDNF (via AAV2) which is essential for survival of catecholaminergic neurons. Immunohistochemical analysis of substantia nigra sections revealed significant GM1 deficiency in TH+ nigral neurons of PD patients. Study of the occipital cortex, a less involved brain region, also revealed significantly lower GM1 in PD, suggesting systemic GM1 deficiency as possible risk factor in idiopathic PD (Hadaczek et al. 2014). That study also demonstrated GM1 association with GFR!1 and Ret, components of the GDNF receptor, and GM1 essentiality in maintaining these receptor proteins in functional mode. The nigral neurons of PD brain showed deficient GDNF signaling. We propose the above HT mouse with partial GM1 deficiency (like that of PD) as an especially useful PD model in reflecting its actual pathophysiology. This is supported by detection of 3 non-movement disorders in HT mice characteristic of PD: (a) gastrointestinal pathology (constipation), (b) cardiac sympathetic denervation, and (c) cognitive impairment. These too were alleviated by LIGA20, further suggesting the potential utility of membrane permeable GM1 analogs for PD therapy. [Supported: NIH grant RO1 NS33912].
Alterations of sphingolipids have been found in many diseases, including neurological disorders such as Alzheimer’s disease, Parkinson’s disease, HIV-induced dementia and peripheral neuropathy. However, the exact contributions of different sphingolipid species and metabolites to disease progression are not clear. The goal of this work is to dissect pathological roles of ceramides and long chain bases. We showed that mutations in ceramide synthase 1 (CerS1), the major enzyme for C18 ceramide synthesis in neurons, cause early-onset cerebellar Purkinje cell death and age-related accumulation of ubiquitin-positive lipofuscin deposits in the mouse brain. A recent study in human also linked CerS1 deficiency to myoclonus epilepsy and dementia. However, because CerS1 deficiency results in abrupt elevation of LCBs and severe reduction of C18 ceramide simultaneously, it was unclear which of these changes is the cause of neuron death. To understand specific roles of LCBs and ceramides in neurodegeneration, we modulated ceramide biosynthesis using a genetic approach, by transgenic expressing CerS2, a non-neuronal ceramide synthase that produces C22 and C24 ceramides, in neurons. This led to reduction of LCBs but only minor changes in ceramides and complex sphingolipids examined. Surprisingly, the CerS1 deficient neuronal pathology was almost completely suppressed by the CerS2 transgene expression. This demonstrates that accumulation of LCBs likely is the cause of neurodegeneration, whereas reduction in ceramide and/or alteration in ratios of ceramide species and complex sphingolipids, at least that between C18 and C22/C24 species, is well tolerated by neurons. This result also sheds light on the roles of LCBs and ceramides in other neurodegenerative diseases in which these lipids display changes.
S04-03 CERAMIDE HOMEOSTASIS IN THE BRAIN Glyn Dawson, Fernando Testai, John Kilkus, Jingdong Qin University of Chicago, Dept Pediat, Chicago, USA Ceramide (N-acylsphingosine: Cer) has long been associated with programmed cell death in the CNS and elsewhere and the mechanisms governing its homeostasis are complex. Cer can be synthesized de novo by many organisms but in mammals this requires molecular oxygen to convert dihydroceramide (DHC) to Cer. Hypoxia and stroke inhibit synthesis of Cer and its precursor (DHC) accumulates in brain and CSF. Cer can also be generated in the lysosome by the action of acid sphingomyelinase. The plasma membrane neutral sphingomyelinase (NSMase2) is regulated by __________________ 2015 Transactions of the American Society for Neurochemistry ®
12
S04 Frontiers of Neurolipids palmitoylation and in various stress situations, the Cer generated by NSMase2 in turn activates PP2A phosphatase and inhibits Akt pathways leading to activation of caspase-3, p53, reduced SIRT-1 expression and cell death. Thus the anti-metastatic drug 4MU induces cell death and inhibits migration and invasion in gliomas by activating NSMase2 at the cell surface, increasing Cer and inactivating hyaluronan synthase2. In contrast, cells lacking NSMase2 show reduced Cer and increased hyaluronan synthesis. Cer is further metabolized to sphingosine and then to sphingosine-1-phosphate (S1P) by the action of kinases SK1 and SK2. S1P is considered to be protective to the brain and is reduced in stroke and MS lesions. S1P can be recycled back to sphingosine and then reacylated to ceramide, the ceramide synthase (mainly CerS2) being inhibited by neurotoxins such as Fumonisin B1. The use of MS/MS has greatly increased our ability to track these changes but many questions about ceramide homeostasis in the brain remain to be answered. (Supported by USPHS Grants NS-36866-40 to GD.
S04-04 CERAMIDE-ENRICHED COMPARTMENTS IN NEURAL STEM CELL DIFFERENTIATION AND NEURODEGENERATION Erhard Bieberich Georgia Regents University, Department of Neuroscience and Regenerative Medicine, Augusta, USA Ceramide is a remarkable sphingolipid in that it organizes membrane microdomains and interacts with protein kinases and phosphatases, thereby eliciting cell signaling pathways regulating neural cell apoptosis, differentiation, cell polarity, and neurodegeneration. We hypothesized that these two functions of ceramide, structural and regulatory, combine the organization of structural cell signaling platforms by ceramide with its function as a specific cell signaling lipid. Using polyclonal antibodies, for the first time generated in our laboratory against specific ceramides, we found that C24:0/C24:1 ceramide was enriched in primary cilia of neural progenitors. Another ceramide species, C18:0 ceramide, was found to induce exosome formation and secretion in astrocytes exposed to A!1-42, a neurotoxic amyloid peptide in Alzheimer’s disease. C24:1 as well as C18:0 ceramide bound to atypical PKC" (aPKC"), a protein kinase C isoform regulating cell polarity. This data suggests that the interaction of C24:0/C24:1 ceramide and C18:0 ceramide with aPKC" (and other regulatory proteins in a ceramide-induced protein complex) is critical for the formation of cilia in neural stem cell differentiation and exosomes in neurodegeneration, respectively. Therefore, cilia and exosomes may constitute two compartments that are critically regulated by distinct ceramide species. Supported by NSF grant 112157 and NIH grant R01AG034389.
2015 Transactions of the American Society for Neurochemistry ®
S04-05 CELL DEATH SPREADING BY EXOSOME SHEDDING: THE ROLE OF CERAMIDE-ENRICHED SECRETED MICROVESICLES IN SYNERGISTIC CYTOKINE TOXICITY Narayan Bhat Medical Univ of South Carolina, Dept Neuroscience, Charleston, USA Ceramide occupies a central place in sphingolipid (SPL) metabolism and functions as an important intracellular signaling molecule to modulate a range of cellular processes. Recent findings have further demonstrated that it participates in ESCRT-independent formation of exosomes i.e., the extracellular microvesicles released by a number of cell types including oligodendrocytes, that are increasingly recognized for their role in inter-cellular communications: both physiological and pathological. While ceramide has been implicated as an intracellular regulator of cytokine- as well as stressor-induced cell death signaling in many cell types, its extracellular role, if any in cytokine-induced synergistic toxicity is unknown. Thus, TNF# and IFN$ exert marginal cytotoxicity individually but together induce marked cell death in different cell systems including oligodendrocyte lineage cells. It is likely that cytokine-exposed cells would release ceramide-laden exosomes that may promote the synergistic actions of the cytokine combination. The results from studies using a human oligodendrocyte cell line as a model lend support to this possibility. Thus, the exosomal preparations from TNF#-treated ‘donor’ cells, while being mildly toxic to fresh cultures, induce enhanced cell death in a fashion resembling the effect of cytokine combination when added to IFN$-primed target cultures. The basic SPL profiles of the secreted exosomes as determined by HPLC-MS/ MS showed that the cytokine treatment time-dependently induced the generation, in particular of C24:1-, C24- and C18-Ceramide species; C24:1-, C24-, and C16-dihydroCer species; and C16-, C24:1- and C24-Sphingomyelin species. Interestingly, the patterns of extracellular rather than intracellular ceramide production correlated with the synergistic cytokine toxicity. That exosome-associated ceramide contributes to the cytotoxic activity was supported by the observation that exogenous C6-Cer replicated, albeit partially, the cytotoxic effect of exosomes released from TNF#-treated cells on IFN$-primed targets. The results suggest that ceramide-laden microvesicles released from TNF#-exposed cells collaterally ‘spread’ the death signal(s) to IFN$-primed target cells to elicit a robust synergistic cell death response. It is likely that exosomes derived from stressed oligodendrocytes and/or their progenitors in vivo may similarly ‘broadcast’ the cell death signal under inflammatory demyelinating conditions.
13
S04 Frontiers of Neurolipids
S04-06 PACLITAXEL TREATMENT GENERATES NEUROTOXIC DEOXYSPHINGOLIPIDS Stefka Spassieva 1, Rita Kramer 1, Jacek Bielawski 2 Erhard Bieberich 3, Emily Kistner-Griffin 5, Drew Kornhauser 1 Thorsten Hornemann 4 1 Medical University of South Carolina, Medicine / Div. Hematology/Oncology, Charleston, USA 2 Medical University of South Carolina, Biochemistry and Molecular Biology, Charleston, USA 3 Georgia Regents University, Department of Neuroscience and Regenerative Medicine, Augusta, USA 4 University Hospital Zurich, Institute for Clinical Chemistry, Zurich, Switzerland 5 Medical University of South Carolina, Public Health Sciences , Charleston, USA A major dose-limiting side effect of chemotherapy is peripheral neuropathy. Currently, the mechanism of this neurotoxicity is poorly understood and there is no prevention or treatment available. In the __________________
2015 Transactions of the American Society for Neurochemistry ®
current study we tested whether a class of neurotoxic deoxysphingolipids is involved. Deoxysphingolipids are produced when the first enzyme of the sphingolipid de novo synthesis, serine palmitoyltransferase, utilizes L-alanine instead of L-serine as its amino acid substrate. The neurotoxicity of the deoxysphingolipids was shown before in hereditary sensory and autonomic neuropathy type I. Our results revealed that treatment of cells with paclitaxel, but not cisplatin (both associated with neuropathy) resulted in elevated cellular levels of deoxysphingolipids, and increased levels and activity of serine palmitoyltransferase. Moreover, our pilot study involving breast cancer patients receiving paclitaxel chemotherapy demonstrated a correlation between a specific deoxysphingolipid species and incidence and severity of peripheral neuropathy. In addition, our latest results with neurons showed that treatment with L-alanine derived deoxysphingolipids, but not Lserine derived regular sphingolipids or paclitaxel resulted in change of neuronal morphology and in neuronal damage. Taken together our results imply that deoxysphingolipids are likely molecular intermediates in paclitaxel-induced peripheral neuropathy.
14
S05 Novel Strategies in STEM CELL Therapy and Clinical Potentials S05-01 NEUROPROTECTION AND GENE-STEM CELL THERAPY FOR ISCHEMIC STROKE Koji Abe Okayama University, Neurology, Okayama, Japan Neuroprotection is essential for therapy in acute stage of stroke. Both NTFs and free radical scavenger can be such neuroprotective reagents with inhibiting death signals and potentiating survival signals under cerebral ischemia. For example, topical application of GDNF greatly reduced the infarct size and brain edema after middle cerebral artery (MCA) occlusion in rats. The reduction of the infarct size was not related to a change of cerebral blood flow (CBF), but was accompanied by marked reduction of positive cells for TUNEL and caspases in the affected area. Thus, GDNF showed a direct protective effect against ischemic brain damage, but not secondary by improving CBF. Edaravone, a free radical scavenger, is the first clinical drug for neuroprotection in the world which has been used from 2001 in most ischemic stroke patients in Japan. Edaravone scavenges hydroxyl radicals both in hydrophilic and hydrophobic conditions, and is especially useful in thrombolytic therapy with tissue plasminogen activator (tPA). Combination therapy of Edaravone with tPA greatly increased survival of stroke animals, reduced infarct size, and inhibited molecular markers of oxidative damage in lipid, protein and DNA. Use of Edaravone greatly reduced hemorrhagic transformation accompanied by tPA treatment,and may also extend therapeutic time window with tPA therapy for more than 3 hr in human stroke patients. It is important for regenerative therapy that the neural stem cells which are intrinsically activated or exogenously transplanted. To support stem cell migration, an artificial scaffold can be implanted to injured brain for promoting ischemic brain repair. Addition of NTFs greatly enhanced an intrinsic migration or invasion of stem cells into the scaffold, which could provide a future regenerative potential against ischemic brain damage at chronic stage. G-CSF may promote bone marrow cell migration into ischemic brain to reduce such a damage.
S05-02 CO-TRANSPLANTATION OF GLIAL RESTRICTED PROGENITOR AND SCHWANN CELLS IN PROMOTING REPAIR AFTER SPINAL CORD INJURY Xiaoming Xu Indiana University School of Medcine, Neurological Surgery, Indianapolis, USA Oligodendrocyte (OL) replacement can be a promising strategy for spinal cord injury (SCI) repair. However, the poor post-transplantation survival and inhibitory properties to axonal regeneration are two major challenges which limit their use as donor cells for repair of CNS injuries. Therefore, strategies aimed at enhancing the survival of grafted oligodendrocytes as well as reducing their __________________ 2015 Transactions of the American Society for Neurochemistry ®
inhibitory properties, such as the use of more permissive oligodendrocyte progenitor cells (OPCs), also called glial restricted precursor cells (GRPs), should be highly prioritized. Schwann cells (SCs) transplantation is a promising translational strategy to promote axonal regeneration after CNS injuries, partly due to their expression and secretion of multiple growth-promoting factors. Whether grafted SCs have any effect on the biological properties of grafted GRPs remains unclear. Here we report that either SCs or SC conditioned medium (SCM) promoted the survival, proliferation, and migration of GRPs in vitro. When GRPs and SCs were cografted into the normal or injured spinal cord, robust survival, proliferation, and migration of grafted GRPs were observed. Importantly, grafted GRPs differentiated into mature oligodendrocytes and formed new myelin on axons caudal to the injury. Finally, co-grafts of GRPs and SCs promoted recovery of function following SCI. We conclude that co-transplantation of GRPs and SCs, the only two kinds of myelin forming cells in the nervous system, act complementarily and synergistically to promote greater anatomical and functional recovery after SCI than when either cell type is used alone.
S05-03 MICRO RNA AND SIGNALING PATHWAYS IN STEM CELL THERAPY AFTER ISCHEMIC STROKE Michael Chopp 1 1 Henry Ford Hospital, Neurology, Research, Detroit, MI, USA 2 Oakland University, Physics, Rochester, MI, USA In this presentation, I will describe how multiple endogenous neurorestorative processes are stimulated after a stroke, and the means by which we can amplify these restorative processes with cell-based therapies. I will focus on the use of multipotent mesenchymal stromal cells (MSCs) for the treatment of stroke and neural injury. Exogenously administered cells communicate with and thereby activate parenchymal cells, primarily astrocytes, to generate proteins and trophic factors which promote the expression of interactive restorative processes, such as angiogenesis, neurogenesis and axonal outgrowth and synaptogenesis, and thereby, mediate neurological recovery post stroke. The entire central nervous system (CNS) is stimulated by the restorative cell-based therapy, and rewiring and remodeling of the hemisphere contralateral to the lesion as well as the spinal cord circuits and the cortical spinal tract (CST) ensue. The underlying molecular mechanisms that mediate these restorative processes and how the exogenously delivered cells communicate with parenchymal cells and alter parenchymal cell gene and protein expression to amplify neurorestoration will be discussed. microRNAs play a fundamental role in mediating a vast array of biological functions. I will describe how cell-based therapies transfer miRNAs via exosomes, and thereby target and alter parenchymal cells. Exosomes are small lipid microvesicles (~30-120 nm) that are active biological containers, which transport regulatory genes, lipids and proteins between cells, and form a major biological communication conduit. Exosomes __________________ 15
S05 Novel Strategies in STEM CELL Therapy and Clinical Potentials protect their cargo from degrading proteases and RNAses. If exosomes mediate the therapeutic response of cell-based therapies, then it may be reasonable to employ these exosomes without the parent cells to treat stroke, neurological disease and injury. I then demonstrate that treatment of stroke and traumatic brain injury with cell-free exosomes provides a potent therapeutic restorative effect. Cell-based therapies are therefore shown to amplify endogenous restorative processes and their ability to promote neurovascular remodeling and plasticity after stroke and neural injury may be mediated by the transfer of non-coding RNAs and proteins via exosomes.
S05-04 IPS CELL TRANSPLANTATION AND COMBINATIONAL THERAPY FOR THE REGENERATIVE TREATMENT AFTER ISCHEMIC STROKE Ling Wei Emory University, Anesthesiology/Neurology, Atlanta, USA Ischemic stroke is a leading cause of death and long-term disability. However, no treatment exists to repair damaged brain tissue. Inducible pluripotent stem (iPS) cells are a novel type of stem cell created by genetically reprogramming adult somatic cells into pluripotent cells which can then be differentiated into functional neurons. The transplantation potential and therapeutic benefits of __________________
2015 Transactions of the American Society for Neurochemistry ®
iPS cells however, is relatively unknown. Our research is to develop novel, safe and effective treatments for stroke by taking a comprehensive approach to promote cell’s ability to survive, migrate, differentiate and integrated in the lesion area after transplantation. Human or mouse pluripotent iPS cells were differentiated into neural progenitors and subjected to hypoxic preconditioning. This procedure significantly promoted cell survival and neuronal differentiation in vitro as well as after transplantation into the ischemic cortex. iPS cell-derived neural progenitors show the expression of trophic factors including VEGF, EPO, SDF-1, FGF, GDNF, and factors promoting neurogenesis or angiogenesis. The levels of these trophic factors is increased while the inflammatory factors are decreased in preconditioned cells. The non-invasive method of intranasal cell delivery was applied several days after stroke in adult or neonatal rodents. iPS cells were successfully differentiated into functional neurons in vitro and after transplantation into the ischemic brain. Stroke animals that received iPS cell transplantation showed increased BrdU-positive cells and increased BrdU/NeuN and BrdU/Collagen IV double-positive cells compared to stroke only rats. iPSC transplantation animals showed improved sensorimotor function. Transplantation of hypoxic preconditioned iPS cells may contribute additional trophic support to increase endogenous progenitor migration to the infarct, cell survival, and endogenous neurogenesis and angiogenesis. We conclude that the combination therapy using iPS cells can increase regenerative activities in the stroke brain and is a potential therapy for ischemic stroke.
16
S06 Autophagy in Neurodegeneration: Role in Disease and Therapy Target S06-01 CELL-PENETRATING LANTHIONINE KETIMINE DERIVATIVE ACTIVATES AUTOPHAGY THROUGH A NOVEL MECHANISM INVOLVING CRMP2 AND MTORC1 Kenneth Hensley University of Toledo, Pathology / Neuroscience, Toledo, USA Autophagy is a fundamental process for recycling macromolecules and whole organelles. Defects in autophagy are becoming associated with many neurodiseases and cancers. Autophagy is largely controlled through inhibition of its initiation, via the mammalian target of rapamycin-complex I (mTOR-Complex I or mTORC1). mTORC1 inhibition by the fungal product rapamycin is clinically important, but no endogenous mammalian small molecule regulator for mTORC1 has been reported. We now report that a cellpenetrating form of the natural sulfur amino acid metabolite, lanthionine ketimine (LK), activates autophagy in neuroblastoma and glioma cells through a mechanism that alters the subcellular localization of mTOR relative to other mTORC1 components on the lysosome. Current data suggests that LK or its ester, LKE, acts on the adaptor protein CRMP2 (collapsing response mediator protein2) to affect autophagy regulation through mTORC1 and possibly other means. These findings are discussed in context of other recent reports that the glutathione and LK-binding protein, LanCL1 (lanthionine synthase like protein-1) regulates neural redox status such that LanCL1 knockout causes neuroinflammation and neurodegeneration. The emerging relationship amongst LK, LanCL proteins and autophagy control proteins will be explored with an eye toward engaging this system for therapeutic purposes in neurodisease. Supported in part by grants from NIH (NS082283) and the Muscular Dystrophy Association (MDA217526).
S06-02 THE ELUSIVE ROLE OF AUTOPHAGY IN TRAUMATIC BRAIN INJURY Marni Harris-White 1, Aleksandra Poteshkina 1, Ming Johnson 1 Pirooz Eslami 1, Kalina Venkova-Hristova 2, Alexandar Hristov 2 Kenneth Hensley 2 1 Veterans Admin/Univ of Calif, Los Angeles, Dept of Medicine, Los Angeles, USA 2 University of Toledo, Dept of Pathology, Toledo, USA The role of autophagy in TBI is clouded by conflicting reports of autophagy being both detrimental and beneficial. Although there is substantial evidence that markers of autophagy are increased in both human and animal brains following TBI, it is not clear whether those changes result in functional autophagy. Protein quality control and degradation play important roles in CNS homeostasis. The fidelity of the autophagic process is particularly relevant to CNS cells, and in particular, post-mitotic neurons that utilize autophagy to selectively target misfolded or aggregated proteins and defective organelles for removal. We have recently demonstrated that the axonal scaffolding protein, CRMP2, is involved in autophagy as engineered knockdown of CRMP2 reduces autophagy flux. Further, __________________ 2015 Transactions of the American Society for Neurochemistry ®
Lanthionine ketimine ethyl ester (LKE), binds to CRMP2 and stimulates autophagy in mammalian CNS cells by inhibition of mTORC1. In this study, we utilized the mouse central fluid percussion model, a model of diffuse axonal injury. The progressive nature of DAI suggests that there is a period of time in which a pharmacological treatment might be effective to stabilize neuronal architecture, stimulate productive autophagy and allow repair mechanisms to function. To this effect, 30 minutes following a moderate TBI, and throughout the survival period, LKE was administered and mice subsequently evaluated for learning/memory impairments and biochemical and histological changes over a 5 week period.
S06-03 AUTOPHAGY-ENDOSOMAL-LYSOSOMAL PATHWAY DYSFUNCTION IN A!(1-42) DEPENDENT NEURODEGENERATION Paul Salvaterra 1, 2, Martha Magallanes 1, Saumya Srivastava 1 1 Beckman Research Institute, Neuroscience, Duarte, USA 2 Irell and Manella Graduate School of Bioscience, Neuroscience, Duarte, USA Dysfunctions of the autophagy and endosomal pathways have long been recognized as an underlying feature of many types of neurodegenerative diseases. The specifics of these dysfunction(s) as well as their relationships to various neurodegenerative disease phenotypes have in most cases not yet been clarified. Autophagy is an important catabolic system whose normal function is required for maintenance of neuronal homeostasis. Endosomal functions are varied and include important aspects of cellular signaling, molecular sorting, delivery of extracellular material to internal cellular compartments and more recently an appreciation of an intimate link with autophagy. The dynamic characterization of autophagy and endosomal pathways at the molecular and cellular level, while still incomplete, has established an important role for these membrane trafficking pathways as various cellular vesicles form, fuse, digest their contents and disappear. We have been exploring a Drosophila model of A!1-42 dependent chronic progressive neurodegeneration. Our results suggest that dysfunctional autophagy-endosomallysosomal (AEL) activity underlies A!1-42 dependent neurodegeneration. Experimentally, we directly express a secretory form of A!142 rather than APP, the normal source of A!1-42. Remarkably, many aspects of direct A!1-42 expression in fly neurons exhibit phenotypic similarities to pathological features of Alzheimer’s disease as well as mammalian models of Alzheimer’s. These include an early accumulation of AEL vesicles resulting from and specific to A!1-42 expression. As animals age, A!1-42 accumulates specifically within dysfunctional AEL vesicles where it exhibits plaque-like characteristics. Expression of A!1-40 even at high levels does not lead to significant neurodegeneration or other AD like phenotypes. Analysis of flies expressing non-secretory A!1-42 suggests that production through secretory routing is necessary to achieve maximum neurotoxicity. We recently built a new human embryonic stem cell model __________________ 17
S06 Autophagy in Neurodegeneration: Role in Disease and Therapy Target of direct A!1-42 expression and initial results using this system suggest the relevance of our Drosophila findings for studies designed to understand human Alzheimer’s-like neurodegeneration.
S06-04 AUTOPHAGOSOME BIOGENESIS IN PRIMARY NEURONS FOLLOWS AN ORDERED AND SPATIALLY REGULATED PATHWAY Sandra Maday, Erika Holzbaur University of Pennsylvania School of Medicine, Department of Physiology, Philadelphia, USA Autophagy is an essential lysosomal degradation pathway in neurons. Neuron-specific knockout of autophagy genes is sufficient to induce axonal degeneration and neuronal death in mouse models. Despite evidence that autophagy is critical for neuronal homeostasis, the mechanisms driving this process in neurons are poorly understood. In primary dorsal root ganglion (DRG) neurons, we have shown that autophagosomes are constitutively generated in the distal axon and undergo robust transport toward the cell soma. As they move proximally, autophagosomes mature into autolysosomes that more effectively catalyze cargo degradation. Here, we use dualcolor live-cell imaging to investigate the neuron-specific mecha__________________
2015 Transactions of the American Society for Neurochemistry ®
nisms of autophagosome biogenesis under basal conditions. Puncta positive for the autophagosome-assembly factors Atg13 and Atg5 appear almost exclusively in the distal axon and grow progressively in size. LC3 is recruited to these nascent autophagosomes, Atg13 and Atg5 dissociate, and the LC3 puncta then grow progressively into ring structures ~800 nm in diameter. Quantitative analysis of the temporal dynamics reveals that this ordered recruitment of assembly factors proceeds with stereotypical kinetics. During formation, we did not observe incorporation of plasma- or mitochondrial-derived membrane into nascent autophagosomes in the distal axon. Rather, autophagosomes are generated at subdomains of the endoplasmic reticulum positive for DFCP1 and distinct from ER exit sites. Remarkably, these biogenesis events are highly enriched in the distal axon, with rates of formation ~20-fold higher than observed along the mid-axon. Autophagosomes form less frequently in the cell soma or mid-axon, indicating that this process is spatially enriched in the distal axon, consistent with a highly compartmentalized pathway for constitutive autophagy in primary neurons. This overall paradigm of distal initiation followed by robust retrograde transport is not limited to developing DRG neurons and is also observed in synaptically-connected hippocampal neurons. We propose that distal enrichment of autophagosome formation facilitates the degradation of damaged mitochondria and long-lived cytoplasmic proteins that reach the axon tip via slow axonal transport.
18
S07 Central and Peripheral Inflammatory and Immune Responses in Parkinson's Disease S07-01 DESTRUCTIVE OR PROTECTIVE IMMUNITY IN PARKINSON'S DISEASE Howard Gendelman University of Nebraska Medical Center, Pharmacology and Experimental Neuroscience, Omaha, USA Adaptive immune responses during central nervous system injury affect the pathogenesis of Parkinson’s disease. Immune responses can exacerbate neurodegenerative activities or, alternatively, induce neuroprotective outcomes. The temporal mechanisms by which these responses occur begin through the emergence of antigen-specific CD4+ T cells. These cells readily traverse the blood brain barrier, engage resident glial cells then affect neuroinflammation and neuronal vitality. The functional endpoints of T cell-glial interactions are dependent on the numerical dominance and functional outcomes of effector or regulatory T cells. Migratory, regulatory and effector functions of these cells are triggered by misfolded and aggregated proteins and leading to altered neuronal behaviors and overt disease. How these events can be harnessed for therapeutic gain is being tested in our laboratories and at thte patient bedside.
S07-02 ROLES FOR MICROGLIAL LRRK2 EXPRESSION AND FUNCTION IN PARKINSON’S DISEASE Matt LaVoie Brigham and Women's Hospital and Harvard Medical School, Ann Romney Center for Neurologic Diseases, Boston, USA Autosomal dominant missense mutations in the multi-domain kinase LRRK2 are the most common genetic cause of Parkinson’s disease (PD). Pathogenic mutations are found in multiple functional motifs of the LRRK2 protein and thus likely exert a complex effect on its function. Biochemical evidence strongly indicate that the most common mutation (G2019S), which is found within the kinase domain, induces a gain-of-function increase in kinase activity while the impact of other mutations is less well understood. Likewise, neuronal phenotypes associated with mutant LRRK2 expression are mild and have not been informative with respect to the primary pathologic features of PD, Lewy body inclusions containing alphasynuclein and the premature degeneration of the dopaminergic neurons of the substantia nigra. Emerging data indicate a potential role for LRRK2 in autoimmune disorders and clearly demonstrate high levels of LRRK2 expression in a variety of immune cell types. To examine the functional role of LRRK2 in the immune system, we studied endogenous LRRK2 expression in a variety of primary monocyte cultures as well as monocytic cell lines. Consistent with prior reports, we found that selective TLR activation resulted in acute changes in LRRK2 phosphorylation. We extended these studies to demonstrate that activation of endogenous LRRK2 was associated with its dimerization and translocation to membranous compartments of the cell. We found little to no effect of LRRK2 on __________________ 2015 Transactions of the American Society for Neurochemistry ®
monocyte phagocytosis or ROS secretion. However, data suggested a critical role for LRRK2 kinase activity in lysosomal degradation associated with autophagy, and that LRRK2 was recruited in part to autophagolysosome membranes. Additional data on the relevance of these findings to the homeostatic control of alpha-synuclein metabolism in PD will be discussed.
S07-03 CRITICAL GASTROINTESTINAL FUNCTIONS AND DISEASE MARKERS IN PARKINSON'S DISEASE Kathleen Shannon Rush Medical College, Department of Neurological Sciences, Chicago, USA Parkinson’s Disease (PD) is a common neurodegenerative synucleinopathy of the elderly with genetic and environmental determinants. Diagnosis rests on the presence of a motor syndrome that reflects a burden of pathology that has accumulated over decades. Clinical trials over the past 30 years have failed to produce a single disease modifying agent, and there is a critical need to develop biomarkers to both increase diagnostic certainty and allow earlier diagnosis, and to identify new therapeutic targets that are relevant to disease mechanisms. Recent epidemiological and clinicopathological studies suggest PD may originate in the gastrointestinal (GI) tract, and that neuroinflammation may be an important driver of disease pathology in PD. The GI tract represents a very large surface that acts as a selective barrier to toxins and pathogens, has important function in innate immunity, and has a robust population of neurons in close proximity to the intestinal wall. Enteric neurons are connected to the dorsal motor nucleus of the vagus nerve, a site of early central nervous system pathology in PD. We have studied critical gastrointestinal functions and biomarkers of synuclein pathology and neuroinflammation in a group of PD and control subjects. Compared to controls, PD subjects show: (1) increased intestinal permeability (absorption of oral sugar load); (2) disruption of tight junction binding proteins ZO-1 and Claudin-1 (immunohistochemistry in colon tissue); (3) exposure to bacterial antigens and the bacterial endotoxin lipopolysaccharide (immunohistochemistry studies of colon biopsy tissue, plasma lipopolysaccharide binding protein changes); (3) inflammatory changes (nitrotyrosine immunostaining, upregulation of inflammatory cytokines); (4) fecal microbiome that differs from control stool (analysis of 16S rRNA gene amplicons); and (5) alpha-synuclein (AS) accumulation in colonic submucosa (immunohistochemistry). AS accumulation can be seen even in tissue samples taken several years prior to the first motor experience of PD. This presentation will review our clinical studies in PD and control subjects and discuss the potential of these biomarkers to aid our understanding of PD pathogenesis as well as to guide earlier diagnosis and future therapeutic strategies.
19
S07 Central and Peripheral Inflammatory and Immune Responses in Parkinson's Disease
S07-04
S07-05
A COMMON GENETIC VARIANT IS ASSOCIATED WITH ALTERED MHC-II EXPRESSION AND SYNERGIZES WITH PYRETHROID EXPOSURE TO INCREASE PD RISK Malu Tansey 1, George T. Kannarkat 1, Darcie A. Cook 1 Jae-Kyung Lee 1, Jianjun Chang 1, Jaegwon Chung 1 Elizabeth Sandy 1, Kimberly C. Paul 2, Beate Ritz 2 Jeffrey Bronstein 3, Stewart Factor 4, Jeremy M. Boss 5 1 Emory University, Physioloy, Atlanta, USA 2 UCLA, Epidemiology, Los Angeles, USA 3 UCLA, Neurology, Los Angeles, USA 4 Emory University, Neurology, Atlanta, USA 5 Emory University, Microbiology and Immunology, Atlanta, USA
INHIBITION OF THE JAK/STAT PATHWAY PROTECTS AGAINST ALPHA-SYNUCLEIN-INDUCED NEUROINFLAMMATION AND DOPAMINERGIC NEURODEGENERATION Etty Benveniste Univ Alabama at Birmingham, Dept Cell, Developmental & Integrative Biology, Birmingham, USA
Many genome-wide association studies have implicated the major histocompatibility complex class II (MHC-II) in risk for idopathic Parkinson’s disease (PD). Specifically, the common non-coding single nucleotide polymorphism (SNP) rs3129882 in HLA-DRA implicates regulation of antigen presentation as a potential mechanism by which immune responses link genetic susceptibility to environmental factors in conferring lifetime risk for PD. A heightened baseline expression and inducibility of MHC class II molecules in B cells and monocytes from peripheral blood of healthy controls and PD patients was associated with homozygosity for G at this SNP. In addition, the risk conferred by this SNP synergizes with exposure to pyrethroids, a commonly used class of insecticide, thereby indicating a novel gene-environment interaction that increases risk for PD. These novel findings suggest that the MHC-II locus may increase susceptibility to PD through presentation of pathogenic, immunodominant antigens and/or a shift toward a more pro-inflammatory CD4+ T cell response and that pyrethroid exposure can modulate the antigen presentation process to the adaptive immune system.
__________________
2015 Transactions of the American Society for Neurochemistry ®
Parkinson’s Disease (PD) is an age-related, chronic neurodegenerative disorder. At present, there are no disease-modifying therapies to prevent PD progression. Activated microglia and subsequent neuroinflammation are associated with the pathogenesis and progression of PD. Accumulation of a-synuclein (a-SYN) in the brain is a core feature of PD and leads to microglial activation, inflammatory cytokine/chemokine production and neurodegeneration. Given the importance of the JAK/STAT pathway in activating microglia and inducing cytokine/chemokine expression, we investigated the therapeutic potential of inhibiting the JAK/STAT pathway using the JAK1/2 inhibitor, AZD1480. We utilized an in vivo rat model of PD induced by viral overexpression of a-SYN. We find that AZD1480 treatment inhibits a-SYN-induced neuroinflammation by inhibiting microglia activation and CD3+ T-cell infiltration, and neurodegeneration by preventing the degeneration of dopaminergic neurons in vivo. Numerous genes involved in cellcell signaling, nervous system development and function, inflammatory diseases, neurological diseases and inflammatory processes are enhanced in the substantia nigra of rats with a-SYN overexpression, and inhibited upon treatment with AZD1480. In vitro, AZD1480 inhibits a-SYN-induced MHC Class II expression and inflammatory gene expression in microglia and macrophages by reducing STAT1 and STAT3 activation. These results indicate that inhibiting the JAK/STAT pathway can prevent neuroinflammation and neurodegeneration by suppressing activation of innate and adaptive immune responses to a-SYN in this PD model. Furthermore, this suggests the feasibility of targeting the JAK/STAT pathway as a neuroprotective therapy for neurodegenerative diseases.
20
S08 Wiring of the Nervous System: From Molecular Mechanisms to Complex Behaviors S08-01 A MOLECULAR PATHWAY REGULATING AXOGENESIS Anthony Barnes Oregon Health and Science Unvi, Pediatrics, Portland, USA The establishment of long-range connectivity represents one of the greatest challenges faced by the developing nervous system. The axons that will ultimately constitute these neural networks are specified during neuronal differentiation. While the phenomenology of this process has been described in some detail for particular cell types in the embryonic brain, much remains to be learned about the cell biological events that underlie this critical event in neuronal morphogenesis. It is now well established that particular signal transduction pathways are required for axogenesis, but it is less clear how these pathways are regulated and how they cross talk with one another. One of the requisite signaling elements of axon formation is the tumor suppressor protein kinase LKB1. We, along with others, have shown that LKB1 contributes to multiple stages of axon development including axon specification, branching and synaptogenesis. Here, we focus on LKB1 regulation via its unconventional activation scheme during axogenesis in the developing cerebral cortex. Specifically, we will describe our genetic and biochemical analysis of the role played by two related pseudokinases in this process. Our data indicate these proteins, STRAD-alpha and STRAD-beta, comprise an essential regulatory node for LKB1 during this early and essential step of neural development.
S08-02 COMBINATORY CIS AND TRANS RET SIGNALING CONTROLS THE SURVIVAL AND CENTRAL PROJECTION GROWTH OF RAPIDLY ADAPTING MECHANORECEPTORS Wenqin Luo University of Pennsylvania, Neuroscience, Philadelphia, USA RET can be activated in cis or trans by its co-receptors and ligands in vitro, but the physiological roles of trans signaling are unclear. Rapidly adapting (RA) mechanoreceptors in dorsal root ganglia (DRGs) express Ret and the co-receptor Gfr!2 and depend on Ret for survival and central projection growth. We found that Ret and Gfr!2 null mice display comparable early central projection deficits, but Gfr!2 null RA mechanoreceptors recover later. Loss of Gfr!1, the co-receptor implicated in activating Ret in trans, causes no significant central projection or cell survival deficit by itself, but Gfr!1;Gfr!2 double nulls phenocopy Ret nulls. Finally, we demonstrate that RET in RA mechanoreceptors is activated by GFR!1 in trans in DRG explants. Taken together, our results suggest that trans and cis Ret signaling function in the same developmental process __________________
2015 Transactions of the American Society for Neurochemistry ®
and that the availability of both forms of activation likely enhances but not diversifies Ret signaling.
S08-03 ROLES FOR NEUROPILIN 2 SIGNALING IN HIPPOCAMPAL- AND CORTICO-STRIATAL-DEPENDENT LEARNING AND MEMORY Tracy Tran 1, Matthew Gavin 1, Michael Shiflett 2 1 Rutgers University, Biological Sciences, Newark, USA 2 Rutgers University, Psychology, Newark, USA The establishment of neuronal morphology, which is crucial for the proper wiring of neural circuits, is essential for complex behavior and cognitive function. However, the identity and function of extrinsic cues that control the development of neuronal morphology and synaptic connections leading to the activation of neural circuitry involved in complex behavior is poorly understood. Semaphorins play a key role in synapse refinement in the mammalian nervous system, and have been implicated in a number of neurological disorders. However, the effects on behavior and mental function of dysregulated Sema3F-Nrp2 signaling have not been addressed. Previously, we demonstrated that the class 3 semaphorin 3F (Sema3F) signaling through Neuropilin 2/Plexin-A3 (Nrp2/PlexA3) holoreceptor complex in vivo to restrain apical dendritic spine morphogenesis of cortical pyramidal and hippocampal neurons during postnatal development and mediates excitatory synaptic transmission. This study is the first behavioral investigation of mice harboring a mutation of the Nrp2 gene. Given that loss of Nrp2 signaling alters cortical and hippocampal synaptic organization, we investigated the performance of Nrp2-/- mice on learning and sensorimotor function that are known to depend on cortical and hippocampal circuitry. Nrp2-/- mice showed striking impairments in object recognition memory and preference for social novelty compared to age-matched controls. Additionally, Nrp2-/mice displayed impaired motor function in the rotarod test and in observations of grooming behavior. Exploration of novel olfactory sensory stimuli and nociception were unaffected by the loss of Nrp2. Furthermore, we have results showing Nrp2-/- animals have abnormal cortico-striatal circuit activity. Currently, we are investigating the role of Sema3F-Nrp2/PlexA3 signaling in corticostriatal circuit function, which is known to underlie goal-directed learning and behavior. To determine whether Sema3F-Nrp2/PlexA3 signaling alters goal-directed learning, we employ instrumental learning paradigms, in combination with anatomical and electrophysiological analyses in mutant mice deficient in Sema3F signaling. Overall, our results suggest that loss of Nrp2 leads to aberrant processing within hippocampal and cortico-striatal networks that may contribute to neurodevelopmental disease mechanisms.
21
S08 Wiring of the Nervous System: From Molecular Mechanisms to Complex Behaviors
S08-04 EXTRINSIC MECHANISMS GOVERNING INJURY-INDUCED AXON DEGENERATION Christopher Deppmann University of Virginia, Department of Biology, Charlottesville, USA Neuronal axon degeneration is a hallmark of all neurodegenerative disorders including Alzheimer’s, Luo Gehrig’s, and Huntington’s disease, as well as neural injury (e.g. stroke, traumatic brain injury and spinal cord injury). Moreover, loss of axons and synapses via degeneration is thought to be a main cause of cognitive decline, movement disorders, and paralysis associated with these pathologies. Despite their importance as therapeutic targets, very little is known about the mechanisms underlying axon loss. Beyond __________________
2015 Transactions of the American Society for Neurochemistry ®
pathological situations, axon degeneration is also an important particular aspect of developmental degeneration that has yet to be discovered in injury-induced degeneration is the ability to respond to axon derived extrinsic factors. In this talk I examine whether extrinsic initiators of developmental axon degeneration programs are also used to coordinate injury-induced axon degeneration (also known as Wallerian degeneration). In particular, I examine family members of tumor necrosis factor receptor (TNFR), which are known to be responsible for developmental neuron death and axon degeneration. I will present preliminary evidence that these receptors do indeed coordinate injury-induced axon degeneration in tissue culture as well as in mice. These are the first receptors discovered, that when lost, prevent Wallerian degeneration.
22
S09 Astrocytes as Obligatory PARTNERS in Purinergic and Glutamatergic Neurotransmission S09-01
S09-03
ASTROCYTES AS OBLIGATORY PARTNERS IN PURINERGIC AND GLUTAMATERGIC NEUROTRANSMISSION Alexei Verkhratsky The University of Manchester, Faculty of Life Sciences, Manchester, United Kingdom
ASTROGLIAL GLUTAMATE TRANSPORT: AT THE INTERSECTION OF EXCITATORY SIGNALING AND BRAIN ENERGETICS Michael Robinson, Joshua Jackson, John O'Donnell Brendan Whitelaw, Elizabeth Krizman University of Pennsylvania/ChildrenÕs Hospital of Philadelphia, Pediatrics/Pharmacology, Philadelphia, USA
Glutamate and ATP and adenosine are the most widespread neurotransmitters in the central nervous system (CNS), which are released from neurons and astroglial cells through several mechanisms that include regulated exocytosis and by transmembrane diffusion via several types of ion channels. Biological actions of glutamate and ATP are mediated through activation of multiple receptors represented by ligand-gated ion channels and G-protein coupled receptors. Astrocytes are central of glutamate metabolism in physiological and pathological settings, accumulating glutamate through dedicated transporters and supplying neurons with glutamine that acts as a glutamate/GABA precursor. Astrocytes express multiple receptors to ATP and ATP regulates numerous homeostatic and defensive functions of astroglia.
S09-02 ON VESICULAR FUSIONS IN ASTROCYTES: SINGLE VESICLE/MOLECULE APPROACHES Vladimir Parpura University of Alabama, Department of Neurobiology, Birmingham, USA Ca2+-dependent release of various transmitters from astrocytes is mediated by regulated exocytosis. These glial cells express the protein components of the SNARE complex, including synaptobrevin 2, syntaxin and SNAP-23, but not neuronal SNAP-25. Using astrocytes expressing synapto-pHluorin, exocytotic sites can be fluorescently imaged. Fusions of synapto-pHluorin labeled vesicles with the plasma membrane can be observed using total internal reflection fluorescence microscopy; the time course of fusion events (burst vs. sustained), their type (“kiss-and-run” vs. full fusion) and spatial relationship between different fusion sites will be discussed. Single molecule investigations of the SNARE complex using force spectroscopy show that the ternary complexes containing SNAP-23 have a shorter spontaneous lifetime than those containing SNAP25B. A model which considers the geometry of the vesicle approach to the plasma membrane with favorable energies and stability as the basis for the preferential usage of the parallel (as opposed to the anti-parallel) ternary SNARE complex in exocytosis is presented. It appears that the spatio-temporal characteristics of astrocytic exocytosis might be, in part, due to the intrinsic properties of the ternary SNARE complex in astrocytes.
2015 Transactions of the American Society for Neurochemistry ®
The glutamate transporters, GLT-1 and GLAST, are found on the fine processes of astrocytes and mediate the bulk of glutamate uptake activity in the mammalian brain. Astrocytes have several functions. In addition to clearing amino acid neurotransmitters, they are responsible for import of most of the substrates that are used to generate ATP in the brain. It has been assumed by many in the field that the fine astrocytic processes are too small in diameter to accommodate mitochondria. In organotypic hippocampal slices, we have selectively transduced astrocytes with a number of different proteins to measure mitochondrial occupancy, mitochondrial mobility, co-localization of mitochondria with glutamate transporters, and Ca2+ signaling. We find that mitochondria are present throughout astrocytic processes. In fact, mitochondria are frequently found in the finest tips of astrocyte processes that we can resolve with conventional confocal microscopy. We show that mitochondrial mobility in astrocytic processes is regulated by neuronal activity in a manner that is dependent upon glutamate transport and an influx of Ca2+ though Na+/ Ca2+ exchange. We have also developed evidence that neuronal activity immobilizes mitochondria near glutamate transporters that cluster at excitatory synapses. Finally we find that stationary mitochondria shape spontaneous calcium signals within astrocytic processes. These results combined with those of several other groups suggest that glutamate transporters functionally couple to/co-compartmentalize with glycolytic enzymes and mitochondria, placing these transporters at the intersection of excitatory signaling and brain energetics.
S09-04 GLUTAMATE AS A KEY ENERGY SUBSTRATE FOR SUPPORTING ASTROCYTIC GLUTAMATE TRANSPORT AND NEURONAL-GLIAL INTERACTIONS Mary McKenna Univ Maryland School of Medicine, Dept Pediatrics and Program in Neuroscience, Baltimore, USA A key function of astrocytes is disposal of glutamate from the synaptic cleft after depolarization to maintain the low resting concentration of glutamate required for continuing neurotransmission. Evidence from in vitro and in vivo studies demonstrates that glutamate is used by astrocytes as a fuel for oxidative energy metabolism. The ability to harvest the energy from the carbon skeleton of glutamate provides astrocytes with a mechanism to offset the high ATP cost of glutamate uptake from the synaptic cleft. The complete oxidation of glutamate requires metabolism of carbons via the pyruvate recycling pathway. Glutamate is preferentially __________________ 23
S09 Astrocytes as Obligatory PARTNERS in Purinergic and Glutamatergic Neurotransmission oxidized for energy by astrocytes even when other energy substrates including glucose, glutamine, lactate or 3-hydroxybutyrate are present. Other substrates can facilitate the uptake and oxidative metabolism of glutamate by astrocytes, and oxidation of glutamate for energy spares glucose and other substrates. Our data provide evidence supporting the importance of glutamate metabolism in __________________
2015 Transactions of the American Society for Neurochemistry ®
astrocytes and provides further evidence of multiple compartments of TCA cycle activity in cultured astrocytes. Utilization of energy substrates and compartmentation of metabolism changes during development and is different in astrocytes and synaptic terminals. Supported by NIH grant 5P01-HD016596.
24
S10 Neuropsychiatric Disorders: New Mechanisms and Models S10-01 CIRCADIAN RHYTHM DISRUPTION IN A MOUSE MODEL OF RETT SYNDROME Dawn Loh University of California - Los Angeles, Psychiatry and Biobehavioral Sciences, Los Angeles, USA Sleep-wake cycle disruptions are common amongst patients with neuronal and psychiatric disorders, resulting in a negative quality of life for the patients. Mouse models of neuropsychiatric diseases mirror such sleep-wake and circadian rhythm disruption, suggesting that the underlying genetic mutations affect the circadian system. One such disease with a known genetic cause is Rett syndrome (RTT), in which mutations of the X-linked Mecp2 gene lead to neurodevelopmental problems and sleep disorders. In the mouse, the Mecp2 gene is highly expressed in the suprachiasmatic nucleus (SCN), the master pacemaker of the circadian system. We explored possible circadian dysfunction in a mouse model of RTT as a strategy to understanding the sleep deficits in RTT patients. Hemizygous MeCP2 mutant mice exhibit decreased amplitude and precision of wheel running rhythms, and altered temporal patterning of sleep. We observed a reduction in the amplitude of rhythms driven by the circadian gene, Period2, in the SCN of mutants, suggesting that MeCP2 plays a role in maintaining the rhythms in molecular clockwork. Furthermore, the neural activity rhythms in the SCN are also affected, suggesting that MeCP2 is an integral component of the SCN timing mechanism. Examination of the molecular oscillator in individual cells confirmed a direct impact on the circadian gene expression in fibroblasts from hemizygous mutants and RTT patients with two different mutations in MeCP2. Finally, we tested the hypothesis that further circadian disruption could alter disease phenotype. Prior work has shown that weekly shifts of the light/dark cycle mimicking jet lag are very disruptive to the circadian system and increase mortality in aged mice. We found significant acceleration in mortality for the MeCP2 mutant mice subjected to chronic jet lag. These results raise the possibility that destabilizing the circadian system is a risk factor for vulnerable RTT patients while a stabilized circadian system may improve the quality of life for the patients.
S10-02 A NATURAL PRODUCT AS A TOOL TO EXPLORE NOVEL THERAPEUTIC TARGETS FOR STRESS-INDUCED IMMUNE CHANGES AND DEPRESSIVE BEHAVIORS Atsushi Kamiya Johns Hopkins University School of Medicine, Department of Psychiatry and Behavioral Sciences, Baltimore, USA There are a number of clinically effective treatments for stress-associated mental disorders, such as depression. Nonetheless, a large __________________
2015 Transactions of the American Society for Neurochemistry ®
portion of those afflicted exhibit treatment-resistance to first-line treatments, which calls for novel interventions based on pathological mechanisms. Alterations in immune and inflammation processes, including changes in expression of pro- and anti-inflammatory cytokines are observed in patients with depression. Notably, many natural products used in traditional Eastern medicine have been shown to have immunomodulatory properties. In fact, traditional Eastern medicine has been empirically used for treatment of depressive symptom over the past centuries. Nonetheless, there is almost no mechanism-based evidence for the effectiveness of traditional herbal medicines in the treatment of depression. Thus, natural products may be useful tools to uncover novel therapeutic targets of depression by deciphering their mechanisms utilizing modern neuroscience approaches. In this talk, I will present our studies to explore the antidepressant effect of pachyman, a polysaccharide extracted from Poria cocos used as a main ingredient in many traditional Eastern medicines. We examine the effects of pachyman on stress-induced immune changes and depressive behaviors to define its mechanisms of action, which may provide a basis for identifying novel drug targets for the prevention and treatment of depression and related mental conditions.
S10-03 GENETIC RISK FACTORS AND BRAIN MALDEVELOPMENT Cristina Ghiani 1, Frank Lee 2, Achilles Aiken 3, Kelly Chan 3 Christina Falcone 3, Chanelle Sy 3, Esteban Dell' Angelica 2 1 University of California Los Angeles, Departments of Psychiatry and Pathology, Los Angeles, USA 2 University of California Los Angeles, Department of Human Genetics, Los Angeles, USA 3 University of California Los Angeles, Department of Psychiatry, Los Angeles , USA Schizophrenia is a major psychiatric disease perhaps triggered by a combination of modest effects caused by susceptibility genes. Thus, functional studies on the products of such susceptibility genes are necessary to demonstrate the “biological plausibility” of their genetic association with the disease. We have previously shown that dysbindin, encoded by the susceptibility gene for schizophrenia, DTNBP1, is a stable component of a multimeric complex named BLOC-1 (Biogenesis of Lysosome-related Organelles Complex 1), of which expression levels are developmentally regulated and it has been implicated in intracellular protein trafficking. Here, we will provide evidence that lack of functional BLOC-1 alters gene expression in the developing central nervous system of homozygous mutant mice, eliciting defective neural cell maturation and malformation of relevant brain areas such as the hippocampus, leading to faulty brain wiring and behavioural abnormalities.
25
S10 Neuropsychiatric Disorders: New Mechanisms and Models
S10-04 MOLECULAR AND GENETIC STUDIES OF A SCHIZOPHRENIA SUSCEPTIBILITY INTERACTOME Victor Faundez 1, Avanti Gokhale 1, Ariana Mullin 1 Stephanie Zlatic 1, Skye Comstra 1, Amelia Burch 1, Arielle Lewis 1 Subhabrata Sanyal 2 1 Emory University, Cell Biology, Atlanta, USA 2 Biogen-Idec, Biogen, Cambridge, USA Dysbindin is a schizophrenia susceptibility factor and subunit of the BLOC-1 complex required for lysosome-related organelle biogenesis and in neurons synaptic vesicle assembly, neurotransmission, and plasticity. Protein networks, or interactomes, downstream of dysbindin-BLOC-1 remain unexplored. We conducted a proteome-wide search for polypeptides whose content is sensitive to dysbindin-BLOC-1 down-regulation. We identified components of the vesicle fusion machinery including the N-Ethylmaleimide Sensitive Factor (NSF) as factors down regulated in dysbindinBLOC-1 deficiency. Human dysbindin-BLOC-1 associates with NSF and both proteins colocalized in a Drosophila model synapse. We examined the role for NSF in dysbindin-BLOC-1-dependent synaptic homeostatic plasticity in Drosophila. As previously described, we found that mutations in dysbindin precluded homeostatic synaptic plasticity elicited by acute blockage of post-synaptic receptors. This dysbindin-mutant phenotype is rescued either by presynaptic expression of dysbindin or Drosophila NSF. Our results demonstrate that dysbindin-BLOC-1 expression defects result in altered cellular content of proteins of the vesicle fusion apparatus and therefore influence synaptic plasticity.
S10-05 DURABLE CONSEQUENCES OF SOCIAL ADVERSITY IN ADOLESCENCE: NEUROBIOLOGICAL AND BEHAVIORAL OUTCOMES FOLLOWING ISOLATION Shannon Gourley Emory University, Pediatrics, Atlanta, USA Early-life social adversity is a risk factor for multiple psychopathologies with neurodevelopmental origins, including adolescentemergent depression and schizophrenia. Neurobiological factors associated with mental health outcomes are still being determined, however. To better understand the long-term consequences of social adversity during adolescence, we isolated female mice from postnatal day 31-60, then re-housed them in enriched social groups. We first found that a history of social isolation persistently decreased expression of CNPase, an oligodendrocyte marker, in several cortical and hippocampal regions. We also discovered that social isolation results in glucocorticoid insufficiency. Glucocorticoid receptor occupancy plays a key role in postnatal neural maturation, and accordingly, deep layer prefrontal cortical dendritic spines retained an immature morphology and density in adult mice with a history of social isolation. Social isolation also induced anhedoniclike behavior, increased vulnerability to stress-related amotivation, and caused a bias towards reflexive habits at the expense of goal__________________
2015 Transactions of the American Society for Neurochemistry ®
directed decision-making strategies. Given increasing concern regarding the utility and safety of classical antidepressants in adolescent humans, we next tested the utility of a Rho-kinase inhibitor in reversing maladaptive outcomes. This inhibitor, fasudil, but not fluoxetine, corrected decision-making strategies and had durable antidepressant-like properties. These findings suggest that Rho-kinase, or other regulators of postnatal neural maturation, may be promising targets in the development of novel treatments for adolescent-emergent psychopathologies.
S10-06 EFFECTS OF FINGOLIMOD ADMINISTRATION IN DYSBINDIN DEFICIENT MICE: A GENETIC MODEL FOR COGNITIVE DEFICITS Antonieta Lavin, Darius Becker-Krail MUSC, Neuroscience, Charleston, USA Impairments in information processing, particularly social information are some of the most commonly noted deficits shared by neurodevelopmental and neurospyschiatic disorders. Dysbindin is part of the Biogenesis of lysosome-related organelle complexes 1 (BLOC-1 complex). This complex has been related to multiple cellular functions including synaptic vesicle dynamics. Using dysbindin deficient mice as a tool to study the role of glutamate in cognitive disabilities, we found that these mice show decreases in the replenishment of the ready releasable pool of synaptic glutamate vesicles in the PFC, decreases in quantal size, decreases in probability of glutamate release, deficits in the rate of endo-and exocytosis, diminished expression of L- and N-type Ca2+ channels and deficits on working memory and social interaction tasks. Fingolimod, a drug used to treat multiple sclerosis and Rhett syndrome, is known to increase endogenous levels of brain derived neurotrophic factor (BDNF), and in turn, it has been shown that BDNF increases N-type Ca2+ channels. To explore a potential mean of restoring glutamate release, and perhaps improving the cognitive deficits, we investigate the effects of fingolimod using a dysbindin mutant mouse. The mice were divided into two groups: saline or fingolimod treatment. We assessed sociability and memory across three genotypes (WT, HET and MUT) using the social choice and preference for social novelty tasks. For both groups, we assayed BDNF concentration in PFC homogenate using an ELISA, and analyzed levels of intracellular [Ca2+] in a crude PFC synaptosome preparation using a Ca2+ assay. Relative to WT mice, dysbindin MUT mice demonstrated impairments in novel social interaction, deficits in memory as measured through preference for social novelty, and a lower presynaptic intracellular [Ca2+]. Fingolimod treated MUT mice show significantly improved social interaction with novel mice, significantly improved memory as measured through preference for social novelty, higher [BDNF], and an increase in presynaptic intracellular [Ca2+]. These results show promise for counteracting social and cognitive deficits associated with schizophrenia, and may illuminate the possible role of dysbindin in symptom pathogenesis.
26
S11 Dissecting the Molecular Mechanisms of Acute Neurologic Injury S11-01
S11-02
INTRA-AXONAL PROTEIN SYNTHESIS AS A MEDIATOR OF LONG-RANGE NEURODEGENERATIVE SIGNALS Ulrich Hengst Columbia University, Pathology & Cell Biology / Taub Institute, New York, USA
THE ROLE OF BETA AMYLOID AND TAU IN THE RESPONSE TO TRAUMATIC BRAIN INJURY Russell Nicholls 1, 2, Edward Vogel 3, Christopher Hue 3 Agnieszka Staniszewski 1, 2, Kesava Asam 1, 2, Siqi Liang 1, 2 John Crary 1, 2, Barclay Morrison 3, Ottavio Arancio 1, 2 1 Columbia University, Department of Pathology and Cell Biology, New York, USA 2 Columbia University, The Taub Institute, New York, USA 3 Columbia University, Department of Biomedical Engineering, New York, USA
As the spread of Alzheimer’s disease (AD) through the brain is determined by connectivity rather than proximity it is important to investigate how neurodegenerative signals in the periphery of neurons change intra-neuritic signaling pathways and how these signals are communicated to the neuronal cell bodies. We discovered a novel signaling mechanism for the long-range, retrograde spread of neurodegeneration in response to soluble oligomeric !-amyloid1-42 (A!). We observed a strong activation of localized protein synthesis within axons when we applied A!1-42 locally to axons, and inhibition of intra-axonal translation or retrograde transport of proteins to the cell body completely prevented cell death in response to A!1-42. Axonal application A!1-42 changed the composition of the axonal transcriptome through recruitment of a defined set of mRNAs, among them the transcript of the transcription factor ATF4. ATF4 was locally synthesized in response to local application of A!1-42 and retrogradely transported to the neuronal soma where it caused transcriptional changes, including CHOP production, leading to cell death. When we selectively depleted axons of Atf4 mRNA we could prevent A!1-42-dependent neurodegeneration both in cultured hippocampal neurons and more importantly in a mouse model for amyloidopathy. This finding identifies intraaxonal ATF4 synthesis as a central component of the signaling pathways transmitting neurodegenerative signals across brain regions. The patho-physiological relevance of our findings is further emphasized by the fact that we observed both ATF4 protein and mRNA with increased frequency in axons in those regions of the brain of AD patients that are thought to be especially vulnerable in AD, such as the entorhinal cortex and the subiculum. Axon-to-soma communication via locally synthesized transcription factors is reminiscent to axonal injury-induced signaling pathways, suggesting that localized protein synthesis might be a general response mechanism to injuries insults. Further, the synthesis of ATF4 is an indicator for the activation of cellular stress response pathways, indicating a functional involvement of the UPR or the integrated stress response (ISR) in the pathogenesis of AD.
Mild to moderate closed head traumatic brain injury (TBI) is a significant public health problem that has received increased public attention due, in part, to concern over the consequences of head injuries suffered by athletes as well as concern over the health effects of improvised explosive device (IED) exposure experienced by military personnel. TBI produces acute increases in two proteins strongly implicated in neurodegeneration: beta-amyloid and phosphorylated tau. We hypothesize that these acute changes can contribute to a neurodegenerative cascade that can result in a progressive neurodegenerative tauopathy referred to as chronic traumatic encephalopathy (CTE), and can also contribute to the development of Alzheimer’s disease. We have developed a mouse model of military blast exposure that elicits acute increases in tau phosphorylation and produces behavioral impairments that are evident at 2 weeks post-injury. We have also developed two novel transgenic mouse models that regulate the activity of the serine/threonine phosphatase, PP2A, and we find that targeting PP2A activity in this way alters tau phosphorylation and the sensitivity of these animals to behavioral and electrophysiological impairments caused by exogenous beta-amyloid exposure. In my presentation, I will describe our blast exposure methodology and our work in characterizing its biochemical and behavioral effects as well as our tests of the hypothesis that altered PP2A methylation may regulate not only beta-amyloid sensitivity but also sensitivity to TBI-related impairments.
S11-03 NEUROTROPHIN REGULATION OF NEURONAL DEATH AFTER INJURY Wilma Friedman 1, Marta Volosin 1, Juan Zanin 1 Barbara Hempstead 2 1 Rutgers University, Biological Sciences, Newark, USA 2 Weill Cornell Medical College, Department of Medicine, New York, USA The neurotrophin family of growth factors regulates neuronal growth, differentiation and survival during development by activating signaling via the Trk family of receptor tyrosine kinases. However, the neurotrophin precursors, or proneurotrophins, are potent ligands for the p75 neurotrophin receptor (p75NTR)-sortilin complex that can induce apoptosis, especially after injury. Our previous studies have demonstrated that p75NTR is upregulated after seizures __________________
2015 Transactions of the American Society for Neurochemistry ®
27
S11 Dissecting the Molecular Mechanisms of Acute Neurologic Injury and mediates neuronal death in response to elevated levels of proNGF. We have now also demonstrated that p75NTR mediates neuronal death after TBI, suggesting that this may be a general injuryrelated mechanism of neuronal death. We previously demonstrated that p75NTR-mediated apoptotic signaling requires activation of the intrinsic caspase pathway, involving caspases-9, -6 and -3, and simultaneous induction of PTEN to suppress Akt activation. ProNGF induction of PTEN occurred at the translational level, and we are currently investigating mechanisms by which this is regulated particularly the role of microRNAs. In addition to promoting neuronal apoptosis, we are also investigating a role for p75NTR in regulating axonal degeneration independent of neuronal death, and we are studying the mechanisms governing that process.
have identified critical caspases in multiple models of brain injury and degeneration. To target specific caspases we have developed novel cell permeant inhibitors specific for individual caspases. By employing specific caspase inhibitors we can define the function of an individual caspase family member and avoid potential side effects which can result from inhibiting multiple caspase family members. Intranasal delivery of a specific inhibitor of caspase-9 provides substantial functional neuroprotection in rodent models of cerebral ischemia. Surprisingly caspase-9 inhibition abrogates edema as well as neuronal death and neurologic dysfunction. We will present our recent studies on how caspase-9 activation leads to the major sequelae of stroke and also address the therapeutic potential of caspase inhibition.
S11-04 MECHANISMS OF AND THERAPIES FOR BRAIN INJURY AND NEURODEGENERATION Carol Troy Columbia Univ Medical Center, Pathology & Cell Biology, Neurology, Taub Institute, New York, USA Activation of specific caspases – a hierarchical family of cell death proteases – in cerebral ischemia causes neuronal death. We __________________
2015 Transactions of the American Society for Neurochemistry ®
28
S12 Signaling Pathways Regulating Remyelination S12-01
S12-02
SOX17 IN WHITE MATTER REPAIR: VENTURING BEYOND WNT Li-Jin Chew 1, Xiaotian Ming 1, Jeff Dupree 2, Vittorio Gallo 1 1 Children's Research Institute, Children's National Medical Center, Center for Neuroscience Research, Washington, USA 2 Virginia Commonwealth University, Dept of Anatomy and Neurobiology, Richmond, VA, USA
ERK2 MAP KINASE REGULATES TIMELY REMYELINATION IN THE ADULT BRAIN Sharyl Fyffe-Maricich, Kelly Michel, Tianna Zhao, Molly Karl Katherine Lewis University of Pittsburgh medical school, Pediatrics, Pittsburgh, USA
Successful remyelination relies on the timely completion of terminal differentiation by oligodendrocyte progenitor cells (OPC) and ensheathment of axons. Signaling events in the adult white matter (WM) that promote these processes are incompletely understood. We have previously identified Sox17 as a positive regulator of postnatal OPC maturation. The peak of Sox17 expression in preoligodendrocytes supports a role in the initiation of OPC differentiation, and its inhibition of Wnt/beta-catenin activity in vitro promotes myelin gene expression. Our studies in novel Sox17 gainand loss- of function mouse strains indicate roles in oligodendrocyte maturation and survival, as well as regeneration after demyelination. Sox17 transgenesis provides protection against lysolecithininduced demyelination through dual mechanisms of reduced cellular apoptosis and preservation of myelin ultrastructure, as well as increased oligodendrogenesis through enhanced OPC production and maturation. In Sox17 transgenic mice, we also observed significantly reduced activation of beta-catenin, and increased Gli2- and Sonic Hedgehog-expressing cells. To determine the contribution of these signaling pathways in Sox17-mediated oligodendrogenesis, acute stereotaxic injections of Hedgehog and beta-catenin antagonists were performed in the adult WM. These showed that Sox17 transgenesis: i) specifically re-directed Smoothened activity from OPCs to O4-pre-oligodendrocytes, enhancing lineage progression, and ii) abolished beta-catenin-mediated inhibition of OPC maturation. In the course of these studies, functional antagonism between GLI and active beta-catenin was demonstrated in adult WM, and beta-catenin activation by GLI inhibition was found to be associated with increased cell death. These suggest that the loss of Shh/ GLI signaling during demyelination contributes to beta-catenininduced cell death, and that Sox17 additionally prevents death by inhibiting injury-induced changes in GLI and beta-catenin activity. Our results thus indicate that both processes of enhanced cell survival and oligodendrogenesis improve repair in WM lesions through simultaneous regulation of canonical Wnt and Hedgehog signaling, and that modulation of crosstalk between these pathways underlies Sox17-dependent protection and lineage progression in remyelinating OPCs.
Multiple sclerosis (MS) is a disease characterized by chronic demyelination in the central nervous system (CNS). At early stages of the disease, patients with MS often exhibit periods of active demyelination that are followed by periods of myelin repair. As the disease progresses, however, myelin repair eventually fails and the reasons for this failure remain unclear. Successful remyelination requires the robust and timely production of myelin proteins in order to generate new myelin sheaths. The underlying regulatory mechanisms and complex molecular basis of myelin regeneration remain poorly understood. ERK MAP kinases are critical intracellular molecules that transduce extracellular signals at multiple stages of OL development, and we have previously shown that ERK2 plays a specific role in the timing of mouse forebrain myelination during postnatal development. Here, we have investigated the role of ERK2 MAP kinase in the timing of remyelination in the adult. Conditional deletion of Erk2 from cells of the oligodendrocyte lineage resulted in delayed remyelination following demyelinating injury to the adult mouse corpus callosum. The delayed repair occurred as a result of a specific deficit in the expression of the major myelin protein, MBP. MBP mRNA levels were unaffected by the loss of ERK2. In the absence of ERK2, activation of the ribosomal protein S6 kinase (p70S6K) and its downstream target, ribosomal protein S6 (S6RP), was impaired at a critical time when pre-myelinating oligodendrocytes were transitioning to mature cells capable of generating new myelin sheaths. These results suggest a potential role for ERK2 in the translational control of MBP, a myelin protein that appears critical to ensure the timely generation of new myelin sheaths following demyelinating injury.
S12-03 GLIA-DERIVED BDNF AFFECTS RECOVERY FROM A DEMYELINATING INJURY Cheryl Dreyfus Rutgers-Robet Wood Johnson Med Sch, Neuroscience & Cell Biology, Piscataway, NJ, USA Although it is established that BDNF enhances oligodendrocyte differentiation following a demyelinating lesion, the sources of BDNF that may be harnessed to reverse deficits associated with such lesions are poorly defined. Here we will discuss roles of astrocytes in serving such a role. First we will discuss previous culture studies indicating that cultured astrocytes respond to glutamate metabotropic stimulation by increasing BDNF synthesis and release. Second, we will focus on effects of the general metabotropic glutamate agonist, ACPD in vivo. Following cuprizoneelicited demyelination, astrocytes increase levels of metabotropic glutamate receptors and contain BDNF. When ACPD is injected into the demyelinating lesion, increases in BDNF and myelin __________________
2015 Transactions of the American Society for Neurochemistry ®
29
S12 Signaling Pathways Regulating Remyelination proteins are evident. These increases in myelin proteins are eliminated by co-injection of trkB-Fc to locally deplete BDNF actions. Moreover, increases in myelin proteins are eliminated by deletion of astrocyte-derived BDNF. The data suggest that astrocytes may serve as a source of BDNF, and possibly other growth factors, that can be used to reverse deficits following demyelination. (Supp: NIH NS036647, HD23315 and the NMSS)
S12-04 THERAPEUTIC INTERVENTION TO INDUCE OLIGODENDROCYTE SURVIVAL AND DIFFERENTIATION IN MOUSE MODELS OF DEMYELINATION Seema Tiwari-Woodruff, Jonathan Hasselamnn, Anna Khalaj John Katzenellenbogen School of Medicine at the University of California at Riverside, Biomedical Sciences, Riverside, USA Restoration of the previously damaged myelin sheath by replenishment of oligodendrocytes (OLs), should not only be useful in restoring saltatory axon conduction, but should represent a major boost to axon survival. Transition of early OL progenitors (OPCs) __________________
2015 Transactions of the American Society for Neurochemistry ®
to late OPCs requires Erk1/2 signaling, and transition of immature OLs to mature OL requires mTOR signaling. PI3K/Akt/mTOR pathway plays a major role in the late stages of OL differentiation and myelination. We hypothesized that therapeutic intervention to decrease demyelination and axon damage will involve the activation of these pathways to enhance endogenous OLP/OL population and axon remyelination. We have shown that therapeutic treatment with the modestly selective generic estrogen receptor (ER) ! ligand diarylpropionitrile (DPN) confers functional neuroprotection in chronic experimental autoimmune encephalomyelitis (EAE) by stimulating endogenous myelination. More potent, selective ER! agonists improved clinical disease and retard motor performance. Increased colossal myelination and mature oligodendrocyte numbers correlated with improved callosal conduction and refractoriness. Therapeutic treatment EAE with various ER! agonists resulted in a significant increase in BDNF and both phosphorylated (p) -AKT and p-motor levels compared to vehicle-treated EAE mice. Valuable insights about the appropriate timing and nature of the signaling that play central roles in the regulation of endogenous precursor cells and their capacity to remyelinate could serve as useful targets to enhance remyelination in neurological diseases.
30
S13 Mechanisms of Cell Death and Diseases S13-01 CONVERGENT ZINC AND CALCIUM SIGNALING CASCADES IN Kv2.1-ENABLED NEURONAL CELL DEATH Elias Aizenman University of Pittsburgh School of Medicine, Neurobiology, Pittsburgh, USA A simultaneous increase in cytosolic Zn2+ and Ca2+ accompanies the initiation of neuronal cell death signaling cascades. However, the molecular convergence points of cellular processes activated by these cations are poorly understood. We have reported that oxidantinduced intraneuronal Zn2+ liberation triggers a syntaxin-dependent incorporation of Kv2.1 channels into the plasma membrane. This can be detected as a marked enhancement of delayed rectifier K+ currents, a process responsible for the cytoplasmic loss of K+ that enables protease and nuclease activation during apoptosis. We demonstrate that an oxidative stimulus also promotes intracellular Ca2+ release and activation of CaMKII, which, in turn, modulates the ability of syntaxin to interact with Kv2.1. Pharmacological or molecular inhibition of CaMKII prevents the K+ current enhancement observed following oxidative injury and, importantly, significantly increases neuronal viability. We next tested whether disrupting the interaction of Kv2.1 and syntaxin promoted the survival of cortical neurons following injury. Syntaxin is known to bind to Kv2.1 in a domain comprised of amino acids 411-522 of the channel’s cytoplasmic C-terminus (C1a). We observed that this domain is required for the apoptotic K+ current enhancement. Moreover, expression of an isolated Kv2.1-derived C1a peptide is sufficient to suppress the injury-induced increase in currents by interfering with Kv2.1/syntaxin binding. By sub-dividing the C1a peptide, we were able to localize the syntaxin binding site on Kv2.1 to the most plasma membrane-distal residues of C1a. Importantly, expression of this peptide segment in neurons prevented the apoptotic K+ current enhancement and cell death following an oxidative insult, without largely impairing baseline K+ currents or normal electrical profiles of neurons. These results establish that binding of syntaxin to Kv2.1 is crucial for the manifestation of oxidant-induced Zn2+/Ca2+-mediated apoptosis, and thereby suggest a potential new direction for therapeutic intervention in the treatment of neurodegenerative disorders.
S13-02 THE SOMATODENDRITIC K+ CHANNEL KV2.1 IS A CRITICAL REGULATOR OF NEURONAL SURVIVAL AND DEATH Durga Mohapatra 1, 2, Andrew Shepherd 1, 2, Lipin Loo 1 1 The University of Iowa, Department of Pharmacology, Carver College of Medicine, Iowa City, USA 2 Washington University, Department of Anesthesiology, School of Medicine, St. Louis, USA The intrinsic excitability of neurons is homeostatically regulated by voltage-depenent K+ (Kv) conductances across the plasma mem__________________ 2015 Transactions of the American Society for Neurochemistry ®
brane, which under pathological conditions is often threatened, leading to collapse of cellular integrity and death. Our recent findings show distinct signaling pathways originating from multiple neuropathological assaults on mammalian brain converge on the major neuronal somato-dendritic Kv channel Kv2.1, and influence the intrinsic cellular mechanism of survival and death in neurons. Our results show that dynamic Ca2+/calcineurin-dependent dephosphorylation of Kv2.1 in response to the acute phase of assaults, such as ischemic stroke and exposure to human deficiency virus type-1 (HIV-1) glycoprotein gp120, leads to enhanced channel activation at less depolarizing potentials. This, accompanied with the redistribution of Kv2.1 channel protein across the plasma membrane in somatodendritic compartments, serves as an intrinsic mechanism for neuroprotection. However, during the chronic phases of ischemic stroke/reperfusion and HIV-1 gp120 exposure, distinct Src- and p38 mitogen-activated protein kinase (MAPK)-dependent phosphorylation of Kv2.1 lead to increases in the surface trafficking of the channel protein and sustained efflux of K+ from neuronal cytoplasm. This results in the collapse of neuronal plasma membrane and mitochondrial membrane integrity, ultimately leading to the initiation of apoptotic cell death in neurons. In line with these findings, pharmacological blockade of Kv2.1 led to the attenuation of such apoptotic cell death in neurons. Our findings suggest that distinct phospho-modifications in the Kv2.1 channel could constitute a therapeutic target for providing neuroprotection to apoptotic death associated with multiple neurodegenerative pathologies.
S13-03 MITOCHONDRIAL BASE EXCISION REPAIR ATTENUATES REPERFUSION INJURY IN ISCHEMIC STROKE Roger Simon Morehouse School Medicine, Neurobiology, Atlanta, United States of America Endovascular and thrombolytic stroke treatment attenuate infarction but also result in reperfusion injury from induced generation of reactive oxygen species (ROS). ROS produce injury to DNA bases. Mitochondrial DNA (mtDNA) is far more sensitive to such oxidative damage than is the nuclear genome. The propensity for cytotoxicity is inversely related to the efficiency of mtDNA repair. We have shown this relationship in ischemic brain. Moreover, DNA glycosylases, the first and rate limiting enzyme in base excision repair, is neuroprotective in the setting of oxidative DNA damage in vitro and in modeled stroke in vivo. Accordingly we devised a novel fusion protein construct, with TAT, targeting DNA repair glycosylases to mitochondria and show in rodent models of ischemia/reperfusion that such treatment attenuates reperfusion injury, does so in a dose response manner, and has a three hour time window of effect. Thus pharmacologic enhancement of mtDNA repair attenuates the degree of brain infarction following reperfusion and does so in a time window that is highly translational.
31
S13 Mechanisms of Cell Death and Diseases
S13-04 'THE SUR1-TRPM4 CHANNEL IN CNS INJURYŸ J. Marc Simard University of Maryland School of Medicine, Neurosurgery, Baltimore, USA "Accidental necrosis" due to profound adenosine triphosphate (ATP) depletion or oxidative stress is distinguished from regulated necrosis by the absence of death receptor signaling. However, both accidental and regulated necrosis have in common the process of " oncosis," a physiological process characterized by Na(+) influx and cell volume increase that, in necrotic cell death, is required to produce the characteristic features of membrane blebbing and membrane rupture. Emerging data indicate that the monovalent cation channel, Sur1-Trpm4, plays a crucial role as end executioner in the accidental necrotic death of ATP-depleted or redox-challenged CNS cells, both in vitro and in vivo. We will review emerging evidence that Sur1-Trpm4 is involved in the cell death process of oncosis in acute diseases of the CNS including stroke, subarachnoid and intraventricular hemorrhage, hemorrhagic forms of encephalopathy of prematurity, and traumatic brain and spinal cord injury.
S13-05 WNK3-SPAK/OSR1-NKCC1 SIGNALING PATHWAY IN ISCHEMIC BRAIN INJURY Dandan Sun University of Pittsburgh, Neurology, Pittsburgh, USA Ischemia-mediated activation of Na-K-Cl cotransporter (NKCC1) leads to Na+ overload, cytotoxic edema, and cell death. However, the molecular mechanisms underlying these processes are not understood. The WNK (with no lysine = K) and Ste20/SPS1-related proline-alanine-rich protein kinase (SPAK)/oxidative stressresponsive 1 (OSR1) kinase are evolutionarily conserved serinethreonine kinases which regulate activities of multiple ion transporters and channels. Our recent study demonstrates that genetic deletion in mice of WNK3, a WNK family member highly expressed in brain, is neuroprotective after transient focal ischemia. We show that genetic deletion of WNK3 or siRNA-mediated knockdown of SPAK or OSR1, protects against ischemic neuroglial death triggered __________________
2015 Transactions of the American Society for Neurochemistry ®
by oxygen-glucose deprivation/reoxygenation. Using phospho specific antibodies, we detected robust up-regulation of p-NKCC1 as well as p-SPAK/p-OSR1 expression in peri-infarct cortex, striatum and corpus callosum in wild type (WT) mice after focal ischemia. In WNK3 knockout (KO) mice, ischemia-induced hyper-phosphorylations of both the SPAK/OSR1 catalytic T-loop and NKCC1 are abolished, infarct volume and axonal demyelination are reduced, and neurobehavioral recovery is accelerated. Magnetic resonance imaging detected less brain edema and white matter injury not only in WNK3 KO but also in SPAK heterozygous (Het) and SPAK KO mice after ischemia. These data provide fresh insight into the roles of ion transporters and their regulatory kinases in ischemic neuroglial injury, and identify the WNK3-SPAK/OSR1 kinase complex as a compelling target for novel neuroprotective strategies following stroke. Support: NIH R01NS38118
S13-06 DYSFUNCTION OF MEMBRANE TRANSPORTER/EXCHANGER AND HYBRID CELL DEATH IN NEURONAL AND CANCER CELLS Shan Ping Yu Emory University, Anesthesiology, Atlanta, USA Accumulating evidence reveals that metabolism disruption due to mitochondrial damage is a critical pathological event after stroke and neurodegenerative diseases. The energy failure of ATP deficiency directly leads to the dysfunction of Na+/K+-ATPase and disruption of ionic homeostasis. Moreover, the Na+/K+-ATPase and exchangers in the plasma membrane have recently become the novel targets in cancer therapy. Our investigations identified regulatory mechanisms of Na+/K+-ATPase such as Src tyrosine phosphorylation and inhibition by reactive oxygen species (ROS). Resent data show that blocking Na+/K+-ATPase can lead to a hybrid form of cell death containing concurrent apoptotic and necrotic features. We further demonstrated that the hybrid cell death is a common form of cell death in responding to multiple insults involving the Na+ pump or the Na+/Ca2+ exchanger. A better understanding of the hybrid cell death will provide new strategies in the treatment of stroke as well as brain cancers such as gioblastoma.
32
S14 Retromer and Neurodegenerative Diseases S14-01 CHEMICAL CHAPERONES AS AN APPROACH TO TREATMENT OF ALZHEIMER’S DISEASE Dagmar Ringe Brandeis University, Biochemistry, Waltham, USA Chemical (pharmacological) chaperones have been used for therapeutic purposes for some time, the first example being tetrahydrobiopterin (BH4) for treatment of some forms of phenyl ketone urea. Here we are using the same strategy to increase levels and function of a complex implicated in both sporadic and familial Alzheimer’s disease (AD), and more recently in familial Parkinson’s disease (PD). Retromer is a multi-protein complex that traffics endosomal cargo back to the Golgi. It has been shown to traffic the Amyloid Precursor Protein (APP) away from the endosome, where the beta-secretase is optimally active, thereby regulating A! peptide accumulation. We have identified pharmacological chaperones that enhance retromer stability and function, with the goal of redirecting APP away from those compartments in which it is proteolyzed. First, we used the crystal structures of retromer proteins to identify the ‘weak link’ of the complex, and to complete an in silico screen of small molecules predicted to enhance retromer stability. Among the hits, an in vitro assay identified one molecule that stabilized retromer against thermal denaturation by more than 10 deg. C. Second, the levels of retromer proteins in hippocampal neurons are increased in the presence of the small molecule, shifting APP away from the endosome, and decreasing A! accumulation dramatically, in a dose-dependent manner. Together, these findings clarify mechanisms of retromer stability, and identify a pharmacological chaperone that has therapeutic potential for AD and possibly other neurodegenerative disorders.
S14-02 VPS10-FAMILY RECEPTORS IN NEURODEGENERATION AND INSULIN RESISTANCE Sam Gandy 1, Elysse Knight 1, Alan Attie 2, Christoph Buettner 1 Michelle Ehrlich 1 1 Icahn School of Medicine, Neurology, Pediatrics, Endocrinology, New York, USA 2 University of Wisconsin, Biochemistry, Madison, USA Members of the vacuolar protein sorting 10 (Vps10) family of receptors (including sortilin, SorL1, SorCS1, SorCS2, and SorCS3) play pleiotropic functions in protein trafficking and intracellular and intercellular signaling in neuronal and non-neuronal cells. Interactions have been documented between Vps10 family members and the retromer coat complex, a key component of the intracellular trafficking apparatus that sorts cargo from the early endosome to the trans-Golgi network. In recent years, genes encoding several members of the Vps10 family of proteins, as well as components of the retromer coat complex, have been implicated as genetic risk factors for sporadic and autosomal dominant forms of neurodegenerative diseases as well as for type 2 diabetes and atherosclerosis. Endosomal sorting of the Alzheimer amyloid precursor protein (APP) plays a key role in the biogenesis of the amyloid-! __________________ 2015 Transactions of the American Society for Neurochemistry ®
(A!) peptide. Genetic lesions underlying Alzheimer's disease (AD) can act by interfering with this physiological process. Specifically, proteins involved in trafficking between endosomal compartments and the trans-Golgi network (TGN) [including the retromer complex (Vps35, Vps26) and its putative receptors (sortilin, SorL1, SorCS1)] have been implicated in the molecular pathology of lateonset AD. Previously, we demonstrated a role for SorCS1 in APP metabolism and A! production, but the underlying mechanism was, at that time, poorly understood. We will present evidence for the existence of a motif within the SorCS1c cytoplasmic tail that, when manipulated, results in perturbed sorting of APP and/or its fragments to endosomal compartments, decreased retrograde TGN trafficking, and increased A! production in H4 neuroglioma cells. These perturbations apparently do not involve turnover of the cell surface APP pool, but rather they involve intracellular APP and/or its fragments, downstream of APP endocytosis. We will also highlight the emerging data implicating the Vps10 family of receptors and the retromer in physiological intracellular trafficking and signaling and in the pathogenesis of neurodegeneration.
S14-03 VPS35-DEFICIENCY IS A RISK FACTOR FOR THE PATHOGENESIS OF BOTH ALZHEIMER’S DISEASE AND PARKINSON’S DISEASE Wen-Cheng Xiong Georgia Regents University, Neuroscience & Regenerative Medicine & Neurology, Augusta, USA VPS35 (vacuolar protein sorting 35) is a major component of the retromer complex that is essential for the selective endosome-toGolgi retrieval of membrane proteins. Human clinical investigations have suggested that dysfunction of retromer is a risk factor for pathogenesis of both Alzheimer’s disease (AD) and Parkinson’s disease (PD). We have taken advantage of Vps35-deficient animal model to address this issue and to investigate the underlying mechanisms. VPS35 is highly expressed in mouse hippocampus, an ADvulnerable brain region. Vps35 hemizygotes (Vps35+/-) show increased soluble amyloid beta (Abeta) level and reduced glutamate neurotransmission in hippocampus. However, the AD-relevant neurophysiological deficits, including reduced LTP (long term potentiation) in hippocampal neurons, defective spatial learning and memory by Morris water maze assay, and Ab plaques, were not observed. When Vps35+/- mice were crossed with Tg2576 mouse model of AD, the AD-relevant deficits were accelerated and enhanced, suggesting that Vps35-deficicency promotes AD neuropathology. Whereas multiple mechanisms may underlie this event, VPS35-regulation of BACE1-mediated APP cleavage and Abeta production appears to be a crucial one. Interestingly, mutations in the Vps35 gene are identified in the autosomal dominant PD patients, raising questions of whether Vps35 mutation acts as Vps35-loss of function mutant and if Vps35deficiency results in PD-like pathologic deficit. VPS35 was also expressed in mouse dopamine (DA) neurons in substantia nigra pars compacta (SNpc) and striatum, regions that are PD-vulnerable. Vps35+/- mice exhibited PD-like deficits including loss of DA in __________________ 33
S14 Retromer and Neurodegenerative Diseases these areas and accumulation of a-synuclein in SNpc-DA neurons. These results thus support the view for Vps35-deficiency as a risk factor for pathogenesis of both AD and PD. However, the underlying molecular and cellular mechanisms may be different, which is an important question for future investigations.
S14-04 PARKINSON DISEASE-ASSOCIATED MUTANT VPS35 CAUSES ABNORMAL MITOCHONDRIAL DYNAMICS AND MITOCHONDRIAL DYSFUNCTION Xiongwei Zhu Case Western Reserve University, Pathology, Cleveland, USA Mitochondria play a crucial role in neuronal function and, not surprisingly, mitochondrial dysfunction is a prominent feature in the brain of various neurondegenerative diseases including Parkinson disease (PD). Recent studies suggest that mitochondria are highly dynamic organelles characterized by a delicate balance of fission and fusion. This concept has revolutionizes our basic understanding of the regulation of mitochondrial structure, function and distribution which has far-reaching significance in studies of health and disease. Increasing evidence suggest abnormal mitochondrial dynamics and quality control as an important mechanism underlying mitochondrial dysfunction and neuronal dysfunction in PD. Vps35 is the third dominant gene associated with PD after alpha__________________
2015 Transactions of the American Society for Neurochemistry ®
synuclein and LRRK2. However, despite the knowledge of the role of VPS35 as a key component of the retromer complex important for the retrieval of membrane proteins, how VPS35 mutant leads to neurodegeneration in PD remains elusive. Given the critical role of mitochondrial dysfunction in the pathogenesis of PD, we explored the potential role of VPS35 in the regulation of mitochondrial function. We found that PD-associated VPS35 D620N mutation caused extensive mitochondrial dysfunction along with significant neuronal death in M17 neuroblastoma cell line and in primary cortical neurons. Interestingly, we noticed significant mitochondrial fragmentation in cells expressing VPS35 D620N mutant and the inhibition of mitochondrial fission could almost completely block VPS35 D620N-induced mitochondrial deficits and neuronal deficits. Importantly, VPS35 D620N mutant also caused mitochondrial fragmentation and cell death in TH-positive neurons in mouse substantia nigra which could be alleviated by the inhibition of excessive mitochondrial fission. We further demonstrated that mitochondrial became fragmented and dysfunctional in fibroblasts from human PD patient with VPS35 D620N mutation. More detailed studies demonstrated a direct regulatory role of VPS35 on the mitochondrial fission/fusion machinery which is disturbed by the PD-assocaited mutation. Our studies revealed a novel cellular mechanism of the involvement of VPS35 in the regulation of mitochondrial dynamics, dysregulation of which is likely involved in the pathogenesis in VPS35 mutation-associated familial PD and possibly also in the sporadic form of this disease.
34
S15 Endoplasmic Reticulum Stress in Myelin Disorders S15-01 IMPAIRED EIF2B ACTIVITY IN OLIGODENDROCYTES CONTRIBUTES TO VANISHING WHITE MATTER DISEASE PATHOGENESIS Wensheng Lin University of Minnesota, Neuroscience, Minneapolis, USA Vanishing White Matter Disease (VWMD) is an inherited autosomal-recessive leukodystrophy caused by mutations in eukaryotic translation initiation factor 2B (eIF2B). eIF2B mutations predominantly affect the brain white matter, and the characteristic features of VWMD pathology include myelin loss and foamy oligodendrocytes. Activation of pancreatic endoplasmic reticulum kinase (PERK) has been observed in oligodendrocytes in VWMD. PERK activation in response to endoplasmic reticulum stress attenuates eIF2B activity by phosphorylating eIF2!, suggesting that impaired eIF2B activity in oligodendrocytes induced by VWMD mutations or PERK activation exploit similar mechanisms to promote selective white matter pathology in VWMD. Using transgenic mice that allow for temporally-controlled activation of PERK specifically in oligodendrocytes, we discovered that strong PERK activation in oligodendrocytes during development suppressed eIF2B activity and reproduced the characteristic features of VWMD in mice, including hypomyelinating phenotype, foamy oligodendrocytes, and myelin loss. Notably, impaired eIF2B activity induced by PERK activation in oligodendrocytes of fully-myelinated adult mice had minimal effects on morphology or function. Our observations point to a cell-autonomous role of impaired eIF2B activity in myelinating oligodendrocytes in the pathogenesis of VWMD.
S15-02 OLIGODENDROCYTE PROTECTION BY THE INTEGRATED STRESS RESPONSE: A POTENTIAL PHARMACOLOGICAL TARGET Brian Popko The University of Chicago, Center for Peripheral Neuropathy, Chicago, USA The integrated stress response (ISR), which is a conserved feature of all eukaryotic cells, is activated by, and provides protection to, a variety of cytotoxic insults. ISR signaling leads to the phosphorylation of the protein translation initiating factor eIF2-alpha, which reduces overall cellular translation rates and places the cell in a cytoprotective state. In our studies we are exploring the possibility that the induction of an enhanced ISR response, using genetic and pharmacological approaches, will provide increased protection to the CNS in response to neurological disorders. We have focused our efforts on oligodendrocytes, which are the myelinating cells of the __________________
2015 Transactions of the American Society for Neurochemistry ®
CNS. Oligodendrocyte apoptosis, in response to CNS inflammation, contributes to the development of the demyelinating disorder multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), such that approaches designed to protect these cells will likely have therapeutic value. Moreover, there is evidence that the ISR is activated in oligodendrocyts within demyelinating lesions in MS and EAE. Our work has demonstrated that ISR activation in oligodendrocytes protects the cells against inflammatory cytokines and significantly attenuates EAE disease severity, which is associated with reduced oligodendrocyte apoptosis, demyelination, and axonal degeneration. These results provide support for the premise that pharmacological agents that result in an enhanced ISR response will have therapeutic value in inflammatory demyelinating disorders.
S15-03 ENDOPLASMIC RETICULUM STRESS IN CHARCOT MARIE TOOTH NEUROPATHY Lawrence Wrabetz 1, Maria Paola Sidoli 1, Nicolo Müsner 1 Maurizio D'Antonio 2, Maria Laura Feltri 1 1 SUNY at Buffalo School of Medicine, Hunter James Kelly Research Institute, Buffalo, USA 2 San Raffaele Scientific Institute, DIBIT, Milan, Italy Endoplasmic Reticulum (ER) Stress has been implicated in the pathogenesis of both acquired and hereditary neuropathies. For example, mutant proteins associated with several Charcot-MarieTooth (CMT) neuropathies provoke ER stress and an unfolded protein response (UPR), and perturbations of UPR alter the severity of neuropathy. P0 glycoprotein is abundantly synthesized in myelinating Schwann cells. The mutant P0S63del causes CMT1B neuropathy in humans, and a very similar demyelinating neuropathy in S63del transgenic mice. P0S63del is retained in the ER of Schwann cells where it promotes unfolded protein stress and elicits an UPR associated with translational attenuation. We have shown that ablation of CHOP, a UPR mediator downstream of the PERK kinase sensor of unfolded proteins, ameliorates demyelination in S63del nerves. In addition, Gadd34 is a detrimental effector of CHOP that reactivates translation too aggressively in myelinating Schwann cells. Limitation of Gadd34 function improves myelination in S63del nerves, and reduces accumulation of P0S63del in the ER by maintaining translational attenuation. Surprisingly, ablation of PERK in Schwann cells, which activates translation and therefore should worsen demyelination, actually improves myelination and neuropathy. PERK may have additional targets outside of the UPR. Limiting Gadd34 in order to reset translational homeostasis may provide a therapeutic strategy in tissues impaired by misfolded proteins.
35
S15 Endoplasmic Reticulum Stress in Myelin Disorders
S15-04 ER STRESS AND SPINAL CORD INJURY Scott Whittemore, Sujata Saraswat Ohri, S Ashley Mullins Michal Hetman University of Louisville, KY Spinal Cord Injury Res Ctr, Louisville, USA Spinal cord injury (SCI) initiates a cascade of secondary cell death mechanisms in most resident cells. These pathophysiologic responses have been shown in non-CNS cells to initiate the endoplasmic reticulum stress response (ERSR). We addressed the extent to which and in what cell types, the ERSR was initiated after contusive thoracic SCI in adult mice. All three signaling branches (PERK, ATF6, and IRE1) are rapidly up regulated as early as 6 hours post-SCI and remained elevated until 72 hours post-SCI as detected by both PCR and immunohistochemistry. CHOP expression was observed in neurons and oligodendrocytes, but not in astrocytes at 72 hours post-SCI. In CHOP null mice, the ERSR was attenuated and hindlimb locomotor function significantly improved. At both acute (72 hours) and chronic (6 weeks) times after SCI, oligodendrocytes were significantly spared. Thus, blocking the proapoptotic aspects of the ERSR in oligodendrocytes was one mecha__________________
2015 Transactions of the American Society for Neurochemistry ®
nism underlying behavioral recovery. To explore if pharmacological enhancement of the homeostatic arm of PERK signaling was similarly therapeutically protective after SCI, mice were treated with salubrinal, which inhibits the dephosphorylation of eIF2! by both PPP1R15A/GADD34 and PPP1R15B/CReP, the stressinducible and constitutive eIF2! phosphatases, respectively. Salubrinal treatment resulted in similar ERSR attenuation, oligodendrocyte sparing, and locomotor recovery. Guanabenz, an FDA approved, specific inhibitor of PPP1R15A/GADD34 promoted the survival of OPCs treated with tunicamycin in vitro and enhanced eIF2! phosphorylation after SCI. However, SCI in wild type mice after treatment with guanabenz or in GADD34 null mice showed differential attenuation of the ERSR and no locomotor improvement. Since, GADD34 is an essential component of a negativefeedback loop operating under stress, GADD34-mediated translational de-repression is important not only for translation of the downstream effectors but also for the expression of essential intermediates in the activation of stress-induced gene expression programs. While ERSR remains an attractive therapeutic target after CNS injury, the exact protein(s) to optimally target remains elusive. Supported by NS073584 and GM103507.
36
S16 Wiring Axons by mRNA Regulation in Neuronal Health and Disease S16-01
S16-02
OPPOSING ROLES OF HUD AND KSRP IN SHARED TARGET MRNA STABILITY AND AXONAL GROWTH Nora Perrone-Bizzozero 1, Amy Gardiner 1, Cynthia Gomes 2 Jeffery Twiss 2 1 University of New Mexico, Neurosciences, Albuquerque, USA 2 University of South Carolina, Biology, Columbia, USA
FUTSCH/MAP1B IS A TRANSLATIONAL TARGET OF TDP-43 AND IS NEUROPROTECTIVE IN A DROSOPHILA MODEL OF ALS Daniela Zarnescu 1, Alyssa Coyne 1 Bhavani Bagevalu Siddegowda 1, Patricia Estes 1 Jeffrey Johannesmeyer 1, Tina Kovalik 2, Scott Daniel 1 Antony Pearson 1, Robert Bowser 2 1 University of Arizona, Molecular and Cellular Biology, Tucson, USA 2 Barrow Neurological Institute, Neurobiology, Phoenix, USA
Post-transcriptional mechanisms are known to play important roles in neuronal development as they allow for faster responses to environmental cues and provide spatially restricted compartments for local control of protein expression. Among these mechanisms, the control of mRNA stability by the interaction of RNA-binding proteins (RBPs) with AU-rich instability elements (ARE) in the 3’ UTR is one of the least understood. We have recently shown that two ARE-BPs, HuD and KSRP, compete for binding to the ARE in GAP-43 mRNA (Bird et al., 2013). Our previous work demonstrated that HuD binding stabilizes and localizes GAP-43 mRNA, promoting axonal outgrowth (Anderson et al., 2000, 2001, Bolognani et al., 2006, 2007) while KSRP binding destabilizes GAP-43 mRNA and stunts outgrowth (Bird et al., 2013). These results suggest that these proteins compete for the control of target expression and process outgrowth during neuronal development. Supporting this idea, we recently found that KSRP and HuD show different temporal patterns of expression both in culture and in vivo, with HuD preceding KSRP expression. To initially identify the repertoire of neuronal transcripts regulated by these ARE-BPs, we used UV-crosslinking and RNA immunoprecipitation followed by Affymetrix arrays (CLIP-chip). Briefly, E18 mouse neocortical tissue was processed for CLIP using antibodies to HuD, KSRP or control IgG. Microarray results were validated using qRT-PCR and KSRP KO mice. We found that KSRP and HuD bound both distinct and shared sets of target mRNAs. Besides GAP-43, other common targets included Actb, Cttnb1, Cdc42, Eif4g2, Fscn1, Hspa8, Marcks, Stmn1, Ttr and Tubb2b. Interestingly, many of these targets are localized to axons and upregulated in KSRP KO mice. These results suggest that the opposing roles HuD and KSRP in axonal outgrowth are due in part to the differential regulation of shared targets, with HuD stabilizing and KSRP destabilizing localized transcripts. Supported by NIH (R01 NS30255 to NPB and NS041596 to JLT).
TDP-43 is an RNA binding protein linked to ALS that is known to regulate the splicing, transport and storage of specific mRNAs into stress granules. Although TDP-43 has been shown to interact with translation factors, its role in axonal protein synthesis remains unclear. Here we provide evidence that TDP-43 associates with futsch mRNA in a complex and regulates its expression at the larval neuromuscular junction (NMJ) synapse in Drosophila. In the context of TDP-43 induced proteinopathy there is a significant reduction of futsch mRNA at the NMJ compared to motor neuron cell bodies where we find higher levels of transcript compared to controls. TDP-43 also leads to a significant reduction in Futsch protein expression at the NMJ and polysome fractionations coupled with qRT-PCR indicate that TDP-43 leads to a futsch mRNA shift from actively translating polysomes to non-translating RNA granules. These data suggest that in addition to its effect on localization, TDP-43 also regulates the translation of futsch mRNA in axons. We also show that futsch overexpression is neuroprotective by extending lifespan, reducing TDP-43 aggregation and suppressing ALS-like locomotor dysfunction as well as NMJ abnormalities by increasing microtubule and synaptic stabilization. Furthermore, the localization of MAP1B, the mammalian homolog of Futsch is altered in ALS spinal cords in a manner similar to our observations in Drosophila motor neurons. These results suggest that microtubule stability defects in motor neuron axons caused by TDP-43 dependent alterations in futsch mRNA localization and translation in vivo are in part responsible for the disease mechanism.
S16-03 RNA PROTEIN INTERACTIONS FOR AXONALLY TRANSPORTED MRNAS Jeffery Twiss, Seung Joon Lee, Cynthia Gomes Sharmina Miller-Randolph University of South Carolina, Biological Sciences, Columbia, USA Proteins synthesized locally in mature PNS axons are needed for responses to injury and regenerative growth. Analyses of axons isolated from PNS and CNS neurons has shown a surprisingly complex population of several hundred mRNAs. These mRNAs are actively transported into neuronal processes and this requires that the transcripts interact with RNA binding proteins (RBP). Work from our lab indicates that some axonal mRNAs compete for binding to limited quantities of RBP, and this determines how much __________________ 2015 Transactions of the American Society for Neurochemistry ®
37
S16 Wiring Axons by mRNA Regulation in Neuronal Health and Disease of an mRNA localizes. For example, !-actin and GAP-43 mRNAs compete for binding to ZBP1/Imp1, and transcriptional induction of GAP-43 mRNA after axotomy decreases axonal transport of !-actin mRNA. Other mRNAs show increased localization with neuronal injury, suggesting that either levels or activities of their needed RBPs are regulated by axotomy. NMP35 and Nrn1 are examples of mRNAs that move from the cell body into axons after axotomy. Still other axonal mRNAs are constitutively transported into axons where their translation appears to be regulated by different stimuli. Axonal levels of amphoterin/HMGB1 mRNA do not change with injury, but the transcript appears to be translated at higher efficiency in axons of injury-conditioned neurons. Despite advances in understanding how transport can be regulated, we know of relatively few RBPs that are needed for axonal localization. Using transfection of heterologous reporter mRNAs, we have determined ‘motifs’ in untranslated regions (UTRs) for several axonal mRNAs that are necessary and sufficient for their subcellular localization in cultured neurons. These motifs have provided a platform for isolation of RBPs from axons, and we have identified high affinity and low affinity interacting RBPs for these localization motifs. These RBPs were not previously known to localize into axons, so they provide novel insight into RNA-protein interactions that are needed for axonal mRNA transport [Supported by grants from National Institutes of Health (R01-NS041596), Department of Defense Orthopaedic Research Program (W81XWH-13-1-0308), and the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation].
S16-04 IMPAIRED MRNP COMPLEX ASSEMBLY AND AXONAL MRNA LOCALIZATION IN A MOUSE MODEL OF SPINAL MUSCULAR ATROPHY Gary Bassell, Paul Donlin, Claudia Fallini, Jeremy Rounet Shirley Huang, Kathryn Williams, Wilfried Rossoll Emory University, Cell Biology, Atlanta, USA Spinal muscular atrophy (SMA) is a neuromuscular disease characterized by a specific degeneration of motor neurons. SMA results from a reduction in the survival of motor neuron (SMN) protein, which has a well characterized role in assembly of small nuclear ribonucleoproteins (snRNPs). Studies from our laboratory and others have revealed impairments in axonal mRNA localization in SMA models. We have discovered specific defects in the axonal localization of mRNAs (!-actin, Gap43) and mRNA-binding proteins (HuD, IMP1) in axons and growth cones of primary motor neurons from SMA mice or depleted of SMN. These findings led to the hypothesis that SMN plays a critical role in the assembly and/or trafficking of messenger ribonucleoproteins (mRNPs) in neuronal processes. However, the molecular function(s) of SMN in the biology of mRNPs remained unclear due to the lack of specific assays. To investigate the biological role of SMN and the effects of SMN deficiency in axonal mRNA regulation, we have established a trimolecular fluorescence complementation (TriFC) assay in motor neuron cultures as a sensor for mRNA and protein association, which permitted an analysis of the proposed role of SMN in mRNP assembly and localization. Our findings revealed a deficiency in the __________________
2015 Transactions of the American Society for Neurochemistry ®
assembly of IMP1 protein/!-actin mRNA containing complexes in SMA motor neurons, which is consistent with SMN acting as a chaperone for mRNP complex assembly. We further show that SMN-deficiency results in defects in axonal local translation, likely a downstream consequence of impaired mRNP transport complex assembly. Local translation of !-actin and GAP43 are known to play an important role in axon outgrowth and branching. Our findings reveal a novel function for SMN in the assembly of axonally transported mRNP complexes, and uncover consequent effects on axonal mRNA localization and translation that may contribute to SMA pathology.
S16-05 COORDINATING SYNTHESIS OF AXONAL CYTOSKELETAL PROTEINS WITH CELL SIGNALING PATHWAYS THROUGH RNA REGULATION BY HNRNP K Ben Szaro State University of New York at Albany, Department of Biological Sciences, Albany, USA During axonal outgrowth the synthesis of the structural proteins that organize the axonal cytoskeletal polymers must be coordinated both with one another to prevent the formation of aggregates and with the dynamics of axonal elongation to match supply with demand. This coordination is accomplished, in large part, posttranscriptionally through the actions of shared RNA binding proteins regulated by cell signaling pathways to control the trafficking and translation of the mRNAs. Our studies in the developing embryonic nervous system and regenerating optic nerve of the frog Xenopus laevis have established that the RNA binding protein hnRNP K plays a central role in orchestrating the trafficking and translation of multiple cytoskeletal-related RNAs during axonal outgrowth. In the brain, hnRNP K associates endogenously with mRNAs related to all three cytoskeletal polymers, including Type IV neurofilament proteins as well as several microtubule and microfilament associated proteins, such as tau, ARP2 and GAP-43. Suppressing hnRNP K protein expression in the embryo leads to disorganized arrays of neuronal cytoskeletal polymers and the loss of the axon itself, despite neurons maintaining their cell polarity. Upon hnRNP K knockdown, although hnRNP K-regulated mRNAs are transcribed and spliced, they are exported less efficiently from the nucleus and are not translated into protein. The same defects are seen with hnRNP K knockdown in retinal ganglion cells during optic axon regeneration, indicating that this RNA binding protein plays a similar role in axon regeneration. During axonogenesis, hnRNP K’s actions are regulated by JNK, a kinase whose cytoplasmic activity is required for axon outgrowth. JNK-phosphomimetic, but not -phosphodeficient, hnRNP K rescues developing axons both from hnRNP K knockdown and from pharmacological JNK inhibition, and at the molecular level, can initiate translation of its regulated, cytoskeletal-related transcripts. These studies indicate that hnRNP K is a nexus for coordinating the nuclear export and translation of multiple cytoskeletal-related RNAs with cell signaling pathways involved in wiring the brain. Supported by the NSF.
38
S17 Stress Organelles and Neurodegenerative Diseases S17-01 INHIBITING HISTONE DEACETYLASE 6 AS A THERAPEUTIC STRATEGY FOR TAUOPATHIES Leonard Petrucelli, Casey Cook Mayo Clinic, Neuroscience, Jacksonville, USA Given the accumulation of hyperphosphorylated tau in neurofibrillary tangles (NFTs) is a neuropathological hallmark of tauopathies, including Alzheimer’s disease (AD) and Chronic traumatic encephalopathy, therapeutic strategies targeting tau may hold great promise. Toward that end, we have recently discovered that the enzyme histone deacetylase 6 (HDAC6) directly influences and modulates phosphorylation and acetylation on the tau protein. Offering novel insight into disease mechanisms, our data also suggest these modifications on tau compete to maintain the balance between health and disease. Specifically, we show HDAC6 deacetylates tau on KXGS motifs, thus influencing susceptibility to phosphorylation on this same motif. We observe robust inhibition of tau aggregation following acetylation, an effect that is dependent upon acetylation of KXGS motifs. Using a novel, site-specific antibody to detect acetylation of KXGS motifs, we demonstrate that these sites are hypoacetylated in patients with AD as well as a mouse model of tauopathy. As the loss of acetylation in these critical regions of tau may be indicative of HDAC6 hyperactivity in patients with AD, we predicted that lowering HDAC6 activity therapeutically would increase acetylation and effectively prevent the accumulation of toxic tau. We found chronic treatment with a selective and blood-brain barrier permeable HDAC6 inhibitor significantly decreases levels of hyperphosphorylated tau in mice. We have also recently conducted proteomic studies to identify additional acetylation sites on tau that are regulated by HDAC6, in order to further characterize how acetylation and HDAC6 modulate tau’s propensity to aggregate. Taken together, our data demonstrate HDAC6 is an exploitable and drug-able target to block the formation of pathogenic tau species in disease.
S17-02 ROLE OF C-ABL AND P53 IN AUTOPHAGIC DEFECTS ASSOCIATED WITH ALPHA-SYNUCLEINOPATHY IN VIVO Michael Lee, Elly Liao, Razaul Larim, Justin Barnes University of Minnesota, Neuroscience, Minneapolis, USA Neurodegeneration in PD is thought to be the consequence of alpha-synucleinopathy. Our studies of a transgenic (Tg) mouse model of alpha-synucleinoapthy show that a stress-activated kinase, c-Abl, is activated with the disease in the HuaS(A53T) Tg mice. Previous studies showed that c-Abl is activated in PD brains and active c-Abl could phosphorylate and inactivate parkin. Our results show that aS pathology is sufficient to cause c-Abl activation. We also found that alpha-synucleinopathy associated c-Abl activation is __________________
2015 Transactions of the American Society for Neurochemistry ®
accompanied by phosphorylation and deactivation of Hdm2/Mdm2. Because Hdm2/Mdm2 is an E3-ubiquitin ligase that promotes degradation of p53, c-Abl activation would promote p53-dependent cell death in PD. In addition to the potential effects of p53 on cell death, our studies indicate that increased activation of p53 might be responsible for alpha-synucleinopathy associated autophagy defect. In neuronal cells, inhibition of c-Abl or p53 can stimulate authophagy. Significantly, treatment of HuaS(A53T) Tg mouse model with c-Abl inhibitor, nilotinib, can reduce disease associated activation of p53 and relieve autophagic defects. Further, c-Abl inhibitor can significantly delay the onset of late onset disease associated with aging of HuaS(A53T) Tg mouse model as well as the rapid onset disease caused by alpha-synuclein fibril innoculation of HuaS(A53T) Tg mice. Our studies indicate that pathological effects of alpha-synucleinopathy-dependent c-Abl activation is mediated by p53 and that inhibition of c-Abl, Mdm2, and/or p53 could provide disease modifying therapies for PD and other alpha-synucleinoapthies. Supported by NS38065, NS086074, and Plimpton Fund.
S17-03 REGULATION AND FUNCTION OF NEURONAL SURVIVAL FACTOR BY MULTIPLE STRESS ORGANELLES IN PARKINSON’S DISEASE Zixu Mao 1, Qian Yang 1, 2, Hua She 1, Wenming Li 1, Li Gao 2 Guodong Gao 2 1 Emory University School of Medicine, Pharmacology, Atlanta, USA 2 Tangdu Hospital, The Fourth Military Medical University, Neurosurgery, Xi'an, China Neurons constantly experience stress. We investigated the mechanisms of neuronal stress response by determining the function and regulation of a neuronal survival protein myocyte enhancer factor 2D (MEF2D) in neurons and in models of Parkinson’s Disease (PD). Our studies showed that MEF2D functions in several organelles to mediate stress response. In addition to the nucleus, MEF2D resides in mitochondria to regulate complex I activity, which is impaired by toxic stress associated with PD. Oxidative condition led to MEF2D oxidization, reducing its level in the nucleus and mitochondria. Damaged MEF2D in the cytoplasm was degraded by lysosomes via chaperone-mediated autophagy to maintain its functional homeostasis. This process was vulnerable to genetic risk factor associated with PD. Therefore, various neuronal toxic conditions associated with PD interfere MEF2D function via multiple mechanisms, impairing neuronal ability to maintain homeostasis. Our findings suggest that failure at multiple organelles reduces the ability of neurons to copy with stress and may underlie in part the pathogenic process in PD.
39
S17 Stress Organelles and Neurodegenerative Diseases
S17-04 MITOCHONDRIAL HOMEOSTASIS AND DENDRITIC STABILIZATION: BEYOND MITOPHAGY Charleen Chu University of Pittsburgh, Pathology, Pittsburgh, USA The sequestration and degradation of damaged mitochondria by mitophagy represents a major mechanism ensuring the maintenance of high quality mitochondria. We found that mitophagy is triggered in multiple models of Parkinson’s disease to include complex I inhibitors, oxidative neurotoxins, PINK1 deficiency and mutant __________________
2015 Transactions of the American Society for Neurochemistry ®
LRRK2 expression. However, data derived from inhibiting autophagy suggest that the mitophagic response to stress functions as a double-edged sword in primary neurons. Excessive mitochondrial clearance from dendrites that is not balanced by adequate replacement results in neurodegeneration. Our data indicate that the mitochondrial kinase PINK1 plays multiple roles in regulating autophagy and mitochondrial turnover. In neuronal cells with functioning mitochondria, PINK1 supports dendrite extension in part by promoting anabolic, and suppressing catabolic, intracellular signaling pathways.
40
S18 Cys-loop Receptors in Neurotransmission: Structural Aspects of Regulation by Exogenous and Endogenous Compounds S18-01
S18-02
GABA-A RECEPTOR SUBTYPES - RECENT DEVELOPMENTS IN PHARMACOLOGY Margot Ernst Medical University of Vienna, Department of Molecular Neurosciences, Vienna, Austria
INTERACTION OF NICOTINIC RECEPTORS WITH THREEE-FINGER NEUROTOXINS AND LY6 PROTEINS Victor Tsetlin 1, Igor Kasheverov 1, Ekaterina Lyukmanova 1 Yuri Utkin 1, Dmitry Dolgikh 1 1 Institute of Bioorganic Chemistry, Department of molecular basis of neurosignaling, Moscow, Russia 2 Institute of Bioorganic Chemistry, Department of Bioengineering, Moscow, Russia
GABA-A receptors are the most important inhibitory neurotransmitter receptors in the mammalian central nervous system. These GABA gated chloride channels possess many allosteric binding sites, at which ligands enhance or reduce GABA induced currents, or elicit GABA independent currents. GABA-A receptors are targets of many clinically used medications for the treatment of a broad range of conditions: Sleeping aids, anxiolytics, anti-epileptics and narcotics are well established. More than thirty GABA-A receptor subtypes have been described and additional ones will likely be identified in the future. The question which receptor subtype mediates which physiological function is under intense research. Compounds that interact with these receptors either do this in a subtype-selective manner, or broadly und unselectively. Selectivity can occur at the level of binding, ligands in this case bind to a single subtype or to a small group of subtypes with high affinity, and with low affinity or not at all to other subtypes. Functional selectivity, in contrast, occurs if ligands bind at multiple subtypes, but elicit modulatory effects only in a single subtype. Unselective interaction leads to either a general increase, or a decrease of the action of GABA, and to a range of partly overlapping pharmacological effects such as sedation, anxiolysis, muscle relaxation and anticonvulsant effects if GABA action is enhanced. The development of highly subtype selective ligands holds the promise of medications with fewer side effects such as anxioselective compounds. Moreover, selective agents allow development of novel medications potentially useful in conditions that so far were not addressed by targeting of GABA-A receptors such as pain, depression, schizophrenia, cognitive deficits, addiction/ alcoholism and more. We identified a novel binding site on these receptors, and present the first roadmap of its pharmacology obtained by two electrode voltage clamp experiments using recombinant receptor subtypes. Selective ligands of that site and their possible applications as research compounds and as leads for a future drug development will be presented. Recent structural data accelerates atomic level insight on binding determinants, homology models will be discussed.
2015 Transactions of the American Society for Neurochemistry ®
Our work addresses different binding modes of various threefinger proteins (3FP) to nicotinic acetylcholine receptors (nAChRs). These 3FPs include snake venom proteins !- and "-neurotoxins, and some non-conventional neurotoxins. 3F-folding is also a feature of Ly6 proteins, some of them being endogenous modulators of nAChR activities. The talk covers structural analysis of these 3FP classes and their interactions with nAChRs. !-Neurotoxins (like !bungarotoxin) helped to isolate Torpedo nAChR and acetylcholinebinding protein (AChBP) and are still widely used in pharmacology. We discovered dimeric !-cobratoxin wherein two !-cobratoxins joined by 2 intermolecular S-S-bonds; this post-translational modification did not abolish blocking of !7 and muscle-type nAChRs, but created capacity to block neuronal !3#2 nAChRs (Osipov et.al., J.Biol.Chem. 2008, 2012). Snake “non-conventional” 3FPs structurally are most close to the Ly6 proteins since they both contain 5th disulfide not in the central loop II (as !-bungarotoxin), but in N-terminal loop I. Ly6 protein Lynx1 from mammalian brain has a GPI anchor attaching it close to nAChRs, information about Lynx1 modulation of nAChRs being available mostly from over-expression or knock-out of the respective gene. A water-soluble form of Lynx1 lacking the GPI anchor (ws-Lynx1) was prepared and found to bind to the classical binding site for agonists/competitive antagonists at AChBP and muscle-type nAChR, but outside it at neuronal nAChRs; several ws-Lynx1 residues involved in binding to its distinct targets were identified (Lyukmanova et al., J.Biol.Chem., 2011, 2013). A common feature of ws-Lynx1 and non-conventional toxin WTX is their µM affinity for different nAChRs and allosteric interactions with muscarinic acetylcholine receptors. Contrary to high-affinity (nM) and irreversible binding of !-neurotoxins, ws-Lynx1 can be easily washed out. Thus, our work revealed both common and opposite features in binding modes for the two classes of 3FPs which may be the reason of their different types of action.
41
S18 Cys-loop Receptors in Neurotransmission: Structural Aspects of Regulation by Exogenous and Endogenous Compounds
S18-03
S18-04
PRESYNAPTIC !7 NICOTINIC ACH RECEPTORS ENHANCE HIPPOCAMPAL MOSSY FIBER GLUTAMATERGIC TRANSMISSION VIA PKA ACTIVATION Jerrel Yakel National Institute of Environmental Health Sciences/NIH, Neurobiology, Durham, USA
BACTERIAL AND BACTERIAL-HUMAN CHIMERAS AS MODELS TO STUDY THE MOLECULAR MECHANISMS GOVERNING PENTAMERIC LIGAND-GATED ION CHANNELS Pierre-Jean Corringer 1, 2 1 Institut Pasteur, Neurosciences, Paris, France 2 CNRS, UMR2182, Paris, France
Nicotinic acetylcholine receptors (nAChRs) are expressed widely in the CNS, and mediate both synaptic and perisynaptic activities of endogenous cholinergic inputs and pharmacological actions of exogenous compounds (e.g. nicotine and choline). Behavioral studies indicate that nicotine improves such cognitive functions as learning and memory. However, the mechanism of nicotine’s action on cognitive function remains elusive. We performed patch-clamp recordings from hippocampal CA3 pyramidal neurons to determine the effect of nicotine on mossy fiber glutamatergic synaptic transmission. We found that nicotine, via activation of the !7 nAChR, strongly potentiated the amplitude of evoked EPSCs (eEPSCs), and reduced the EPSC paired-pulse ratio. BAPTA applied postsynaptically failed to block the action of nicotine and NS1738, suggesting again a presynaptic action of the !7 nAChRs. We also observed !7 nAChR-mediated calcium rises at mossy fiber giant terminals, indicating the presence of functional !7 nAChRs at presynaptic terminals. The potentiating effect of !7 nAChR activation on eEPSCs was abolished by inhibition of protein kinase A. In cultured hippocampal neurons, activation of the !7 nAChR increased the levels of cytoplasmic cyclic adenosine monophosphate (cAMP) via a direct activation of the calcium- and calmodulinactivated adenylyl cyclase, AC1; this form of AC is highly expressed in the dendritic arbors of the dentate gyrus and the mossy fiber projections. Our findings indicate that activation of !7 nAChRs at presynaptic sites, via a mechanism involving PKA, plays a critical role in enhancing synaptic efficiency of hippocampal mossy fiber transmission. Furthermore this provides the first direct evidence to link activation of !7 nAChRs to a cAMP rise, which defines a new signaling pathway employed by !7 nAChRs. Our study sheds light into the molecular mechanisms of the positive cognitive actions of !7 nAChR agonists and development of therapeutic treatments for cognitive impairments.
2015 Transactions of the American Society for Neurochemistry ®
Pentameric channel-receptors, including nicotinic acetylcholine and GABAA receptors, play a key role in fast excitatory and inhibitory transmission in the nervous system and are the target of numerous therapeutic and addictive drugs. They carry several neurotransmitter binding sites which govern the opening of a transmembrane ion channel. Extensively expressed in animals, they were found in several bacteria, especially the homolog from the cyanobacteria Gloeobacter violaceus (GLIC) which functions as a proton-gated ion channel. The simplified architecture of this archaic homologue, as well as its prokaryotic origin, allowed solving its Xray structure in two closed and one open conformation. This suggests that channel opening occurs through symmetrical quaternary twist and “blooming” motions, together with tertiary deformation, according to a global transition that couples channel opening with reorganization of the binding pockets for neurotransmitters and allosteric effectors. In addition, we generated a functional chimera made up of the extracellular domain of GLIC coupled to the transmembrane domain of the human alpha1 glycine receptor. Combined structural and functional studies give insights into the allosteric mechanisms operating in these integral membrane proteins, and help understanding the molecular mechanisms of allosteric mutations, notably those that produce hyperekplexia.
42
S19 Mechanisms of Cell Death in Inflammatory Processes S19-01 NEUROPROTECTIVE EFFECTS VIA PERIPHERAL ADMINISTRATION OF XPRO1595 TO BLOCK SOLUBLE TNF SIGNALING IN A RAT MODEL OF PD Malu Tansey 1, Christopher J. Barnum 1, Xi Chen 1, Jaegwon Chung 1 Jianjun Chang 1, Martha Williams 1, Nelly Grigoryan 1 Raymond J. Tesi 2 1 Emory University, Physioloy, Atlanta, USA 2 FPRT Bio, Inc, Scranton, USA Parkinson’s disease (PD) is a complex multi-system age-related neurodegenerative disorder. Targeting the ongoing neuroinflammation in PD patients is one strategy postulated to slow down or halt disease progression. Proof-of-concept studies from our group demonstrated that selective inhibition of soluble Tumor Necrosis Factor (solTNF) by intranigral delivery of dominant negative TNF (DN-TNF) inhibitors reduced neuroinflammation and nigral dopamine (DA) neuron loss in endotoxin and neurotoxin rat models of nigral degeneration. As a next step toward human clinical trials, we aimed to determine the extent to which peripherally administered DN-TNF inhibitor XPro®1595 could: i) cross the blood-brain-barrier in therapeutically relevant concentrations, ii) attenu- ate neuroinflammation (microglia and astrocyte), and iii) mitigate loss of nigral DA neurons in rats receiving a unilateral 6hydroxydopamine (6-OHDA) striatal lesion. Rats received unilateral 6-OHDA (20 micrograms into the right striatum). Three or 14 days after lesion, rats were dosed with XPro®1595 (10mg/kg in saline, subcutaneous) every third day for 35 days. Forelimb asymmetry was used to assess motor deficits after the lesion; brains were harvested 35 days after the lesion for analysis of XPro®1595 levels, glial activation and nigral DA neuron number. Peripheral subcutaneous dosing of XPro®1595 achieved plasma levels of 1–8microgram/mL and CSF levels of 1–6ng/mL depending on the time the rats were killed after final XPro®1595 injection. Irrespective of start date, XPro®1595 significantly reduced microglia and astrocyte number in SNpc whereas loss of nigral DA neurons was attenuated when drug was started 3, but not 14 days after the 6-OHDA lesion. Our data suggest that systemically administered XPro®1595 may have disease-modifying (neuroprotective) potential in the early stages of PD prior to significant degeneration of dopaminergic neurons.
S19-02 CELL DEATH PATHWAYS IN HOST DEFENSE, INFLAMMATION, AND DEVELOPMENTAL FAILURE William Kaiser Emory University, Microbiology and Immunology, Atlanta, USA The kinases RIP1 and RIP3 coordinate apoptosis, necrosis, and inflammation. Evidence indicating programmed necrosis occurs in natural settings emerged with identification that multiple pathogen sensor pathways induce RIP3-dependent necrosis and with recognition that pathogens have evolved to directly subvert this __________________ 2015 Transactions of the American Society for Neurochemistry ®
death pathway for pathogenesis. Though important in host defense, the potential deleterious consequence of RIP1/RIP3 dysregulation in vivo is perhaps most starkly illustrated by my finding that Caspase-8 suppression of RIP1/RIP3 is a requisite for mammalian development, T cell proliferation, hematopoiesis, and suppression of fatal chronic inflammation. The molecular basis of programmed necrosis and the role of this pathway in host defense will be discussed as well as strategies for therapeutic intervention.
S19-03 TARGETING ASTROCYTE SIGNALING PATHWAYS PROTECTS HIPPOCAMPAL SYNAPTIC FUNCTION FOLLOWING TRAUMATIC BRAIN INJURY Christopher Norris, Jennifer Furman, Susan Kraner, Esther Putman Melanie Pleiss, Pradoldej Sompol, Stephen Scheff University of Kentucky, Sanders-Brown Center on Aging, Lexington, USA Astrocyte activation is strongly associated with both acute and chronic neurodegenerative conditions. Nevertheless, the impact of activated astrocytes on neurologic function remains poorly understood. The activated astrocyte phenotype appears to be regulated, in part, by the protein phosphatase calcineurin (CN) and the CN-dependent transcription factor, NFAT (Nuclear Factor of Activated T Cells). Here, we report evidence for increased DNA binding of NFATs 1 and 4, but not for NFATs 2 and 3 in adult rat hippocampus 7 days after a unilateral cortical contusion injury (CCI). Confocal microscopy showed that the increased expression of NFAT4, but not necessarily NFAT1, was most strongly associated with activated astrocytes in injured hippocampal tissue, especially in CA1. To assess the impact of elevated CN/NFAT expression/activity in astrocytes following injury, we injected adeno-associated virus (AAV) vectors containing the NFAT inhibitor, VIVIT, or a control EGFP construct into the hippocampus of adult Sprague Dawley rats 2 months prior to receiving CCI. The human GFAP promoter (Gfa2) was used to restrict transgene expression to astrocytes. At 7 days post-CCI, hippocampal slices were harvested for the assessment of CA3-CA1 synaptic strength and plasticity under investigator-blind conditions. AAV-Gfa2-EGFP control rats exhibited significantly reduced basal synaptic strength and robust LTD in injured relative to unijured hippocampus (and also compared to sham animals). In contrast, synaptic strength in AAV-Gfa2-VIVIT rats was very comparable across the injured and uninjured hemispheres and nearly indistinguishable from sham rat slices. Moreover, slices from AAVGfa2-VIVIT rats showed almost no LTD. Finally, AAV-Gfa2VIVIT provided significant protection from the loss of key synaptic proteins (e.g. GluR1, PSD-95, and synapsin 1) following injury. The results demonstrate that aberrant CN/NFAT activity in hippocampal astrocytes contribute to synaptic dysfunction associated with acute neural injury. Ongoing studies are determining the extent to which astrocytic CN/NFAT activity regulates neuroinflammatory markers following CCI.
43
S20 Neuron-Oligodendrocyte Interactions in Development and Disease S20-01 ACTIVITY-BIASED SELECTION OF AXONS FOR MYELINATION IN VIVO Bruce Appel 1, Jacob Hines 1, Andrew Ravanelli 1, Rani Schwindt 1 Ethan Scott 2 1 University of Colorado School of Medicine, Pediatrics, Aurora, USA 2 The University of Queensland, School of Biomedical Sciences, St. Lucia, Australia An essential feature of vertebrate neural development is the ensheathment of axons with myelin, an insulating membrane formed by oligodendrocytes. However, not all axons are myelinated and the mechanisms that select some axons but not others for myelination are not known. Using zebrafish as a model system, we investigated whether neuronal activity influences axon selection for myelination. Pharmacological and genetic inhibition of activity and axonal exocytosis reduced the frequency with which a specific spinal cord axon was wrapped, indicating that activity biases axon selection. In vivo time-lapse microscopy revealed that only 25% of nascent sheaths are initially stabilized during normal myelination, and that initial ensheathment does not require activity-dependent secretion. Instead, nascent sheaths wrapping silenced axons are retracted more frequently. To investigate the molecular basis for activity-dependent sheath stabilization, we tested expression of myelin genes. Notably, transcripts encoding Myelin basic protein (Mbp) were reduced approximately 50% in larvae in which neuronal activity was silenced. We propose that axon selection is biased by activity-dependent Mbp production and stabilization of myelin sheaths following indiscriminate sheath initiation.
S20-02 NEONATAL BRAIN INJURY CAUSES ABNORMALITIES IN NEURON-NG2 CELL SYNAPTIC COMMUNICATION Vittorio Gallo Children's Natl. Med. Ctr., Children's Research Inst., Washington, USA SA major cause of chronic disability in survivors of premature birth is diffuse white matter injury (DWMI) and hypomyelination. Altered development of the WM is directly associated with adverse outcomes, including cerebral palsy, cognitive delay and neurobehavioral problems. The cellular pathophysiology underlying DWMI and abnormal myelination is complex and not fully understood. WM glia, and particularly oligodendrocytes (OLs) and their progenitors (OPCs), are susceptible to hypoxic injury that often occurs in premature birth. In an animal model of hypoxia (HX)-induced global WMI, we demonstrated that OPCs display delayed maturation, which results in abnormal myelination and altered WM func__________________
2015 Transactions of the American Society for Neurochemistry ®
tion. We have begun to uncover major aspects of the cellular pathology underlying HX-induced hypomyelination, including enhanced OPC proliferation associated with decreased differentiation, and disrupted myelin ultrastructure. We have also defined potential interventions that induce complete recovery of functional myelination and WM-associated behaviors. In my presentation, I will discuss recent studies that define mechanisms that cause OPC proliferation and delayed OL differentiation after HX. In particular, I will focus on neuron-OPC synapses in different white matter region of the CNS, and their role in normal OPC development and in developmental abnormalities observed after HX. These studies will not only shed light on crucial cellular mechanisms of HX-induced delay in WM maturation, but might also lead to the development of new therapeutic approaches aimed at lessening the long-term neurological sequelae of premature birth. Supported by NINDS.
S20-03 IMMUNE-MEDIATED DEMYELINATION AND NEURONOPATHY IN AN AUTOIMMUNE MULTIPLE SCLEROSIS MODEL David Pleasure, Athena Soulika, Peter Bannerman, Emily Mills Ko Monica Moreno UC Davis School of Medicine, Neurology and Pediatrics, Davis, USA The interrelationships between immune-mediated acute oligodendroglial pathology and treatment-refractory chronic axonopathy and neuronopathy in multiple sclerosis (MS) have not been fully clarified. We approached this issue in a mouse MS model, MOG peptide-induced experimental autoimmune encephalomyelitis (EAE). As in MS, spinal cord and subcortical white matter of MOG peptide EAE mice develop subacute focal inflammatory demyelinative lesions, followed by progressive, dissseminated axon loss. Motor cortex also demyelinates, accompanied by a diminution in upper motor neuron perikaryal presynaptic contacts and a reversible loss of upper motor neuronal differentiation markers. Demyelination and axon loss in these mice are diminished by two interventions that inhibit CNS accumulation of M1 (classically activated) macrophages: astroglial conditional deletion of the monocyte chemokine CCL2, or depletion of systemic monocytes by intravenous infusions of clodronate-containing liposomes. In contrast, astroglial conditional deletion of the lymphocyte chemokine CXCL10, though minimizing demyelination, neither suppresses CNS accumulation of M1 macrophages nor prevents long-term axon loss. A current focus of our laboratory is on the relative contributions of monocyte-derived vs microglia-derived M1 macrophages to axonopathy and neuronopathy in EAE and, by extension, in MS.
44
S20 Neuron-Oligodendrocyte Interactions in Development and Disease
S20-04 OLIGODENDROLGIA SUPPORT OF NEURONS AS A BASIS FOR NEURODEGENERATION INITIATION Jeffrey Rothstein Johns Hopkins University, Neurology, Baltimore, USA Studies of human diseases and animal models suggest that oligodendroglia play a supportive role for axons (and neurons). The exact nature of this supportive role was not clear until recent evidence demonstrated that oligodendroglia provide critical metabolic support to axons in the form of monocarboxylates (i.e., lactate). Alterations of the monocarboxylate transporter on oligodendroglia (i.e., MCT1) produce axon and neuron degeneration. This transporter also appears to be important for human disease since MCT1 is downregulated in motor cortex of amyotrophic lateral sclerosis (ALS) patients and spinal cord of a mouse model of __________________
2015 Transactions of the American Society for Neurochemistry ®
ALS. Additionally, protecting oligodendroglia in this model of ALS, by removing the mutant gene specifically from oligodendroglia, attenuates the loss of MCT1, delays onset, and prolongs survival. There is also accumulating evidence that oligodendroglia may be important contributors to neuron injury in other human diseases, including progressive multiple sclerosis and multiple system atrophy. Despite these advances, little is known about the source of oligodendroglial lactate, if cellular linkage between glia is essential for its transport, whether components of the lactate pathway are responsive to neuron/axonal hyperactivity, whether physiologic neuronal activation is dependent on oligodendroglial metabolic support, or whether failure of this oligodendroglial-centric metabolic pathway contributes to motor neuron degeneration in ALS. We will discus new data that comprehensively evaluates the metabolic linkage from oligodendroglia to axons in normal physiology and ALS.
45
S21 Glia Amino Acid Transporters in Health and Disease S21-01
S21-02
FUNCTIONAL CROSS-TALK BETWEEN DIFFERENT SYSTEMS OF GLUTAMATE TRANSPORT AND METABOLISM IN THE SPINAL CORD Georgi Gegelashvili 1, 2, Ole Jannik Bjerrum 2 1 Ilia State University, Institute of Chemical Biology, Tbilisi, Georgia 2 University of Copenhagen, Drug Design and Pharmacology, Copenhagen, Denmark
THE SLC38 FAMILY OF GLUTAMINE TRANSPORTERS AND THEIR CONTRIBUTION TO THE GLUTAMATE/GABA-GLUTAMINE CYCLE Farrukh Chaudhry University of Oslo, Institute of Basic Medical Sciences, Oslo, Norway
Neurons sensing noxious stimuli and conducting pain signals from periphery to the spinal cord are predominantly glutamatergic. Members of the SLC1A family of high-affinity glutamate transporters, GluTs (GLAST/EAAT1, GLT1/EAAT2, EAAC1/EAAT3, and EAAT4) are differentially expressed in sensory neurons and surrounding glial cells. These plasma membrane proteins together with glutamate/cystine exchanger, xCT, are responsible for fine tuning of glutamate concentrations at glutamate receptors and, thus, modulation of excitatory signalling in the spinal cord. Several compounds, believed to affect high-affinity glutamate transport system, including therapeutically promising beta-lactams, have been examined in an in vivo model of neuropathic pain. Both pain behavior and glutamate transporter expression have been investigated in this model at various time-points. For the first time, changes in the expression of rare splice variants of glial glutamate transporter GLT1 have been demonstrated in rats with induced neuropathic pain. The dynamics of expression of high-affinity glutamate transporter subtypes and pattern of nociceptive pointed at complex relations between the functional state of glutamate transport system and the levels of analgesia provided by the tested compounds. The glutamate transport system has been also studied in co-cultures of dorsal root ganglion (DRG) neurons and spinal glial cells. In this in vitro model system, that partially recapitulates primary pain signaling path, both glial and neuronal glutamate carrier proteins undergo changes in expression, as well as post-translational modifications, including proteolytic truncation. Such regulated cleavage depends on phosphorylation state of intracellular domains of glutamate carrier proteins. These functional modifications alter cell surface targeting of glutamate transporters, as well as elicit downstream signaling. This also affects GluT interaction with other components of the glutamate sensing- and metabolizing machinery, including mechanisms of refilling and recycling of synaptic vesicles in DRG neurons and spinal glia. The elucidated regulatory pathways seem to provide fine tuning of excitatory signaling in the spinal cord and, can, thus, emerge as prospective drug targets for chronic pain treatment.
Replenishment of the fast neurotransmitters glutamate and GABA in the central nervous system has been enigmatic. According to the glutamate/GABA-glutamine cycle (GGG cycle) hypothesis glutamate and GABA are released exocytotically and activate their specific receptors. Such neuronal signaling is terminated by transport of the neurotransmitters out of the synaptic cleft by specific plasma membrane transporters to a large extent into astroglial cells. Here they are converted to glutamine which is then shuttled back to the nerve terminals for resynthesis of these neurotransmitters. Although existence of such a cycle is widely accepted, the transport of glutamine across astroglial and neuronal membranes has eluded characterization and regulation of the GGG cycle remains unknown. We have provided compelling evidence that the system N transporters SN1 (Slc38a3) and SN2 (Slc38a5) mediate Na+-dependent transport of glutamine which is coupled to counter-transport of H+. This makes the transport electroneutral and allows these transporters to work in both directions. In particular, it allows SN1 and SN2 to release glutamine from astroglial cells. The homologoues system A transporters SAT1 (Slc38a1) and SAT2 (Slc38a2) are localized on the cell membranes of GABAergic and glutamatergic neurons, respectively, and mediate Na+-dependent transport of glutamine. As this is not coupled to counter-transport of H+ such electrogenic transport is unidirectional and allows for accumulation of glutamine in neurons for resynthesis of GABA and glutamate. We now show that the complementary expression of the system A and system N transporters is widespread and allows for intercellular and interorgan shuttling of glutamine to sustain glutamine metabolism involved in a variety of pathways. Furthermore, we show that the activity and membrane trafficking of the transporters of the Slc38 family are dynamically regulated by ions and intracellular phosphorylation events with potential impact on neurotransmitter synthesis and synaptic plasticity.
S21-03 GLAST-DEPENDENT CONTROL OF THE GLUTAMATE/GLUTAMINE SHUTTLE: MOLECULAR TARGETS OF POLLUTANTS Arturo Ortega, Luz María Del Razo, Andrea De Vizcaya Cinvestav-IPN, Toxicology, Mexico City, Mexico L-Glutamate (Glu) is the major excitatory neurotransmitter in the vertebrate Central Nervous System. The extracellular levels of this amino acid are tightly regulated through the activation of a family of Na+ -dependent excitatory amino acid transporters (EAATs). Glia cells either through EAAT-1 (GLAST) or EAAT-2 (GLT-1) carry out
2015 Transactions of the American Society for Neurochemistry ®
46
S21 Glia Amino Acid Transporters in Health and Disease most of Glu uptake. Within the cerebellum, GLAST is responsible of the removal of Glu and is expressed abundantly in Bergmann glia cells, which completely ensheat the parallel fibers-Purkinje cells synapses in the molecular layer. The Glu-glutamine (Gln) cycle, also known as the Glu/Gln shuttle consists of glial uptake of the glutamate released by neurons into the synaptic cleft where it is converted into glutamine by the glial-enriched enzyme glutamine synthetase. Subsequently, glutamine is released into the extracellular space to be taken up by glutamatergic neurons and enter a new cycle of transmitter synthesis. Previous work from our group has demonstrated a Glu-dependent GLAST down regulation of its activity and gene transcription. Moreover, Glu uptake also regulates a physical and activity coupling between GLAST and the Gln transporter SNAT3 (sodium dependent neutral amino acid transporter 3). These results clearly suggest a fundamental role of glia cells in Glu homeostasis. With this in mind, we decided to explore the possibility that pollutants, such as particulate matter (PM) or fluorine could disrupt amino acid transport and by these means exert their neurotoxic effects. Indeed this is the case. These and other recent results that reinforce the involvement of glia cells in synaptic transmission in what is currently known as the tripartite synapse will be discussed.
mechanisms associated with this protection have yet to be elucidated. Given that E2 increases astrocytic expression of glutamate transporter-1 (GLT-1), which would prevent excitotoxic-induced neuronal death, we proposed that GPR30 mediates E2 action on GLT-1 expression. To investigate this hypothesis, we examined the effects of G1, a selective agonist of GPR30, and GPR30 siRNA on astrocytic GLT-1 expression, as well as glutamate uptake in rat primary astrocytes, and explored potential signaling pathways linking GPR30 to GLT-1. G1 increased GLT-1 protein and mRNA levels, subject to regulation by both MAPK and PI3K signaling. Inhibition of TGF-" receptor suppressed the G1-induced increase in GLT-1 expression. Silencing GPR30 reduced the expression of both GLT-1 and TGF-" and abrogated the G1-induced increase in GLT-1 expression. Moreover, the G1-induced increase in GLT-1 protein expression was abolished by a protein kinase A inhibitor and an NF#B inhibitor. G1 also enhanced cAMP response element-binding protein (CREB), as well as both NF-#B p50 and NF-#B p65 binding to the GLT-1 promoter. Finally, to model dysfunction of glutamate transporters, manganese was used, and G1 was found to attenuate manganese-induced impairment in GLT-1 protein expression and glutamate uptake. Taken together, the present data demonstrate that activation of GPR30 increases GLT-1 expression via multiple pathways, suggesting that GPR30 is worthwhile as a potential target to be explored for developing therapeutics of excitotoxic neuronal injury.
S21-04 GPR30 REGULATES GLUTAMATE TRANSPORTER GLT-1 EXPRESSION IN RAT PRIMARY ASTROCYTESITLE Michael Aschner 2, Pratap Karki 1, Eunsook Lee 1 1 Meharry Medical College, Neurology, Nashville, USA 2 Albert Einstein College of Medicine, Molecular Pharmacology, Bronx, USA The G protein-coupled estrogen receptor GPR30 contributes to the neuroprotective effects of 17!-estradiol (E2); however, the __________________
2015 Transactions of the American Society for Neurochemistry ®
47
S22 Calpain Inhibitors in Preclinical Models of Neurodegenertion and Neurotrauma S22-01
S22-02
EFFECTS OF CALPAIN INHIBITION IN TRAUMATIC BRAIN INJURY AND AXONAL DEGENERATION Kathryn Saatman University of Kentucky, Spinal Cord and Brain Injury Research Center, Lexington, USA
ISCHEMIA ACTIVATES CALPAINS IN EXPERIMENTAL RETINAL NEUROPATHIES
Traumatic brain injury (TBI) is a leading cause of death and disability in the United States. Moderate to severe TBI is often associated with long-term motor and cognitive deficits, but even mild TBI can result in learning and memory impairment. Trauma-induced neuronal death and axonal injury result from a multitude of secondary injury mediators, including early calcium dysregulation sufficient to trigger activation of calpains. Many studies have demonstrated calpain activation and concomitant calpain-mediated proteolytic degradation of cellular proteins following TBI. Targeting this calpain cascade through the administration of exogenous calpain inhibitors appears effective in decreasing substrate proteolysis and improving behavioral outcome in experimental TBI, but typically fails to protect against neuronal death. As an alternate approach to avoid potential problems with CNS delivery and bioavailability of pharmacological compounds, we used a transgenic mouse in which human calpastatin (hCAST) is overexpressed in order to investigate the effects of calpain inhibition in a mouse model of contusion brain injury. Constitutive calpastatin overexpression attenuated levels of calpain-generated proteolytic fragments of spectrin, collapsin response mediator protein-2, and the voltage-gated sodium channel within the first 24 h after TBI. Brain-injured mice overexpressing calpastatin also exhibited improved motor and cognitive function compared to wildtype mice. However, regional neurodegeneration was unaffected by calpastatin overexpression, suggesting that behavioral improvements may be related to other aspects of neuronal function or plasticity. Because TBI is associated with widespread axonal injury, this same transgenic mouse model was used to test the efficacy of calpastatin overexpression to attenuate Wallerian degeneration. Degeneration of both central and peripheral nerves was slowed after transection, and the structure, but not the function, of the neuromuscular junction was preserved. While calpains represent a viable therapeutic target for reducing posttraumatic morbidity, the cellular mechanisms through which calpain inhibition improves neurobehavioral function remain unclear. To achieve neuroprotection with behavioral efficacy, calpain inhibitors may need to be coupled with another neuroprotective treatment in a combination therapy approach.
2015 Transactions of the American Society for Neurochemistry ®
Yoshiyuki Tamada 1, R. Suzuki 1, T. Oka 1, E. Nakajima 1, 2 T. Shearer 2, M. Azuma 1, 2 1 Senju Pharmaceutical Co., Research & Development Division, Kobe, Japan 2 Oregon Health & Science University, Health & Science, Portland, OR Retinal ischemia is a common clinical condition and the major cause of human visual impairment and blindness. For example, in anterior ischemic optic neuropathy (AION), infarction of capillaries damages the anterior optic nerve and leads to sudden loss of vision. Common cardiovascular risk factors are associated with AION, but no clinically effective treatment exists. Acute angle-closure glaucoma also causes ischemic retinal neuropathy. Calcium overload, glutamate toxicity, and oxidative stress are hypothesized to cause these neuropathies. Experimental models reproduce some of the pathological features observed in human AION and acute angle-closure glaucoma. The current study measured the involvement of calpain proteolytic enzymes in several experimental models of ischemic retinal neuropathy. AION was produced in rats by intravenous injection of Rose Bengal and photoactivation at the optic nerve head by irradiation with a green laser. Acute ocular hypertension was produced in rats by elevation of intraocular pressure to 110 mm Hg for 40 min. These procedures caused increased retinal calcium, activation of calpains, and proteolysis of calpain substrates. After appearance of TUNELpositive cells, retinal ganglion cells were lost. Calpain inhibitor SNJ-1945 inhibited proteolysis of calpain substrates and ameliorated retinal degeneration. Retinal neuronal cells and retinal tissues were also cultured under hypoxic conditions. As observed in the in vivo models, activation of calpain, proteolysis of calpain substrates, and neuronal cell death were observed. Calpain inhibitor SNJ-1945 reduced these changes. Calpain activation appears to play an important role in retinal ischemia in experimental models. Calpain inhibitor SNJ-1945 is a possible candidate drug for delaying progression of retinal degeneration in patients with ischemia-induced retinal neuropathy. Dr. Shearer receives a research contract and consulting fees from, and Drs. Azuma and Nakajima are employees of Senju Pharmaceutical Co., Ltd.
48
S22 Calpain Inhibitors in Preclinical Models of Neurodegenertion and Neurotrauma
S22-03
S22-04
CHRONIC INTERMITTENT ETHANOL-INDUCED AXON AND MYELIN DEGENERATION IS ATTENUATED BY CALPAIN INHIBITION Supriti Samantaray 1, Varduhi Knaryan 1, Kaushal Patel 1 Patrick Mulholland 2, 3, Howard Becker 2, 3, 4, Naren Banik 1, 4 1 Medical University of South Carolina, Neurosciences, Neurology, Charleston, USA 2 Medical University of South Carolina, Neurosciences, Institute of Psychiatry, Charleston, USA 3 Medical University of South Carolina, Charleston Alcohol Research Center, Charleston, USA 4 Ralph H Johnson Veterans Medical Center, Research Services, Charleston, USA
CALPAIN INHIBITORS: A SURVEY OF COMPOUNDS IN THE PATENT AND SCIENTIFIC LITERATURE Isaac Donkor The University of Tennessee Health Science Center, Pharmaceutical Sciences, Memphis, USA
Chronic alcohol consumption causes multifaceted damage to the central nervous system (CNS), underlying mechanisms of which are gradually being unraveled. In our previous studies, activation of calpain, a calcium-activated neutral protease has been found to cause detrimental alterations in spinal motor neurons following ethanol (EtOH) exposure in vitro. However, it is not known whether calpain plays a pivotal role in chronic EtOH exposure-induced structural damage to CNS in vivo. To test the possible involvement of calpain in EtOH-associated neurodegenerative mechanisms the present investigation was conducted in a well-established mouse model of alcohol dependence - chronic intermittent EtOH (CIE) exposure and withdrawal. Our studies indicated significant loss of axonal proteins (neurofilament light and heavy, 50-60 %), myelin proteins (myelin basic protein, 20-40 % proteolipid protein, 25 %) and enzyme (2', 3'-cyclic-nucleotide 3'- phosphodiesterase, 21-55 %) following CIE in multiple regions of brain including hippocampus, corpus callosum, cerebellum, and importantly in spinal cord. These CIE-induced deleterious effects escalated after withdrawal in each CNS region tested. Increased expression and activity of calpain along with enhanced ratio of active calpain to calpastatin (sole endogenous inhibitor) was observed after withdrawal compared to EtOH exposure. Pharmacological inhibition of calpain with calpeptin (25 µg/kg) prior to each EtOH vapor inhalation significantly attenuated damage to axons and myelin as demonstrated by immunoprofiles of axonal and myelin proteins, and Luxol Fast Blue staining. Calpain inhibition significantly protected the ultrastructural integrity of axons and myelin compared to control as confirmed by electron microscopy. Together, these findings confirm CIE exposure and withdrawal induced structural alterations in axons and myelin, predominantly after withdrawal and corroborate calpain inhibition as a potential protective strategy against EtOH associated CNS degeneration.
2015 Transactions of the American Society for Neurochemistry ®
The calpain family consists of 15 isoforms of which calpain-1 (µcalpain) and calpain-2 (m-calpain) are the most studied isoforms and are referred to as conventional calpains. Structurally the conventional calpains consist of a large 80 kDa catalytic subunit and a small 30 kDa regulatory subunit. There is considerable interest in the development of calpain inhibitors (CIs) because the enzyme has been implicated in several diseases including neurodegeneration and neurotrauma. The inhibitors may be grouped into peptide-based inhibitors, peptidomimetic inhibitors, and nonpeptide inhibitors most of which contain an electrophilic functionality for covalent modification of an active site cysteine resulting in inactivation of the enzyme. Few allosteric inhibitors that inactivate calpain by binding to the pentaEF hand region of the small subunit of the enzyme are known. Several challenges must be addressed in the design of CIs as potential therapeutic agents. These challenges include: (a) selectivity of the inhibitor for calpain over other proteases; (b) selectivity of the inhibitor for one calpain isoform over others; (c) cellular stability; (d) requirement for desirable pharmacokinetic and pharmacodynamics properties; (e) and ability of the inhibitor to cross the BBB if it is intended to be used in the management of CNS pathologies such as neurodegeneration and neurotrauma. Significant progress has been made in overcoming some of these challenges among which are: (a) discovery of peptidomimetic inhibitors that penetrate the BBB to inhibit spectrin breakdown without overt acute toxicity; (b) development of strategies for selective CNS distribution of CIs; (c) discovery of CIs that combine potency, selectivity, and cytosolic stability within the same molecule; and (d) discovery of undecylenic acid, which permeates the BBB to afford potent neuroprotection. Despite these advances discovery of calpain isoform selective inhibitors continues to be a challenge in calpain inhibitor research. However, the future out look for the discovery of such inhibitors is promising due in part to the disclosure of the X-ray crystal structures of calpain with and without bound inhibitors.
49
S23 Inflammation and Myelination: A Match Made in Heaven? S23-01
S23-03
LATENT HERPESVIRUS 6 INFECTION IMPAIRS HUMAN GLIAL PROGENITOR CELL FUNCTIONS IN VITRO AND IN VIVO Margot Mayer-Proschel, Andrew Campbell, David Mock Chris Proschel University of Rochester, Biomedical Genetics, Rochester, USA
GLIAL INTERACTIONS IN INFLAMMATORY DEMYELINATION AND MYELIN REPAIR: ROLE OF GALECTIN-9 Jianrong Li Texas A&M University, Veterinary Integrative Bioscience, Texas A&M Institute for Neuroscience, College Station, USA
Human herpesvirus 6 (HHV-6), a common resident virus of the human central nervous system (CNS), has been implicated in both acute and chronic inflammatory-demyelinating diseases, including multiple sclerosis. HHV6 has been shown to infect mature oligodendrocytes and astrocytes as well as oligodendrocyte progenitor cells (OPCs). More recently it has been shown that HHV6 can establish a latency state via chromosomal integration into the host genome. Importantly, chromosomal integration of both HHV-6A and B strains (iHHV6) occurs in 1% of live births and has been associated with a number of pathologies ranging from chronic fatigue syndrome to impairment of cognitive functions. iHHV6 has also been seen in MS patients but its contribution to pathology remains unexplored. With the recent discovery of a viral protein, U94, that is expressed during the latency state, it became possible to study the impact of latent HHV6 infections on cellular functions. Using HHV6-infected or U94-transduced human OPCs, we have tested the effect of HHV6 infection on oligodendrocyte precursors using both in vitro and in vivo assays. We show that latency associated expression of viral U94 in hOPC impairs functions that are critical for myelin repair process.
Immune responses in the central nervous system (CNS) are essential protective mechanisms against pathogens and also critical for tissue reparative processes. Persistent and aberrant neuroinflammation, however, may contribute to the pathogenesis of many neurological diseases including multiple sclerosis (MS). Demyelination and axonal damage in MS are thought to be a consequence of inflammatory processes that are perpetuated by activated glia and infiltrating leukocytes. Galectin-9, a !-galactoside-binding protein and a modulator of immune responses and cell-cell interactions, has recently been implicated in MS. However, its function in the CNS remains largely unexplored. We found that galectin-9 is minimally expressed in the normal CNS, but is robustly upregulated in microglia and reactive astrocytes during acute CNS inflammation and experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Increased galectin-9 was also found in reactive astrocytes and microglia/macrophages surrounding active but not inactive MS lesions. Proinflammatory cytokines such as TNF" and IL1! released from activated microglia triggered galectin-9 production from astrocytes, which in turn acted in a feed-forward fashion to further enhance microglial TNF" production. TNF"-stimulated Lgals9+/+ astrocytes induced significantly higher percentage of encephalitogenic T-cell apoptosis and proliferation arrest as compared with Lgals9-/- astrocytes, indicating that galectin-9 negatively regulates encephalitogenic T cells. During MOG35-55-induced EAE, galectin-9 null mice exhibited worse clinical symptoms, exacerbated leukocyte infiltration, enhanced CNS Th17 responses, and increased demyelination when compared with littermate Lgals9+/+ controls. In autoimmunity-independent toxin models of CNS demyelination and remyelination, whereas Lgals9-/- mice had comparable gliosis and demyelination as Lgals9+/+ mice upon lysolecithin or cuprizone challenges, spontaneous remyelination was impaired. Immunohistochemistry analyses of oligodendroglial lineage cells revealed that galectin-9 promoted maturation of oligodendrocytes. Similarly, recombinant galectin-9 facilitated oligodendrocyte maturation in mixed glial cultures. Taken together, our results suggest a dual role for galectin-9 in suppressing Tlymphocytes in the CNS and facilitating oligodendrocyte maturation and myelin repair. Supported by NIH R01NS060017 and the National Multiple Sclerosis Society research grant RG4586
S23-02 HARNESSING THE BENEFITS OF INFLAMMATION FOR REMYELINATION Voon Wee Yong, Manoj Mishra, Khalil Rawji, Michael Keough Sam Jensen University of Calgary, Health Science Center, Calgary, Canada A robust inflammatory response manifested by microglia activation and the recruitment of macrophages is observed following demyelinating injuries to the CNS. Using the lysolecithin model of demyelination, we have characterized the inflammatory response in young and aged mice, and have assessed its impact on myelin clearance, cytotoxicity, gliogenesis, and regulation of the extracellular matrix particularly of the chondroitin sulfate proteoglycans (CSPGs). We describe our recent approaches to stimulate a beneficial inflammatory response for remyelination following lysolecithin injury, using the lipopolysaccharide-enhanced M2 cells, the CSPG-regulating fluorosamine and exercise. The benefits of an inflammatory response for the healing process can be harnessed for injuries of the CNS.
2015 Transactions of the American Society for Neurochemistry ®
50
S23 Inflammation and Myelination: A Match Made in Heaven?
S23-04 LEVERAGING MACROPHAGE ACTIVATION TO PROMOTE OLIGODENDROCYTE GENESIS Dana McTigue, Phillip Popovich, Evan Goldstein, Jamie Church Ohio State University, Neuroscience, Columbus, USA Oligodendrocytes, the myelinating cells of the CNS, are vulnerable to injury and are often lost in regions of trauma or disease, resulting in demyelination and axon pathology. Fortunately, progenitor cells are present throughout the adult gray and white matter that can replace oligodendrocytes under the appropriate conditions. These progenitors are sensitive to changes in local conditions, including expression of inflammatory mediators and growth factors. Common to regions of CNS injury is the presence of activated microglia and macrophages. Since these cells are factories of cytokines and growth factors, they can potently influence progenitor responses. Our work focuses on understanding how __________________
2015 Transactions of the American Society for Neurochemistry ®
activated microglia and macrophages alter progenitor responses in regions in regions of inflammation or trauma. Our data reveal that activating microglia or macrophages in vivo via TLR4 induces a reparative response that promotes marked progenitor proliferation and differentiation into new oligodendrocytes. Ongoing studies are analyzing factors released by these microglia and macrophages that may mediate this response. In addition, we are examining the oligogenic response after spinal cord injury (SCI) in mice that are deficient in TLR4 signaling. The data reveal that the normally robust oligodendrocyte replacement after SCI is muted in TLR4deficient mice and that expression of growth factors such as CNTF, IGF-1, FGF-2 and TGFß is altered. Thus, spontaneous reparative processes initiated after SCI by oligodendrocyte progenitors must be positively influenced by TLR4-activated microglia and macrophages. Overall this work shows that manipulating TLR4 signaling may provide a mechanism for enhancing oligodendrocyte genesis after CNS injury.
51
S24 Modulaton of ABC Transporters and APOE Levels as Therapeutic Targets for Alzheimers S24-01 KEY FUNCTIONS OF ABCA1 IN MULTIPLE TISSUES John Parks Wake Forest School of Medicine, Internal Medicine-Section on Molecular Medicine, Winston Salem, USA ATP binding cassette transporter A1 (ABCA1) is a member of a large family of membrane transporters and functions to efflux free cholesterol (FC) and phospholipid across the plasma membrane to combine with apolipoprotein A-I, the major apolipoprotein of plasma HDL, forming nascent HDL (nHDL) particles that undergo subsequent metabolism to become mature plasma HDL particles, which can then function in reverse cholesterol transport. Mutations that inactivate ABCA1 lead to Tangier disease, a disorder characterized by near absence of HDL, elevated plasma triglycerides, reduced low density lipoprotein concentration, increased storage of cholesteryl esters in macrophages. ABCA1 is expressed in most tissues and its expression is highly regulated at the transcriptional and post-translational level, making in vivo cell-specific ABCA1 function difficult to determine. We addressed this problem by generating cell-specific ABCA1 knockout (KO) mice. Using this approach, we have shown that hepatocyte ABCA1 expression is responsible for ~70-80% of the plasma HDL pool in chow-fed mice, whereas intestinal epithelial cell expression is responsible for ~20%. Subsequent studies revealed some novel and unexpected cell-specific roles for ABCA1, including defective insulin release from pancreatic beta cells, abnormal neuronal structure and function in brain, and hyper-responsiveness to proinflammatory stimuli in macrophages. One potential mechanism to explain the unexpected phenotypes of these cell-specific ABCA1 KO mice may be related to plasma membrane lipid rafts, which we have shown are increased in macrophages from macrophage-specific ABCA1 KO mice. Lipid rafts are specialized regions of the membrane that are enriched in FC and sphingolipids, consist of condensed liquid-ordered phases of membrane, and serve as platforms for signaling complexes. We also documented a direct association of macrophage FC content with membrane lipid raft content and Toll-like receptor 4 stimulated cytokine secretion. Our results suggest that ABCA1 expression may normally function to adjust plasma membrane FC, lipid raft content, signaling events, and ultimately, cellular function.
S24-02 THE THERAPEUTIC POTENTIAL OF ABCA1 FOR ALZHEIMER DISEASE Cheryl Wellington University of British Columbia, Department of Pathology and Labor, Vancouver, Canada The lipid transporter ABCA1 is best known for its role in effluxing cholesterol and phospholipids to apoA-I, the major protein component of plasma “good cholesterol” high-density lipoproteins (HDL). HDL has several beneficial effects on cardiovascular disease risk and has been reported in epidemiological studies to also __________________ 2015 Transactions of the American Society for Neurochemistry ®
protect against Alzheimer Disease (AD). AD is the most common form of dementia in the elderly population, currently affecting over half of persons over 85 years of age. ABCA1 is expressed in all cells of the body. In the central nervous system (CNS), the major lipid acceptor for ABCA1 activity is apoE, which is the major lipidcarrying protein expressed in the brain and the best established genetic risk factor for AD. Both genetic and pharmacological evidence in animal models supports a beneficial role for high ABCA1 activity on minimizing amyloid deposition and cognitive dysfunction. However, it is not yet fully understood how lipidated apoE and apoA-I may each contribute towards an overall reduced AD risk. Potential mechanisms by which ABCA1 may be an attractive therapeutic target for AD will be discussed.
S24-03 APOLIPOPROTEIN E LIPIDATION AS A MECHANISTIC THERAPEUTIC TARGET FOR LOWERING SOLUBLE AMYLOID-! LEVELS IN ALZHEIMER’S DISEASE Mary Jo LaDu University of Illinois at Chicago, Anatomy and Cell Biology, Chicago, USA APOE4 is the greatest genetic risk factor for sporadic Alzheimer’s disease (AD), increasing risk up to 15-fold compared to APOE3, an effect significantly greater in APOE4 females. However, no therapeutics target mechanistic pathways underlying this risk. The amyloid-! peptide (A!42) is considered a major cause of impaired synaptic function in AD, particularly soluble oligomeric A! (oA!). Enabled by the development of novel ELISAs and the new EFAD transgenic (Tg) mouse model (overexpress A!42 with human APOE expression), demonstrate that compared to apoE3, lipoprotein-association/lipidation of apoE4 is reduced, resulting in lower levels of apoE4/A! complex and higher levels of soluble oA!, compromising synaptic viability. Further, with increasing A! pathology, this pathway is also relevant for apoE3. As ATP-bindingcassette-transporter-A1 (ABCA1) is a major transporter of lipid to apoE-containing lipoproteins in the CNS, a promising therapeutic approach for AD is to increase ABCA1 expression/activity. Our hypothesis is: !ABCA1"!apoE4 lipidation"!apoE4/ A!""soluble A!"!synaptic viability"!memory/cognition. Nuclear receptor agonists including bexarotene (RXR agonist) are one approach proposed for increasing ABCA1 expression. However, several issues compromise this approach, particularly questions regarding the safety and mechanism of action i.e. Do RXR agonists increase apoE levels or ABCA1-dependent lipidation of apoE? The latter issue is crucial as debate continues on whether APOE4 imparts a toxic gain-of-function, and thus RXR-agonist induced increased apoE4 levels may exacerbate A! pathology, or if the APOE4 loss-of-function is lipidation of apoE4, then induction of ABCA1 may be beneficial. In EFAD-Tg mice with high A!-pathology, 1-week treatment with RXR agonists (bexarotene and LG268) increased ABCA1 levels, apoE lipidation but not apoE levels, increased apoE/A!, reduced soluble A! (A!42 and oA!) and __________________ 52
S24 Modulaton of ABC Transporters and APOE Levels as Therapeutic Targets for Alzheimers increased synaptic protein expression. These results validate our hypothesis and demonstrate that RXR agonists address a loss-offunction associated with APOE4 and exacerbated by A! pathology, specifically low levels of apoE4 lipidation, thus providing target validation for ABCA1. However, significant side effects of hepatomegaly and steatosis were observed, requiring the development of novel, safer approaches to targeting ABAC1 for the prevention and treatment of AD, particularly for the fragile elderly population.
S24-04 ABCA1 DIRECTED TREATMENT OF APOE4 DRIVEN NEURODEGENERATION IN ALZHEIMER’S DISEASE Daniel Michaelson, Anat Boehm-Cagan, Ishai Luz, Ori Liraz Tel Aviv University, Neurobiology, Tel Aviv, Israel Background. The pathological effects of apoE4, the most prevalent genetic risk factor of Alzheimer's disease (AD), can be due to loss of a structural feature which the " good" apoE3 allele possesses and/or to gain of structural feature specific to apoE4. We will presently examine the degree to which such mechanisms mediate the effects of apoE4. The contribution of gain of structural effects of apoE4 will be assessed by investigating the extent to which the pathological effects of apoE4 can be counteracted by i.p. injection __________________
2015 Transactions of the American Society for Neurochemistry ®
of anti-apoE4 mAbs. The possible role of loss of structure related mechanisms will be assessed by measuring the extent to which the impaired lipidation of apoE4 and the associated pathological effects can be counteracted either by increasing the expression of the ABCA1 lipidation protein utilizing the RXR-agonist bexarotene or by application of ABCA1 agonistic peptides. Results. We have recently shown utilizing apoE3- and apoE4targeted replacement mice that apoE4 triggers cognitive impairments. This is associated with the accumulation of A!42 and of hyperphosphorylated tau in hippocampal neurons and with down regulation the levels of the presynaptic glutamate transporter VgluT1 and of the apoE receptor apoER2. Both i.p. injections of anti-apoE4 mAb and oral application of bexarotene reversed the cognitive impairments of the apoE4 mice and abolished their tau hyperphosphorylation. However, bexarotene, but not the anti-apoE4 mAb, counteracted the effects of apoE4 on A!42 and VgluT1 whereas the anti-apoE4 mAbs, but not bexarotene, counteracted the effects of apoE4 on the apoE receptor apoER2. Further experiments revealed that the brain pathological effects of apoE4 are also counteracted by i.p injections of the ABCA1 agonistic peptide CS6253. This peptide was kindly provided by Artery Ltd. Conclusion. Taken together the findings show that the pathological effects of apoE4 are mediated by both gain and loss of structural features which can be counteracted by anti-apoE4 mAbs and by reversal of the lipidation impairments of apoE4 and suggest that apoE4 treatment in AD should address both mechanisms .
53
S25 Neurodegeneration and Oxidative Stress S25-01
S25-03
SYNAPSE LOSS: AN INITIAL EVENT IN THE PROGRESSION AND PATHOGENESIS OF ALZHEIMERS DISEASE Rukhsana Sultana UT southwestern Medical Centre, Developmental Biology, Dallas, USA
DIETARY SUPPLEMENTATION OF FRUITS ALLEVIATE OXIDATIVE STRESS AND INFLAMMATION IN ALZHEIMER'S DISEASE TRANSGENIC MICE MODEL Mohamed-Essa Musthafa 1, 2, Selvaraju Subash 1, 2 Akbar Mohammed 3, Samir Al-Adawi 2, 4 1 Sultan Qaboos University, Oman, Ageing and Dementia Research group, Al-Khoud, Oman 2 Sultan Qaboos University, Oman, Nutrition, Al-Khoud, Oman 3 NIAA, NIH, Rockville, USA 4 Sultan Qaboos University, Oman, Behavioral Medicine, Al-Khoud, Oman
Alzheimer's disease (AD) is one of neurodegenerative disorders usually occurring after the age of 60 years. It is associated with a gradual loss of cognition and memory. Despite tremendous progress in AD research over the years, we still know very little about the early stages of the disease and the molecular events that lead up to it, although it is thought that synaptic failure is a central feature. Indeed, synapse loss occurs early in the progression of AD, even before the appearance of Amyloid-beta (Abeta) rich deposits within senile plaques (SP) and neurofibrillary tangles (NFTs, rich in hyperphosphorylated tau), suggesting that we should focus our effort on the key molecular players that normally maintain synaptic health and that may go haywire in the initial stages of AD. In my talk I will be focusing on the role of oxidative stress and some key synaptic proteins in the progression and pathogenesis of AD.
S25-02 AN OVERVIEW ON THE MECHANISMS OF MITOCHONDRIAL DYSFUNCTION AND TISSUE INJURY IN NEURODEGENERATIVE DISEASES Mohammed Akbar, Mohamed A. Abdelmegeed, Byoung-Joon Song NIH, Section of Molecualr Pharmacology and Toxicology, Rockville, USA Mitochondria provide the majority of cellular energy ATP. They are also critically important in anti-oxidant defense, fat oxidation, intermediary metabolism and cell death processes. It is wellestablished that mitochondrial functions are suppressed through nitroxidative stress when living cells or organisms are exposed to potentially toxic agents including alcohol, high fat diets, smoking and certain drugs or in many pathophysiological states. Under elevated nitroxidative stress, cellular macromolecules proteins, DNA, and lipids can undergo different covalent modifications, usually leading to disruption of their normal, sometimes critical, physiological functions. Recent reports also indicated that the cellular functions of many mitochondrial proteins are suppressed through various post-translation modifications (PTMs) contributing to mitochondrial dysfunction and cell death, as observed in distinct neurodegenerative diseases including Alzheimer’s Disease, Parkinson’s Disease, Huntington’s Disease, alcoholic dementia, and brain Ischemia-reperfusion (stroke) related injury. Numerous natural and synthetic antioxidants, including naturally occurring flavonoids and polyphenols, that can prevent or delay the formation of ROS/RNS to minimum levels and counteract the deleterious effects of mitochondria-related neurodegenerative diseases. Therefore, we will describe the mechanisms of various PTMs, dietary food supplements and their potential translational research opportunities. 2015 Transactions of the American Society for Neurochemistry ®
Alzheimer's disease (AD) is a devastating age-related neurodegenerative disease with no specific treatment at present. The APPsw/Tg2576 mice exhibit age-related deterioration in memory and learning as well as A! accumulation, and it is considered an effective model for studying brain aging in accelerated senescence. The present study was designed to investigate the dietary supplements and the beneficial effects of pomegranate, figs and the dates on oxidative markers and inflammatory cytokines in APPsw/ Tg2576 mice. Changes in the plasma cytokines and A!, ATP, and redox, inflammatory cytokines investigated in the brain of transgenic mice. Significant increased oxidative stress markers and inflammatory cytokines (IL-1!, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL10, TNF-" and Eotaxin activity) were found AD transgenic mice fed with normal diet. These oxidative and inflammatory markers were significantly decreased by 15 months dietary supplementation of pomegranate, fig and dates (4%) in diets. In addition, putative delays in the formation of senile plaques, as indicated by a decreasing tendency of A! levels in brain A! (1-40, 1-42) content. Thus, a novel pharmacological effect of these dietary supplements mediated by reducing oxidative stress and inflammatory cytokines during aging may be one mechanism underlying the beneficial effects of these fruits in neurodegenerative diseases such as AD, Parkinson's, Huntington's disease HD and other neurological disorders. The study was supported by a research grant from the Research Council, Oman (RC/AGR/FOOD/11/01).
S25-04 EARLY ACTIVATION OF STAT3 REGULATES REACTIVE ASTROGLIOSIS INDUCED BY DIVERSE FORMS OF NEUROTOXICITY James O'Callaghan 1, Kimberly Kelly 1, Reyna VanGilder 2 Michael Sofroniew 3, Diane Miller 1 1 Centers for Disease Control and Prevention, Health Effects Laboratory Division, Morgantown, USA 2 West Virginia University, School of Pharmacy, Morgantown, USA 3 University of California Los Angeles, David Geffen School of Medicine, Los Angeles, USA Astrogliosis, a cellular response characterized by astrocytic hypertrophy and accumulation of GFAP, is a hallmark of all types of CNS. Previously, we used the In a 1-methyl-4-phenyl-1,2,3,6tetrahydropyridine (MPTP) model of dopaminergic neurotoxicity to __________________ 54
S25 Neurodegeneration and Oxidative Stress show the development of neuroinflammation (upregulation of proinflammatory cytokines/chemokines) and activation of the janus kinase and signal transducer and activator of transcription (JAK2STAT3) pathway preceding the up-regulation of GFAP in astrocytes in the brain.Using neurotoxicants to instigate astrogliosis is advantageous because different brain regions and often, different cell types within a given region, can be selectively targeted. Here, we show that multiple mechanistically distinct models of neurotoxicity (MPTP, amphetamine (AMP), methamphetamine (METH), 3,4-methylenedioxyamphetamine (MDA), 3,4-methylenedioxymethamphetamine (MDMA), kainic acid (KA) and trimethyltin (TMT)) engender the same neuroinflammatory response and STAT3 activation in target brain regions of the respective neurotoxicants. Pharmacological antagonism of neurotoxic effects of MPTP, METH and KA with nomifensine, ethanol and diazepam, respectively, blocked the neuroinflammation, phosphorylation of STAT3 and GFAP induction, indicating neuronal damage as a component of the initiation of astrogliosis. Deletion of astrocytic STAT3 in conditional knockout mice prevented the induction of GFAP in MPTP-treated mice. Double immunostaining of GFAP and STAT3 showed enhanced nuclear staining localized to astrocytes in association with the induction of astrogliosis. These findings strongly implicate the STAT3 pathway in astrocytes as a key signaling pathway for astrogliosis.
2015 Transactions of the American Society for Neurochemistry ®
S25-05 INDUCTION OF NEURONAL DEATH BY MICROGLIAL AGE-ALBUMIN: COMMON IMPLICATIONS FOR NEURODEGENERATIVE DISEASES Bonghee Lee Gachon University, LCDI, Incheon, Korea South Advanced glycation end products (AGEs) have long been considered as potent molecules promoting neuronal cell death and contributing to many neurodegenerative disorders. In this study, we demonstrate that AGE-albumin, the most abundant AGE product in human AD, PD, stroke and alcoholic brains, is synthesized in activated microglial cells and secreted into the extracellular space. The rate of AGE-albumin synthesis in human microglial cells is markedly increased by relevant cytokine exposures and oxidative stress. Exogenous AGE-albumin upregulates the receptor protein for AGE (RAGE) and augments calcium influx, leading to apoptosis of human primary neurons. In animal experiments, soluble RAGE (sRAGE) decreased the RAGE levels and thus protected neuronal deaths in rat-mimicking human ND brains. Collectively, these results provide evidence for a new mechanism by which microglial cells promote death of neuronal cells through synthesis and secretion of AGE-albumin, thereby likely contributing to neurodegenerative diseases.
55
S26 CDK5 Signaling: A Double-edged Sword in Normal and Diseased Brain S26-01 IMPORTANT ROLE OF CDK5 IN PAIN MODULATION Ashok Kulkarni, Bradford Hall, Anita Terse, Michaela Prochazkova National Institutes of Health (NIH), Functional Genomics Section, LCDB, NIDCR, Bethesda, USA Cyclin-dependent kinase 5 (Cdk5), a predominantly neuronal serine/tyrosine kinase, is important for brain development and function. We have previously discovered that Cdk5 modulates peripheral pain signaling and that it is required for basal responses to noxious heat. In response to peripheral thermal stimulation, the mice with significantly reduced Cdk5 activity showed hypoalgesia, whereas the mice with significantly increased Cdk5 activity displayed hyperalgesia. Furthermore, the mice deficient in Cdk5 activity only in sensory neurons developed hypoalgesia associated with reduced phosphorylation of the TRPV1 channel. Our current research efforts are focused on characterizing the precise role of Cdk5 in orofacial pain using different behavioral methods and genetically altered Cdk5 mice. We adapted an existing orofacial stimulation test to assess the behavioral responses to mechanical stimulation in the trigeminal region of mice. Using this test we have discovered that Cdk5 activity modulates orofacial mechanical nociception. Along with these studies we have initiated studies to analyze the role of Cdk5 in orofacial thermal sensation. Based on our previous findings showing that Cdk5 phosphorylates TRPV1 during painful thermal stimulation in the spinal system, we are using an oral aversion test to examine the responses of our Cdk5 mouse models to the TRPV1 agonist capsaicin. Our current findings indicate aversive behavior and hypersensitivity to capsaicin in the mice with increased Cdk5 activity as evident from the decreasing number of licks with increasing concentrations of capsaicin. In contrast, mice with reduced Cdk5 activity showed less aversion to capsaicin consumption. Thus, our findings indicate that Cdk5 modulates orofacial mechanical and thermal pain. This research was supported by the Intramural Research Program of the NIH, NIDCR.
S26-02 INTEGRATION OF EXCITATORY AND METABOTROPIC SIGNAL TRANSDUCTION James Bibb, Florian Plattner, Karine Pozo, Tara Tassin Rachel Thomas, Chunfeng Tan, Gabriel Mettlach UT Southwestern Medical Center, Psychiatry Dept., Dallas, USA Environmental experience triggers excitatory neurotransmission, the salience of which is assigned by metabotropic neurotransmission within the limbic system. As a result, meaningful experiences, trigger memory formation through synaptic reorganization and circuitry formation. At the mechanistic level, this requires the integration of different signaling pathways. As a constitutive protein kinase, Cdk5 maintains basal tonus for signaling pathways through its phosphorylation of many substrates. For example Cdk5 modulates cognition and synaptic remodeling through its phosphorylation of Ser1116 of the NR2B receptor, __________________ 2015 Transactions of the American Society for Neurochemistry ®
which maintains its intracellular compartmentalization. Upon excitatory neurotransmission, this site is dephosphorylated, resulting in transient migration of NR2B to the synaptic cell surface where it may mediate further synaptic reorganization such as that which occurs after an experience and results in new memory circuit formation. Targeting this pathway with a small interfering peptide results in increased NR2B surface expression and function, and enhanced memory formation. Cdk5 also maintains basal levels of metabotropic signaling. For example, Cdk5-dependent phosphorylation of members of the PDE4 phosphodiesterase family facilitates its cAMP/PKA-dependent activation. Targeting this pathway attenuates cAMP metabolism, raising basal levels of this second messenger and the activity of PKA. The circuitry in which this pathway mediates emotional salience may be pinpointed to D1 dopamine receptor-positive neurons of the ventral striatum. Selectively deleting Cdk5 or blocking this pathway in these neurons results in antidepressant-like behavior in mice. These pathways, as well as others, by which Cdk5 integrates excitatory and metabotropic neurotransmission will be presented and discussed. By better understanding and targeting these signaling mechanisms, we may develop effective new treatments for neuropsychiatric and neurological disorders.
S26-03 CONTROLLING CDK5 FUNCTION IN NEURONAL AND GLIAL LINEAGES Yue Feng, Andrew Bankston, Megan Allen, Wenqi Li Emory University, Pharmacology, Atlanta, USA Cyclin-dependent kinase 5 (Cdk5) governs brain development and function by phosphorylation of serine and threonine residues on diverse protein targets. Dysregulation of Cdk5 is reported in a growing list of brain disorders, including Alzheimer’s disease, schizophrenia, stroke, and epilepsy. However, molecular mechanisms that control Cdk5 pathway function still remain elusive. Activation of Cdk5 depends on the available amount of p35 or p39, two distinct non-cyclin activators. Rich literature has demonstrated the critical function of p35 in neuronal Cdk5 activity and function. Conversely, the functional importance of abundant p39 expression specifically in the postnatal brain is vastly unknown. Emerging evidence has revealed that besides brain neurons, Cdk5 activity is crucial for the development and function of oligodendrocytes (OLs) that provide myelination and trophic support to brain neurons. The loss of Cdk5 impairs myelinogenesis and myelin lesion repair. We found that in contrast to neurons that harbor high levels of both Cdk5 activators, oligodendrocytes employ p39 to activate Cdk5, which is essential for OL differentiation and myelin repair. Furthermore, we discovered that p39, but not p35, is selectively up-regulated more than ten-fold during neuronal differentiation, thereforeunderlying the developmentally programmed increase of Cdk5 activity and phosphorylation of specific Cdk5 target proteins that promote neuronal circuitry establishment. Interestingly, histoneacetylation plays opposing roles to drive selective up-regulation of __________________ 56
S26 CDK5 Signaling: A Double-edged Sword in Normal and Diseased Brain p39 in the OL and neuronal lineage. Finally, we show that p39 is essential for synaptic development and seizure response. Together, these findings demonstrate distinct mechanisms that control Cdk5 function in neuronal and glia lineages through differential regulation of Cdk5 activators, offering possibilities for differential manipulation of Cdk5 function in various brain disorders where Cdk5 function is differentially affected in OLs and neurons.
S26-04 PHYSIOLOGICAL AND PATHOLOGICAL CONDITIONS ALTER THE ROLE OF PHOSPHORYLATED JIP1 BY CDK5 Dianbo Qu, Doo-Soo Im, David Park University of Ottawa Brain and Mind Research Institute, Faculty of Medicine, Ottawa, Canada
logical conditions. For example, its physiological functions are required for proper development of brain, learning and memory, synaptic function, cytoskeleton dynamic, and membrane transport. In contrast, Cdk5 also display pathological functions, inhibiting prosurvival pathways, such as MEF2D, Prx2 and APE1 signals. Recently, we identified JIP1 (JNK interacting protein 1) as a novel substrate of Cdk5. Our preliminary data showed that phosphorylation of JIP1 by Cdk5 may activate JNK signaling pathway to regulate neuronal death under stress conditions. Of note, phosphorylation of JIP1 by Cdk5 also regulates neuronal migration during brain development. Taken together, our preliminary data identify a novel target of Cdk5 and that this effector may regulate both developmental or pathological consequences.
Cdk5, a member of Cyclin dependent kinase family, plays critical roles in central nervous system under physiological and patho__________________
2015 Transactions of the American Society for Neurochemistry ®
57
S27 Sex-Specific Effects of Adolescent Drugs and Stressors on Neural, Metabolic, Immune and Behavioral Outcomes S27-01 SEX DIFFERENCES IN THE AMYGDALA AS A RISK FACTOR FOR MOOD DISORDERS Bradley Cooke Georgia State University, Neuroscience, Atlanta, USA Women are twice as likely as men to develop a mood disorder such as anxiety and depression. Although gonadal hormones play a major role in the etiology of this sex difference, adverse early experience has a greater impact on the risk for mood disorders in girls than boys. We hypothesize that this sex difference is related to how juveniles perceive and remember stressors, which most often arise from the social world. Thus, my lab is focused on the intersection of early social stress, sex differences in the prepubertal brain, and the effects of puberty on the stress response system. In this talk, I will review how prepubertal sex differences in synapses, dendrites, axons and glia, as well as its large-scale connectivity, make the medial nucleus of the amygdala a potentially crucial substrate of sex differences in the perception and memory of social stressors. I will then describe how social stress elicits sex-specific patterns of neural activity in the medial amygdala of juvenile rats and leads to more severe mood disorder-like behaviors in adult females than in males. Finally, I will discuss how gonadal hormones at puberty alter the expression of corticotropin releasing factor receptors, synaptic connectivity, and the gross morphology of the medial amygdala in such a way as to further differentiate male from female and amplify this brain region’s contribution to the sex-specific perception and memory of stressors. The talk will conclude with a brief description of human research consistent with our results and a consideration of future directions.
S27-02 INCUBATION AND REVERSAL OF ADOLESCENT COCAINE-INDUCED HABITS Shannon Gourley Emory University, Pediatrics, Atlanta, USA Adolescence is a period of vulnerability to the development of many psychiatric disorders, including substance dependence disorders that can persist across the lifespan. Incubation of certain biological factors associated with addiction may play a causal role. We have discovered that mice exposed to subchronic cocaine in adolescence, but not adulthood, are behaviorally insensitive to changes in action-outcome contingencies, generating instead reward-seeking habits. This deficiency incubates during periadolescence, and in tandem, dendritic spines in deep-layer orbitofrontal cortex (oPFC) are eliminated. Meanwhile, remaining spines retain an immature, adolescent-like morphology. oPFCtargeted infusion of STI-571, an Abl-family kinase inhibitor that destabilizes neural structure, also biases responding towards stimulus-response habits, recapitulating the effects of adolescent __________________ 2015 Transactions of the American Society for Neurochemistry ®
cocaine. Conversely, pharmacological inhibition of Rho-kinase (ROCKII) or NR2B-containing NMDA receptors blocks cocaineinduced habits by enhancing new response-outcome associative conditioning. Together, these findings suggest that adolescent cocaine exposure confers behavioral vulnerabilities in adulthood by altering cellular structure. Novel treatment strategies could aim to reverse the chronic, incubated effects of adolescent cocaine exposure, including behavioral, morphological, and neuroplastic consequences. Further, given new evidence from our lab that individual differences in cocaine self-administration patterns during adolescence sex-dependently predict the propensity to engage in habits, such treatment strategies could also be informed by sex.
S27-03 STRESS AND PUBERTY: THE PERFECT STORM OR A CHANGE OF COURSE? Gretchen Neigh 1, 2, Constance Harrell 1, Jilly Beth Burgado 1 Anisha Kalidindi 1, Sean Kelly 1 1 Emory University, Physiology, Atlanta, USA 2 Emory University, Psychiatry & Behavioral Sciences, Atlanta, USA Adolescence is an additional critical period during development in which the brain is particularly influenced by the environment. The plasticity of the brain during adolescence is evidenced by the profound effects of chronic stress during puberty. The mechanisms that mediate adolescent susceptibility to stress exposure are at least partially driven by sex hormones which are undergoing dramatic changes during the period of adolescence. However, it is not abundantly apparent whether the effects of chronic adolescent stress are simply adaptations that garner opportunity costs that have adverse physiological effects, or if the changes observed are truly pathological effects spurned by stress exposure during a sensitive developmental period. The data discussed in this presentation will use sex differences in the effects of adolescent stress exposure to explore the extent to which consequences of chronic adolescent stress are adaptive and/or pathological.
S27-04 ADOLESCENT (IN)VULNERABILITY TO ENDURING EFFECTS OF DRUGS OF ABUSE Kyle Frantz Georgia State University, Neuroscience Institute, Atlanta, USA Adolescent drug abuse is hypothesized to increase the risk of drug addiction. Yet male rats that self-administer drugs of abuse as adolescents show attenuated drug-seeking after abstinence, com__________________ 58
S27 Sex-Specific Effects of Adolescent Drugs and Stressors on Neural, Metabolic, Immune and Behavioral Outcomes pared with adults, in our laboratory. Among other explanatory hypotheses, we have explored a role for neural activity and plasticity-related gene expression in the medial prefrontal cortex (mPFC) and nucleus accumbens (NAcc) in age-dependent drugseeking. Adolescent (~35-day-old at start; adolescent-onset) and adult (~86-day-old at start) male rats acquired lever-pressing maintained by cocaine, morphine, or heroin. Following variable periods of abstinence and/or extinction, rats were tested for reinstatement of drug-seeking triggered by context, cues, or drug-priming. In the case of heroin-seeking, unbiased stereology was then used to estimate the number of Fos-ir(+) and Fos-ir(-) neurons in prelimbic and infralimbic mPFC, core and shell of NAcc. In the case of cocaine-seeking, in situ hybridization was used to explore brain derived neurotrophic factor (BDNF) and activity-regulated cytoskeletal protein (arc) expression in reinforcement-related brain regions. Generally, drug-seeking was attenuated in the younger rats, regardless of initial levels of drug intake during self-administration. __________________
2015 Transactions of the American Society for Neurochemistry ®
The adolescent-onset groups also failed to show significant neural activation in the mPFC during the heroin-seeking test, whereas the adult-onset heroin self-administration group showed two to six times more Fos-ir(+) neurons than their saline counterparts. Moreover, the estimated number of neurons in the infralimbic cortex was greater in rats from the adolescent-onset groups than adults. Finally BDNF mRNA expression was higher among adolescent compared with adult rats. The mPFC may thus play a key role in some age-dependent effects of heroin self-administration. Although no effects of heroin on neuronal activation were observed in the NAcc and gene expression was generally lower in the NAcc than cortical regions, some evidence suggests that heroin experience may differentially affect soma size in core and shell. Such varied neuronal effects of drug intake may contribute to complex patterns of both vulnerability and resistance to some enduring effects of drug exposure during adolescent development.
59
S28 Contribution of Environmental Chemicals to Neurodegenerative Disease S28-01 SLC30A10 IS A CELL SURFACE-LOCALIZED MANGANESE EFFLUX TRANSPORTER Michael Aschner 3, Dinorah Leyva-Illades 1, Pan Chen 3 Charles Zogzas 1, Steven Hutchens 1, Jonathan Mercado 1 Caleb Swaim 1, Richard Morrisett 1, Aaron Bowman 2 Somshuvra Mukhopadhyay 1 1 University of Texas, Division of Pharmacology and Toxicology, Austin, USA 2 Vanderbilt University Medical Center, Department of Neurology, Nashville, USA 3 Albert Einstein College of Medicine, Department of Molecular Pharmacology, Bronx, NY Manganese (Mn) is an essential metal, but elevated cellular levels are toxic and may lead to the development of an irreversible parkinsonian-like syndrome that has no treatment. Mn-induced parkinsonism generally occurs as a result of exposure to elevated Mn levels in occupational or environmental settings. Additionally, patients with compromised liver function attributable to diseases, such as cirrhosis, fail to excrete Mn and may develop Mn-induced parkinsonism in the absence of exposure to elevated Mn. Recently, a new form of familial parkinsonism was reported to occur as a result of mutations in SLC30A10. The cellular function of SLC30A10 and the mechanisms by which mutations in this protein cause parkinsonism are unclear. Here, using a combination of mechanistic and functional studies in cell culture, Caenorhabditis elegans, and primary midbrain neurons, we show that SLC30A10 is a cell surface-localized Mn efflux transporter that reduces cellular Mn levels and protects against Mn-induced toxicity. Importantly, mutations in SLC30A10 that cause familial parkinsonism blocked the ability of the transporter to traffic to the cell surface and to mediate Mn efflux. Although expression of disease-causing SLC30A10 mutations were not deleterious by themselves, neurons and worms expressing these mutants exhibited enhanced sensitivity to Mn toxicity. Our results provide novel insights into the mechanisms involved in the onset of a familial form of parkinsonism and highlight the possibility of using enhanced Mn efflux as a therapeutic strategy for the potential management of Mn-induced parkinsonism, including that occurring as a result of mutations in SLC30A10.
amino-L-alanine (BMAA), is produced by cyanobacteria and that BMAA is abundant in both the traditional diet and brain tissues of ALS/PDC patients reinvigorated interest in this environmental toxin. However, the absence of an unequivocal animal model demonstrating BMAA-induced progressive neurodegenerative disease has slowed broader acceptance of the BMAA/cyanobacterial theory. BMAA is toxic to specific subpopulations of motor neurons through excitotoxic activation of AMPA/kainate receptors. BMAA is also an NMDA agonist. Recent studies have highlighted BMAA toxicity to glial cells. BMAA also activates the mRGLU5 receptor, inducing oxidative stress. In the presence of bicarbonate BMAA forms neurotoxic carbamates, and the isomers of BMAA have also been found to be neurotoxic. Perhaps more importantly, BMAA can be misincorporated for L-serine during protein synthesis, leading to misfolding and protein aggregation. Increased concentrations of Lserine inhibit such misincorporation. FDA-approved human clinial trials to determine if L-serine can slow the progession of ALS are now underway. Protein changes in neonatal mice have been found after BMAA exposures, linked to cognitive deficits in adults. Rats exposed to BMAA have some neuropathologies similar to ALS. However, previous primate experiments, while showing acute neurotoxicity at high doses, did not demonstrate BMAA-induced progressive neurodegeneration. In the United States, France, and the Gulf area, individuals with significant cyanobacterial exposures through contaminated water, shellfish, and desert dust appear to have a higher risk of ALS. Broader acceptance of the BMAA/cyanobacteria theory will likely require development of an animal model, probably in non-human primates, demonstrating that chronic exposure to BMAA results in tauopathies, amyloid plaques, Bunina bodies, and other protein inclusions similar to the neuropathological abnormalities observed in brain tissues from Chamorro ALS/PDC patients in Guam. Increased epidemilogical studies linking cyanobacterial exposures to neurodegenerative disease are also needed to demonstrate broader links between BMAA and human disease.
S28-03 S28-02 THE BMAA/CYANOBACTERIAL THEORY AND ALS/PDC IN GUAM Paul Cox Institute for Ethnomedicine, Ethnobotany, Jackson Hole, USA The mystery of ALS/PDC among the Chamorro people of Guam has fascinated researchers for decdes. Early investigators suggested that environmental exposures may trigger this disease syndrome which combines neuropathological and clinical aspects of amyotrophic lateral sclerosis, Alzheimer's and Parkinson's disease. The twin discoveries that a non-protein amino acid, beta-methyl__________________ 2015 Transactions of the American Society for Neurochemistry ®
MECHANISMS OF GENE-ENVIRONMENT INTERACTIONS IN ALZHEIMER DISEASE Jason Richardson Rutgers Robert Wood Johnson Medical School, Environmental and Occupational Medicine, Piscataway, USA Alzheimer disease (AD) is the most common neurodegenerative disease worldwide and is expected to increase 3-fold over the next 40 years. To date, a massive amount of effort has focused on identifying genetic contributors to AD. Although there is a growing list of susceptibility genes that collectively contribute to AD, the the largest GWAS study published on AD ( > 74,000 individuals) identified only 1 out of 19 loci as an individual strong contributor to AD. Thus, The interactions between senescent-related, genetic, and __________________ 60
S28 Contribution of Environmental Chemicals to Neurodegenerative Disease environmental factors likely contributes to the etiology of late-onset sporadic AD. We recently reported that serum levels of a longlasting residue of the organochlorine pesticide DDT (p,p’-DDE) were significantly higher in patients with AD. Further, those carrying an APOE4 allele and having high serum levels of DDE performed significantly worse on the mini-mental state exam, suggesting the potential for gene-environment interactions. Mechanistically, DDT/DDE appears to increase expression of amyloid precursor protein and secretion of A! in cells and animals. To address the role of gene-environment interactions, APOE 3/3 or 4/4 expressing N2A cells and human neural stem cells expressing these polymorphisms are currently being used to determine the mechanistic basis of these interactions. This presentation will provide a summary of our findings to date and propose future strategies for studying gene-environment interactions in AD. Supported in part by P30ES005022
S28-04 THE STRIATAL SYNAPSE AS A TARGET FOR DAMAGE BY FLAME RETARDANT COMPOUNDS William Caudle 1, 2, Joshua Bradner 1, Kelly Genskow 1, 2 1 Emory University, Environmental Health, Atlanta, USA 2 Emory University, Center for Neurodegenerative Disease, Atlanta, USA
iture, electronics, and baby products. These chemicals readily migrate from these products, resulting in significant exposure of the human population to these chemicals. Indeed, elevated concentrations of these compounds have been found in human tissue, especially the brain. Increasingly, epidemiological and laboratory research has identified exposure to flame retardant compounds as a major contributor to several neurological and neurobehavioral deficits, including learning and memory and autism spectrum disorder. Recent work from our group has delineated the nigrostriatal dopamine system as a critical target for damage by flame retardant exposure. Alterations to this circuit appear to be focused on the dopaminergic synapse in the striatum, where synaptic proteins involved in mediating dopamine handling and neurotransmission are affected. These findings are important as damage to the nigrostriatal dopamine system underlie the major pathological and clinical symptoms of Parkinson disease. Furthermore, exposure to chemicals found in our environment is considered a major risk factor for the development and progression of Parkinson disease. Thus, our findings further contribute to the role of the environment in Parkinson disease etiopathogenesis and highlight specific synaptic proteins and processes that appear to be vulnerable to flame retardant exposure.
Flame retardant chemicals have routinely been used to reduce the flammability of a diverse array of consumer goods, such as furn__________________
2015 Transactions of the American Society for Neurochemistry ®
61
S29 Neuroinflammation and Immune Responses in Autoimmune Demyelination Diseases S29-01 ACTIVATION OF INNATE IMMUNITY BY BLOOD-CLOTTING FACTORS: MECHANISMS, IMAGING, THERAPEUTICS Katerina Akassoglou University of California, San Francisco, Gladstone Institute of Neurological Disease, San Francisco, USA Protection of the CNS from leakage of plasma proteins by the blood-brain barrier (BBB) is lifted in a wide range of neuroimmune and neurodegenerative diseases, as well as after traumatic injury. However, whether plasma proteins contribute to neuroinflammation and neuronal damage remains poorly understood. Our laboratory has discovered pleiotropic functions for the plasma protein fibrinogen in the activation of innate immunity in the CNS, induction of gliotic scar formation, and inhibition of remyelination. Such diverse functions have mechanistic underpinnings on the unique structure of fibrinogen, which contains multiple binding sites for cellular receptors and proteins expressed in the nervous system. Fibrinogen is a potent pro-inflammatory mediator in the nervous system by activating the CD11b/CD18 integrin receptor (also known as Mac-1 and complement receptor 3) in microglial cells. Using in vivo imaging in the mouse spinal cord using twophoton microscopy, we showed that in Experimental Autoimmune Encephalomyelitis (EAE), a model of multiple sclerosis (MS), microglia rapidly perform constant surveillance of blood vessel walls and specifically cluster around blood vessels with fibrin deposition. Pharmacologic or genetic disruption of the fibrinogen/ CD11b interaction suppresses microglial cluster formation, neurologic symptoms, inflammation, demyelination, and axonal damage in EAE. These studies identified fibrinogen as a novel molecular link between BBB disruption and activation of CNS innate immunity. Fibrinogen has the potential for selective drug targeting to suppress its damaging functions in the nervous system without affecting its beneficial effects in hemostasis. Strategies for fibrinselective inhibition of innate immunity will be discussed.
S29-02 REGULATION OF CNS INFLAMMATION BY A NORMAL LIPID-METABOLIZING ENZYME Kareem Graham 1, Bonnie Werner 1, Kimberly Moyer 1 Maria Zoudilova 2, Raymond Sobel 2, Eugene Butcher 2 1 Emory University, Physiology, Atlanta, USA 2 Stanford University, Pathology, Stanford, USA CD4+ T lymphocytes have key roles in the pathology of multiple sclerosis (MS), but the mechanisms that govern CD4+ T cell effector functions within the central nervous system (CNS) are poorly understood. To define novel regulators of pathogenic CNS inflammation, we performed transcriptional profiling of memory phenotype CD4+ T cells isolated from CNS and lymphoid tissues of mice with experimental autoimmune encephalomyelitis (EAE), a __________________
2015 Transactions of the American Society for Neurochemistry ®
widely studied animal model of MS. We found that mRNA for diacylglycerol O-acyltransferase-1 (Dgat1), an enzyme involved in triglyceride formation, was highly upregulated by CNS-infiltrating memory CD4+ T cells from mice with EAE. Administration of a selective DGAT1 inhibitor attenuated EAE, and DGAT1-deficient mice developed less severe clinical EAE than wild type (WT) controls. Compared to their WT counterparts, DGAT1 knockout mice with EAE had fewer mononuclear cell infiltrates within the CNS; and CNS and lymphoid tissues from DGAT1-deficient mice contained a higher proportion of Foxp3+ T regulatory cells. In addition, DGAT1-deficient, in vitro-polarized encephalitogenic Th17 cells were unable to fully induce EAE by adoptive transfer. Our results demonstrate the ability of CNS-specific CD4+ T cell expression profiling to uncover factors of therapeutic relevance in EAE. Through genetic and pharmacologic approaches, we identify DGAT1 as a regulator of T cell-mediated autoimmune CNS inflammation. DGAT1 may therefore represent a novel target for therapy of MS.
S29-03 THE NOVEL FUNCTION OF RGS10 IN EFFECTOR T LYMPHOCYTES TO AUGMENT MOUSE EAE Jae Kyung Lee, Jaegwon Chung, George Kannarkat Kareem Graham, Malu Tansey Emory University, Physiology, Decatur, USA A broad range of immune-based therapeutic drugs have been available for treatment of various autoimmune diseases but many have had limited success. Therefore, it is critical to identify additional cellular targets that can regulate the pathogenic response of immune cells. We hypothesize that GPCR modulator RGS proteins are important modulators of the immune responses involved in the development of multiple sclerosis (MS). Our hypothesis is based on reports that various SNPs in RGS proteins is highly correlated with the diagnosis of MS. Especially, an in-depth search of the GEO profiles database also revealed higher levels of RGS10 transcripts in MS patients’ PBMCs. Here, we showed RGS10-null mice displayed significantly milder clinical symptoms of EAE with reduced incidence and delayed disease onset. We observed that there were less numbers of CD45+ cells and CD4+ T cells in MOG35-55immunized RGS10-null mice in the CNS. RGS10-null LN T cells were less proliferative and produce less IFN-gamma and IL-17 cytokines in response to MOG. Our data suggest a critical role for RGS10 in mediating disease by promoting lymphocyte infiltration or peripheral immune responses during EAE. This is the first study ever conducted to elucidate the function of RGS10 in effector lymphocytes in the context of EAE. The identification of RGS10 as a central regulator of the inflammatory processes will open a possibility for developing more specific targeted therapy for the treatment of MS.
62
S29 Neuroinflammation and Immune Responses in Autoimmune Demyelination Diseases
S29-04 FREQUENCY AND AFFINITY OF T CELLS IN THE CNS DETERMINES EXTENT OF NEUROINFLAMMATION Brian Evavold Emory University, Microbiology and Immunology, Atlanta, USA Disease outcome for experimental autoimmune encephalomyelitis (EAE) is dependent on the number and specificity of antigen reactive and regulatory T cells. We use a novel micropipette adhe__________________
2015 Transactions of the American Society for Neurochemistry ®
sion frequency assay to provide the most sensitive method for characterizing T cell frequency and affinity as disease progresses in the CNS. Our data reveals the importance of T cell two-dimensional (2D) affinity for myelin antigens as a factor in disease progression. In addition, we are able to characterize regulatory T cell specificity for myelin and determine their impact in overall disease outcome. Our results highlight the importance in determining the number and specificity of antigen reactive T cells in the CNS.
63
S30 Pharmacologically Induced Hypothermia and Clinical Implications S30-01 NEUROTENSIN RECEPTOR 1 AGONISTS INDUCED HYPOTHERMIA FOR THE TREATMENT OF STROKE AND TBI Shan Yu Emory University, Anesthesiology, Atlanta, USA Stroke and traumatic brain injury (TBI) are leading causes of human death and disability and there are few effective treatments for those patients. The failure of many clinical trials in recent years has generated the consensus that for a therapy to be effective against complicated CNS disorders such as cerebral ischemia and TBI, it requires overwhelming protective effects on multiple pathways and multiple cell types. To this end, therapeutic hypothermia stands out for its versatile protective effects on the brain. Mild-to-moderate hypothermia has shown remarkable neuroprotective effects (up to 90% infarct reduction) against brain ischemia in animal and human studies. Some of the drawbacks to current cooling techniques of physical means are that they are slow (!3 hrs), difficult to control and require general anesthesia to battle against shivering, which have hampered clinical applications of hypothermia therapy. Thus, chemical compounds that can be utilized for hypothermia therapy have long been sought after for clinical treatments. Using druginduced hypothermia, it is expected that even a small drop in body temperature (1-2°C) is beneficial for preventing the detrimental post-injury hyperthermia, delay the evolution of the secondary injury, and thereafter extend the therapeutic window for other interventions. We have developed novel neurotensin derivatives such as HPI201 (ABS201) and HPI363 that can pass through the bloodbrain barrier to induce “regulated hypothermia”, reducing body and brain temperature by 3-5°C in around 30 min without causing shivering. Post-ischemic administration of these compounds markedly attenuates ischemia-induced neuronal cell death, bloodbrain barrier damage, inflammation, and improved functional recovery. In a hemorrhagic stroke model of the mouse, HPI201 administration 24 hrs after the onset of stroke still showed significant neuroprotection and functional benefits. Our recent investigation also showed protective effects of drug-induced hypothermia against TBI. Systemic studies, blood tests, and autopsy examinations showed no toxic or adverse effects of these compounds. It is expected that drug-induced hypothermia can be developed as a new category of global brain protection drugs and help to translate the pharmacological hypothermic therapy into clinical applications.
S30-02 TRANSLATING DRUG-INDUCED HIBERNATION TO THERAPEUTIC HIBERNATION Kelly Drew, Tulasi Jinka University of Alaska Fairbanks, Institute of Arctic Biology, Fairbanks, USA Hibernation is a physiological state of energy conservation associated with a decrease in core body temperature (Tb) and oxygen __________________ 2015 Transactions of the American Society for Neurochemistry ®
consumption (VO2). Mild therapeutic hypothermia, where Tb is cooled to 32-34°C for 12, 24 or 72h, is becoming the standard of care for cardiac arrest and neonatal hypoxic ischemic encephalopathy and is in clinical trials for stroke. Cooling conscious stroke patients is complicated by energy demanding processes. Shivering and nonshivering thermogenesis may counter beneficial effects of cooling and be uncomfortable for the patient. Therapeutic hibernation refines temperature management by suppressing thermoregulatory and other energy demanding processes to decrease metabolic demand (VO2) that leads to a subsequent decrease in Tb. In recent work, we showed that the arctic ground squirrel (AGS) enters hibernation through activation of CNS adenosine A1 receptors (A1AR), in a manner consistent with the inhibition of thermogenesis. Torpor in hamsters and mice also depends on central A1AR activation. Here we tested the hypothesis that an A1AR agonist combined with lower Ta would provide a means to manage Tb below 37°C for therapeutic purposes. We stimulated A1AR with the A1AR agonist N6-cyclohexyladenosine (CHA) and targeted the CNS by co-administration of 8-(p-Sulfophenyl) theophylline (8SPT), a nonspecific adenosine receptor antagonist that does not cross blood brain barrier. Ambient temperature (Ta) was varied between 16-29°C. Drugs were administered to naïve rats and rats subjected to 8 minutes of asphyxial cardiac arrest (male SpragueDawley, 2 to 3 months old, 375-400g). Results show that CHA (1mg/kg, IP every 4h for 20h at Ta of 16°C) induces and maintains Tb between 29-31°C for 24h in both naïve rats and rats subjected to 8 minutes of asphyxial cardiac arrest. 8-SPT reversed bradycardia without affecting Tb. More stable hypothermia was achieved by continuous infusion of CHA delivered subcutaneously via minipumps. Animals subjected to cardiac arrest and cooled by CHA survived better and showed less neuronal cell death than normothermic control animals. In conclusion, central A1AR activation in combination with a thermal gradient shows promise as a novel and effective pharmacological adjunct for inducing safe and reversible therapeutic hibernation.
S30-03 DO CHILI PEPPERS PROVIDE THE SPICE OF LIFE? HOW TRPV1 AGONISM PROMOTES REVERSIBLE HYPOTHERMIA AND NEUROPROTECTION FOLLOWING STROKE Sean Marrelli Baylor College of Medicine, Anesthesiology, Houston, USA Hypothermia may offer a much needed additional therapeutic option for stroke patients. However, more effective and less physiologically stressful methods of producing hypothermia in the conscious stroke patient are needed. In the following study, we have explored the therapeutic effectiveness of TRPV1 channel agonism in promoting sustained mild hypothermia following ischemic stroke. TRPV1 channels are heat and capsaicin-activated ion channels prevalent in the thermoregulatory system. By pharmacologically activating these channels, we provide false temperature __________________ 64
S30 Pharmacologically Induced Hypothermia and Clinical Implications information to the hypothalamus, thus triggering a centrally-driven suppression of heat-producing mechanisms and a drop in core temperature. We employed a mouse model of transient distal middle cerebral artery occlusion (2 hours) followed by one month of reperfusion. At 90 minutes reperfusion, infusion of either vehicle or dihydrocapsaicin (DHC; TRPV1 agonist) was initiated by implanted osmotic pump. In DHC-treated mice, hypothermia (32-34 °C) was maintained for a period of 8 hours beginning 3.5 hours after stroke initiation. In a third group, a sham surgery was performed. All studies were performed in adult male C57BL6 mice. Following stroke (or sham surgery), mice were evaluated by performance testing through 28 days reperfusion and histology at 30 days. Performance testing demonstrated improved neurofunction in the DHC-treated mice in foot fault, swimming time, and turning latency. No differences were measured in RotaRod, beam walking, or grip strength tests. Total necrosis area in Nissl-stained brain sections showed a trend toward reduced area in DHC-treated mice, but was not statistically significant with the current sample size (n=5-7). However, DHC treatment did significantly reduce the volume of thalamic neuronal degeneration, a form of delayed secondary damage mediated via thalamic projections into the injured cortex. In summary, these studies demonstrate that TRPV1 agonism can produce effective sustained hypothermia in a conscious animal subject and provide long-term neuroprotection following ischemic stroke.
S30-04 MULTI-TARGET TREATMENT IN ACUTE ISCHEMIC STROKE Flemming Fryd Johansen University of Copenhagen, Biomedical Department, Copenhagen, Denmark Stroke is a major health problem worldwide causing death and disability. The only probate treatment is thrombolysis. Unfor__________________
2015 Transactions of the American Society for Neurochemistry ®
tunately, thrombolysis is just accessible to few patients due to a narrow 4! hours therapeutic window. Most new stroke treatments that reach clinical trials are mono-therapies although reviews unanimously picture that stroke in time and space progress sequentially through multiple pathomechanisms. Drug-induced hypothermia is an undiscovered potential in future stroke treatment. Supplementary to thrombolysis, we suggest that future stroke treatment can be improved by drug-induced hypothermia at the first responder’s pick up of the stroke patient before hospitalization. We have performed an intelligence based systematic review to seek information about the sequential order of pathomechanisms in stroke, and matched each pathomechanism with appropriate drugs in clinical trials to elucidate a multi-target treatment in acute ischemic stroke (AIS). In order to find pathomechanisms, we searched PubMed for review articles on the pathophysiology of AIS and selected abstracts. Additionally, we searched PubMed for articles describing potential neuroprotective drugs in clinical trials. Pathomechanisms were classified according to time up to thrombolysis (48h). Drugs in clinical trials were matched with their respective target pathomechanism(s). Once paired, drugs counteracting thrombolysis and standard hospital stroke treatment were discharged. The remaining drugs were scored for adverse events, and drugs with only mild side effects were finally revised for possible drug interference. This sequential list of a potential multi-target treatment in AIS supplementary to thrombolysis were then matched with the possible use of drug-induces hypothermia. In conclusion, drug-induced hypothermia during the initial hours after stroke prepare the patient for later endovascular cooling at the hospital by lowering the shivering threshold, and further prolong the period of therapeutic hypothermia in stroke treatment. As druginduced hypothermia is not obstructing rt-PA-thrombolysis, and can be induced simply by swallowing a pill, this treatment can be initiated very early in the sequential cascade of ischemic pathomechanisms. These are ameliorated even by mild hypothermia. The use of dopaminergic, serotonergic and cannabinergic drugs in druginduced mild hypothermia will be discussed.
65
S31 Revisit Mitochondria in Neurodegeneration S31-01 IMPAIRED MITOCHONDRIAL PROTEIN IMPORT IN NEURODEGENERATIVE DISEASES Hiroko Yano 1, Sergei Baranov 2, Oxana Baranova 2, Jinho Kim 2 Yanchun Pan 1, Svitlana Yablonska 2, Diane Carlisle 2 Robert Ferrante 2, Albert Kim 1, Robert Friedlander 2 1 Washington University School of Medicine, Neurological Surgery, St. Louis, USA 2 University of Pittsburgh, Neurological Surgery, Pittsburgh, USA Mitochondrial dysfunction is thought to play an important role in the pathogenesis of many neurodegenerative disorders, including Huntington’s disease (HD), a progressive neurodegenerative disease caused by an abnormal polyglutamine expansion in the huntingtin (Htt) protein. However, the mechanisms driving mitochondrial dysfunction and neurodegeneration in HD remain largely unknown. Translocation of nuclear-encoded mitochondrial proteins into the mitochondria is essential for mitochondrial function and homeostasis. We identified a specific interaction between mutant Htt and the TIM23 complex, the translocase of the inner mitochondrial membrane, which mediates import of presequence-containing proteins to the matrix, the inner membrane, and the intermembrane space. Remarkably, recombinant mutant Htt directly inhibited mitochondrial protein import in vitro. Furthermore, we observed defective protein import in mitochondria isolated from brain synaptosomes of presymptomatic HD model mice and from mutant Htt-expressing primary neurons, suggesting that deficient protein import is an early event in HD. Importantly, the mutant Htt–induced impairment of mitochondrial protein import and subsequent neuronal death were attenuated by overexpression of TIM23 complex subunits. Collectively, these findings provided a mechanism by which mutant Htt directly impairs mitochondrial protein import through an interaction with the import machinery and demonstrate that defective TIM23-dependent protein import triggers mutant Httinduced neuronal death. Given that impairment of mitochondrial protein import has also been implicated in other neurodegenerative diseases, including amyotrophic lateral sclerosis and Alzheimer’s disease, restoring mitochondrial protein import activity may represent a new therapeutic strategy for neurodegenerative diseases exhibiting mitochondrial protein import deficiency.
S31-02 LOSS OF BAX-INTERACTING FACTOR-1 EXACERBATES ALZHEIMER’S DISEASE PATHOLOGY Richard Morrison 1, David Wang 1, Yoshito Kinoshita 1 Chizuru Kinoshita 1, Takuma Uo 1, Bryce Sopher 2, Gwenn Garden 2 Christopher Keene 3, Karen Gylys 4, Hong-Gang Wang 5 1 Univeristy of Washington, Neurological Surgery, Seattle, USA 2 University of Washington, Neurology, Seattle, USA 3 University of Washington, Pathology, Seattle, USA 4 UCLA, Nursing, Los Angeles, USA 5 The Pennsylvania State University , Pharmacology, Hershey, USA Bax-interacting factor-1 (Bif-1) is a multifunctional protein involved in regulation of apoptosis, autophagy and mitochondrial function. We recently described neuron-specific alternatively __________________ 2015 Transactions of the American Society for Neurochemistry ®
spliced isoforms of Bif-1 that confer neuroprotection. To examine whether Bif-1 mediated neuroprotection could be a novel therapeutic target for Alzheimer’s Disease (AD) we employed a double mutant amyloid precursor protein and presenilin 1 (APPswe/ PS1dE9) mouse model of AD and observed that expression of neuron-specific Bif-1 isoforms is decreased with disease progression. To determine if reduced Bif-1 participates in AD pathogenesis, we crossed Bif-1 knockout mice (Bif-1 KO) with APPswe/PS1dE9 mice. The absence of Bif-1 accelerated disease onset and progression in AD/Bif-1 KO mice, which showed more plaques, astrogliosis, synaptic degeneration, cognitive impairment and mortality than APPswe/PS1dE9 mice. In mouse primary cortical neuron cultures, overexpression of neuron specific Bif-1 isoforms protected against beta-amyloid induced apoptosis and mitochondrial dysfunction. Protein and mRNA levels of neuron-specific Bif-1 isoforms were also selectively decreased in the cerebral cortex of patients with Alzheimer’s disease, suggesting that loss of Bif-1 mediated neuroprotection may participate in AD pathogenesis and that restoration of Bif-1 function could be a novel therapeutic target for AD. We also observed that Bif-1KO mice develop synaptic degeneration and behavioral impairment by 12 months, underscoring the importance of Bif-1 in neuronal homeostasis. Since exposure to beta amyloid leads to reduced expression of Bif-1 and loss of Bif-1 leads to increased accumulation of beta amyloid in vivo, our findings suggest that Bif-1 is a key mediator of a feed forward mechanism of AD disease pathogenesis in which beta-amyloid deposition reduces neuron-specific Bif-1, which in turn enhances beta-amyloid accumulation and neuronal sensitivity to beta-amyloid toxicity.
S31-03 REGULATION OF MITOCHONDRIAL FISSION IN HUNTINGTON’S DISEASE Xin Qi Case Western Reserve University, Physiology and Biophysics, Cleveland, USA Huntington’s disease (HD) is a fatal and inherited neurodegenerative disease which is caused by a glutamine-coding CAG expansion within exon 1 of the huntingtin gene. Although the genetic mutation associated with the disease has been identified, the molecular and cellular mechanism of HD is not yet understood and successful treatment for this disease remains elusive. Accumulating evidence indicates that mitochondrial dysfunction plays an important role in the pathogenesis of HD. Mitochondria are highly dynamic organelle which frequently undergo fusion and fission. This dynamic process controls not only mitochondrial morphology, but also the subcellular location and function of mitochondria. Defects in either fusion or fission limit mitochondrial motility, decrease energy production and increase oxidative stress, thereby promoting cell dysfunction and death. Dynamin-related protein 1 (Drp1) is a large GTPase and a primary protein governing mitochondrial fission. Recent studies have highlighted the causal role of Drp1-mediated excessive mitochondrial fission in neuronal death in a variety of HD models. However, how Drp1 hyperactivation mediates mitochondrial damage and neurodegeneration in HD and whether pharmacological inhibition of Drp1 activation is sufficient __________________ 66
S31 Revisit Mitochondria in Neurodegeneration to reduce mutant Htt (mtHtt)-induced neurotoxicity and neurodegeneration are not known. Using a rational approach for peptide design, we recently developed a peptide inhibitor, P110, which selectively blocked Drp1-induced excessive mitochondrial fission. We found that treatment with P110 inhibited mtHtt-induced excessive mitochondrial fragmentation and improved mitochondrial functions in various HD cell culture models. Moreover, P110 treatment reduced neurite shortening and cell death in patient induced pluripotent stem cells (iPS cells)-derived GABAergic neurons and medium spiny neurons, which are susceptible to neurodegeneration in HD patients. Furthermore, sustained treatment with P110 for eight-weeks in HD transgenic R6/2 mice reduced mitochondrial dysfunction, motor deficits, neuropathology and mortality. Together, these findings suggest a predominant role of mitochondrial fission impairment in the pathogenesis of HD. Therefore, inhibition of Drp1-dependent excessive mitochondrial fission with a P110-like inhibitor may prevent or slow down the progression of HD in humans.
S31-04 MITOCHONDRIAL PROTEIN DEACETYLATION AND NEURODEGENERATION Wenzhen Duan Johns Hopkins University, Psychiatry/Neurobiology, Baltimore, USA
neuronal survival. In addition to their prominent role in energy metabolism, mitochondria perform other essential functions, including the regulation of calcium homeostasis, oxidative stress response, and activation of cell death pathways. Consequently, mitochondrial pathophysiology aggressively promotes neuronal dysfunction and loss of synaptic viability, leading ultimately to neurodegeneration. These broad functions explicate the need for the well-orchestrated biogenesis of mitochondrial proteins and balanced dynamics to avoid neurodegeneration and pathological consequences. The most compelling evidence implicates mitochondrial impairment is involved in neurodegenerative disorders. Mitochondria are highly dynamic organelles that, upon the neuron’s metabolic demands or pathological conditions, frequently change their shape. A number of mitochondria shaping proteins control mitochondrial fission and fusion events, leading to a continuous remodeling of mitochondrial networks: increased fission or decreased fusion can lead to mitochondrial fragmentation. Mitochondrial dynamics involves the shape, size, distribution, transport and number of mitochondria in the neuron and is controlled by a balance between mitochondrial fusion and fission events. In addition, mitochondrial proteins are subject to extensive lysine acetylation and proper localization. Recent studies suggest that mitochondrial fragmentation, import and protein deacetylation play a pivotal role in neurodegeneration. We will present our recent research progress in understanding the critical roles of mitochondrial protein deacetylation in Huntington’s disease, and highlighting a potential novel approach for therapeutic development by targeting mitochondria in Huntington’s diseases.
Mitochondria are unique and complex organelles intimately involved in several key cellular processes that are critical to ensure __________________
2015 Transactions of the American Society for Neurochemistry ®
67
Colloquia C01 Proinflammatory Mechanisms in Neurodegeneration C01-01
C01-02
TARGETING SIGNALING PATHWAYS THAT LEAD TO INFLAMMATORY CYTOKINE DYSREGULATION IN NEURODEGENERATIVE DISORDERS Linda Van Eldik 1, 2, Adam Bachstetter 1, D Martin Watterson 3 1 University of Kentucky, Sanders-Brown Center on Aging, Lexington, USA 2 University of Kentucky, Anatomy & Neurobiology, Lexington, USA 3 Northwestern University Feinberg School of Medicine, Pharmacology, Chicago, USA
THE CONTRIBUTION OF APP TO MICROGLIAL ACTIVATION IN ALZHEIMER’S DISEASE Colin Combs Univ of North Dakota, Dept of Pharm., Phys. & Ther., Grand Forks, USA
Evidence from clinical studies and preclinical animal models suggests that proinflammatory cytokine overproduction from activated glia is a potential driving force for pathology progression in neurodegenerative conditions such as Alzheimer’s disease and sequelae to traumatic brain injury. There are also potential clinical susceptibility linkages when proinflammatory cytokine responses are part of the causative pathophysiology progression. For example, a prior head injury can increase the risk for later development of dementia. This raises the possibility that selective targeting of the dysregulated cytokine response as a component of the stress responses contributing to neuronal/synaptic dysfunction may be a useful therapeutic approach, if the appropriate dosing (e.g., time window and duration) can be elucidated. We have developed two distinct classes of CNS-active, small molecule experimental therapeutics that selectively restore injury- or disease-induced overproduction of proinflammatory cytokines back towards homeostasis. One class is a novel, isoform-selective p38! MAPK inhibitor drug candidate currently in IND-enabling preclinical development. The p38! MAPK is a key protein kinase involved in the pathological upregulation of proinflammatory cytokines and other neuroinflammatory responses in activated glia, as well as in stress-related axonal and synaptic dysfunction in neurons. The second class of experimental therapeutics has entered clinical development. These are not p38 inhibitors, but instead emerged from a less biased, functional discovery approach for compounds that selectively suppress stressor-induced glial activation and overproduction of proinflammatory cytokines that contribute to pathology progression. Both classes of compounds attenuate excessive glial activation, cytokine production and downstream neurologic sequelae in CNS disease models. Our ongoing in vivo studies with these two distinct and complementary drug candidate classes add to the growing body of evidence that documents dysregulated glial activation and proinflammatory cytokine overproduction as a common early pathophysiologic mechanism that is a viable pathway based therapeutic target for diverse neurodegenerative disorders.
2015 Transactions of the American Society for Neurochemistry ®
Proteolytic processing of the amyloid precursor protein (APP) leads to generation of the amyloid beta (A") peptide that accumulates in the brain as extracellular plaques in normal aging and Alzheimer’s disease where it may serve as an activating stimulus for microglia. Our prior work has shown that APP can serve as an adhesion-associated proinflammatory receptor on monocytic cells leading to tyrosine kinase-based activation and A" secretion. We hypothesized that microglia might also use APP as an adhesion-associated receptor involved in regulating their response to A" deposits in the brain. To test this idea we stimulated human THP-1 monocytes and primary murine microglia with A" oligomers or fibrils to find that only cells expressing APP were stimulated by oligomeric peptide. We made similar observations in vivo following intracerebroventricular infusion of oligomeric A" into mice. Finally, we crossed APP-/- mice with mice over-expressing human mutant APP/PS1 under the control of the prion promoter to determine whether loss of APP would alter microgliosis in a setting in which both oligomeric and fibrillar peptide are present in vivo. The APP-/x APP/PS1 mice had similar fibrillar A" plaque deposition compared to APP/PS1 mice. However, as predicted, APP-/- x APP/PS1 had significantly decreased microgliosis compared to APP/PS1 mice which correlated with improved memory performance.
C01-03 EFFECTS OF COMPLEMENT ACTIVATION FRAGMENT C5A IN ALZHEIMER'S DISEASE MOUSE MODELS. Andrea Tenner University of California - Irvine, Dept Molecular Biology & Biochem, Irvine, USA Components of both the classical and the alternative pathways of complement, an effector mechanism of the innate immune system that can be activated by fibrillar amyloid beta (A"), have been found to colocalize with fibrillar A" plaques in brains of Alzheimer’s disease (AD) patients as well as in mouse models of the disease. Studies in animal models of AD have provided evidence that initial complement activation components can be beneficial while the later activation events of the cascade, such as the generation of the proinflammatory peptide C5a, can be detrimental. To validate the potential of C5a and/or C5aR1 as a therapeutic target, C5aR1 (CD88) knockout mice were crossed to the Arctic transgenic AD mouse model, and mice containing a brain specific expression transgene for C5a were crossed to Arctic mice and to the 3xTg a __________________ 68
C01 Proinflammatory Mechanisms in Neurodegeneration distinct murine model of AD. While Arctic mice exhibited deficits in the hippocampal dependent object location memory task (OLM) task at 10 month old of age, the deficits were not apparent in the absence of C5aR1, although the amount of amyloid in the Arctic model was unaffected by the absence of the C5aR1. In contrast, behavioral deficits were accelerated in both the Arctic and 3xTg AD models overexpressing C5a, while there was no such impairment in any of the non-AD mice ie. in the absence of fibrillar amyloid !, consistent with a detrimental effect of C5a when present in conjunction with fibrillar A! in brain. These results support the hypothesis that upon generation of C5a as a consequence of activation of the complement pathway by fibrillar amyloid, the interaction with its receptor C5aR1 results in a neurotoxic and/or inflammatory environment that contributes to the loss of cognitive function. The data further implicate the potential of this activation fragment or its proinflammatory receptor as a therapeutic target to slow the progression of cognitive loss seen in the human disease.
C01-04 INFLUENCE OF AMYLOID-BETA AGGREGATION STATE ON INNATE IMMUNE PATHWAYS Michael Nichols, Geeta Paranjape, Shana Terrill-Usery Sanjib Karki, Benjamin Colvin, Lisa Gouwens University of Missouri-St. Louis, Chemistry and Biochemistry, St. Louis, USA Inflammation in Alzheimer’s disease (AD) arises from accumulation of activated microglia around extracellular plaques in the brain. The plaques, composed primarily of amyloid-! peptide (A!), are fibrillar at the core but recent data suggest that the plaque __________________
2015 Transactions of the American Society for Neurochemistry ®
composition is more complex. Our own in vitro studies demonstrate that microglia are sensitive to structural changes in A! and different A! species. We found that soluble size-exclusion chromatography (SEC)-isolated A!42 protofibrils were much stronger stimulators of microglia than insoluble A!42 fibrils or SEC-purified A!42 monomer. Characterization by electron microscopy revealed classic curvilinear structures for protofibrils with lengths less than 100 nm but significant polydispersity. Fibrils were substantially longer at greater than 1 µm. Dynamic and multi-angle light scattering measurements of the protofibrils revealed a mean hydrodynamic radius of 21 nm and a molecular weight range of roughly 200-2500 kD (45-550 monomers) over multiple preparations. Confocal microscopy studies showed significant binding of protofibrils to the microglia surface with little observed for fibrils and monomers. Additional analyses revealed rapid uptake ( 1 mm length. Our results are consistent with early neurodevelopmental defects observed in mice deficient of ceramide synthase 2, the enzyme synthesizing C24:1 ceramide, and the protective effect of HDAC6 inhibitors on neurons. In summary, our study suggests that C24:1 ceramide is an endogenous inhibitor of HDAC6, which is equally important for primary cilium formation and neurite extension. Supported by NSF grant 112157.
OP01-02 INOSITOL PROTECTS ZEBRAFISH IN MODEL OF FETAL ALCOHOL SYNDROME Sparkle Williams, Latoya Paul, Brian Sims University of Alabama Birmingham, Pediatrics/Neonatology, Birmingham, USA Fetal Alcohol Syndrome accounts for a number of children with developmental disabilities. Exposure to alcohol during development affects multiple organs including the brain, eyes, heart and bones. The most affected of these is the brain. The exact mechanism involved in ethanol-induced teratogenesis is under intense investigation. One critical pathway linked to attenuating the effects of __________________ 2015 Transactions of the American Society for Neurochemistry ®
ethanol induced toxicity is the sonic hedgehog pathway. Sonic hedgehog is a transcription factor that has a key role in the development of the central nervous system. We have found a novel regulator of sonic hedgehog in D-inositol. D-inositol is important in brain development but to what extent is still unclear. In this study, we have found that D-inositol decreases sonic hedgehog by more than 5-fold. Inositol also had a dose dependent improvement in neurobehavior which was statistically significant. It is very plausible that the regulation on sonic hedgehog has a significant effect on embryogenesis and brain maturation. The knowledge gained in this project will give valuable insight into potential strategies that may address the pathogenesis of ethanol-induced toxicity.
OP01-03 SELECTIVE REGULATION OF CDK5 ACTIVATORS BY HUD CONTROLS THE DEVELOPMENT AND FUNCTION OF HIPPOCAMPAL NEURAL CIRCUITRY Megan Allen, Wenqi Li, Guanglu Liu, Wei Feng, Andrew Bankston Yue Feng Emory University, Department of Pharmacology, Atlanta, USA The RNA-binding protein HuD plays key roles in neuronal development and synaptic plasticity through post-transcriptional regulation of its mRNA targets. Recent studies from model cell lines suggest that HuD stabilizes the mRNA that encodes p35. The p35 protein isan activator for the serine/threonine kinase Cyclin-dependent kinase 5 (Cdk5), whichplays key roles in brain development. However, Cdk5 can be activated not only by p35, but also by another distinct Cdk5 activator called p39. Whether HuD indeed regulates Cdk5 activity in brain neurons and the functional importance of such regulation still remains unknown. Here we report that HuD binds and stabilizes not only p35 mRNA, but also p39 mRNA, in cultured brain neurons and hippocampus in vivo. Surprisingly, elevated HuD expression in the hippocampus leads to increased protein expression of p39, but not p35, possibly involving up-regulation of miRNAs that selectively target p35. Importantly, Cdk5 activity and target phosphorylation are increased in the hippocampus of transgenic mice that harbor exogenous hippocampal HuD expression (HuDtg+). HuDtg+ mice also display aberrant over-projection of mossy fiber axons from DGCs, a hallmark of pathological plasticity in epileptogensis. Interestingly, genetic removal of p39 ameliorates this mossy fiber phenotype. Furthermore, preliminary data suggest HuD-dependent regulation of p39 may contribute to seizure response in a pharmacological model of epilepsy. In conclusion, our studies reveal a novel functional link between HuD and the Cdk5 pathway in controlling the development and function of the neuronal circuitry in the hippocampus that plays important roles in epileptogenesis. 77
OP01 Oral Session I - Post Doc Neurochemistry Symposium This work was supported by NIH BCMB training grant 5T32GM008367
OP01-04 ROLE OF RNA/DNA BINDING PROTEINS, TDP-43 AND FUS IN DNA DAMAGE RESPONSE: ETIOLOGICAL LINKAGE TO AMYOTROPHIC LATERAL SCLEROSIS Muralidhar Hegde Houston Methodist Research Institute, Radiation Oncology and Neurology, Houston, USA Accumulation of genome damage including oxidized bases, single- and double-strand breaks, in affected brain cells has been linked to Motor neuron diseases including ALS whose underlying cause(s) are not completely understood. We recently demonstrated that transition metals iron and copper that accumulate in neurodegenerative brain act as a double-edged sword by both increasing oxidative genome damage and preventing their repair and thus could play a role in accumulation of unrepaired genome damage in neurons leading to cell death (Hegde et al, J Biol Chem 2010: 285, 28812-25). Here, we provide evidence for the first time that RNA binding proteins TDP-43 and FUS, whose nuclear clearance and simultaneous cytoplasmic deposition is a hallmark feature in ALS and other neurodegenerative diseases, is required for efficient DNA strand break repair in neurons. We demonstrate that: (1) TDP-43 stably interacts with DSBR proteins in hNSC-derived Spinal Motor Neurons, which was enhanced after treatment with DSB-inducing radiation/bleomycin. (2) TDP-43 is recruited to the DSB sites neuronal cells, and (3) TDP-43’s overall as well as nucleus-specific depletion markedly increased accumulation of DSBs in motor neurons and sensitized the cells to radiation. These results are consistent with the dramatic accumulation of unrepaired DSBs in postmortem brains of ALS-affected human patients and a distinct nuclear clearance of TDP-43 in these affected neurons. Our more recent studies show that FUS, unlike TDP-43 is required for single strand break repair in neurons via activation Ligase III protein. Thus deficiency in DNA strand break repair may be a key etiologic factor in neurodegenerative diseases. These results suggest that defects in genome damage repair in neuronal genome represents a common basis for ALS and other neurodegenerative diseases. (Supported by Alzheimer’s Association, ALS Association and Muscular Dystrophy Association).
OP01-05 MYELIN-ASSOCIATED GLYCOPROTEIN MODULATES PROGRAMED CELL DEATH OF MOTONEURONS VIA NGR/P75NTR RECEPTOR-MEDIATED RHOA ACTIVATION Anabela Palandri 1, Victoria Rozés Salvador 1, Jose Wojnaki 1 Ana L. Vivinetto 1, Ronald L. Schnaar 3, Pablo H. H. Lopez 1, 2 1 Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Laboratorio de Neurobiología, Cordoba, Argentina 2 Universidad Nacional de Córdoba, Facultad de Psicología, Cordoba, Argentina 3 Johns Hopkins University, Departaments of Pharmacology and Neuroscience, Baltimore, USA Myelin-Associated Glycoprotein (MAG) is a minor constituent of the nervous system expressed at the periaxonal layer of myelinated axons. By engaging multiple axonal receptors, including Nogoreceptors (NgRs), MAG exerts a nurture effect on axons it ensheaths. Pharmacological activation of NgRs has a modulatory role on p75NTR-dependent postnatal apoptosis of motoneurons (MNs). NgRs are part of a receptor complex which includes p75NTR, Lingo-1 and gangliosides. Upon ligand binding, this multimeric complex activates RhoA/ROCK signalling in a p75NTR-dependent manner. The aim of this study was to analyze a possible modulatory role of MAG on MNs apoptosis during postnatal development. A time course study showed that Mag-null mice suffer a loss of MNs during the first postnatal week. Also these mice exhibited increased susceptibility in an animal model of p75NTR-dependent MNs apoptosis induced by nerve-crush injury, which was prevented by treatment with MAG-Fc. The protective role of MAG was further confirmed in in vitro models of p75NTR-dependent MN apoptosis including the MN cell line MN1. Lentiviral expression of shRNA sequences targeting NgRs abolished protection by MAG-Fc. Analysis of RhoA activity using a FRET-based RhoA biosensor showed that MAG-Fc activates RhoA. Pharmacological inhibition of p75NTR/RhoA/ROCK pathway or overexpression of a p75NTR mutant lacking binding to RhoA blocked MAG-Fc protection. The role of RhoA/ROCK signaling was confirmed in the nerve-crush model, where pre-treatment with Y27632 (ROCK inhibitor) blocked pro-survival effect of MAG-Fc. Overall these findings identify a new protective role of MAG as a modulator of apoptosis of MNs during postnatal development by a mechanism involving p75NTR/ RhoA/ROCK signaling pathway. In addition our results highlight the relevance of the nurture/protective effect of myelin on neurons.
OP01-06 ADDRESSING THE IN VIVO CONTRIBUTION OF JNK3 TO HUNTINGTON’S DISEASE PATHOGENESIS Rodolfo Gatto 1, Ehsan Tavasoli 1, Yaping Chu 2, Jeffrey Cordower 2 Gerardo Morfini 1 1 University of Illinois, Anatomy and Cell biology, Chicago, USA 2 Rush University Medical Center, Neurosurgery, Chicago, USA Expansion of a polyglutamine tract in the protein huntingtin (htt) results in Huntington disease (HD), a disease categorized by progressive degeneration of specific neuronal populations within the cerebral cortex and the striatum. Work from our group and others indicate that axonal degeneration represents a critical pathogenic __________________ 2015 Transactions of the American Society for Neurochemistry ®
78
OP01 Oral Session I - Post Doc Neurochemistry Symposium event in HD-affected neurons. Accordingly, multiple independent reports showed that pathogenic forms of Htt (mHtt) inhibit axonal transport, a cellular process essential for the appropriate maintenance of axonal connectivity. Further, our prior work indicated this toxic effect of mHtt involves abnormal activation of the c-Jun amino-terminal kinase (JNK) pathway. Despite this knowledge, the relationship between JNK activation and axonal degeneration in HD remains unknown.__________________
2015 Transactions of the American Society for Neurochemistry ®
Results from quantitative immunohistochemical and immunoblotting studies here revealed increased activation of JNKs in both the R6/2 mouse model and in HD brains. Significantly, active JNK mainly localized to HD-vulnerable neuronal populations within the striatum and in the cerebral cortex. Together, results from these studies suggest that differential distribution and activation of JNK kinases might represent an important factor contributing to the cellular topography of HD.
79
OP02 Oral Session II - Graduate Student Neurochemistry Symposium OP02-01 VISUALIZING THE NEUROPATHOLOGY UNDERLYING GRAY MATTER ATROPHY IN EAE USING CLARITY Allan MacKenzie-Graham 1, Rory Spence 2, Florian Kurth 1 Noriko Ito 1, Chandler Mongerson 1, Shannon Wailes 1, Mavis Peng 1 1 University of California, Los Angeles, Neurology, Los Angeles, USA 2 Scripps College, Science, Los Angeles, USA Gray matter atrophy correlates strongly with disease duration and clinical disability in multiple sclerosis and disease duration in experimental autoimmune encephalomyelitis (EAE). However, the mechanisms that underlie gray matter atrophy remain unknown. In this study we combined magnetic resonance imaging (MRI) with clear lipid-exchanged acrylamide-hybridized rigid imaging-compatible tissue-hydrogel (CLARITY), a recently developed optical clearing technology that permits microscopic imaging of the entire brain, to better understand these mechanisms. EAE was induced in female THY1-YFP+ mice which were then scanned using MRI. The mice were sacrificed, their brains and spinal cords optically cleared using CLARITY, and imaged intact using confocal microscopy. MRI volumetry demonstrated decreased cerebral cortex volumes in mice with EAE compared to healthy controls. Axons were followed longitudinally in intact spinal cords revealing that 61% axons exhibited ovoids in mice with EAE with a mean of 22 ovoids per axon over a 5 mm length. 8% of axons in mice with EAE exhibited end bulbs. Healthy control mice exhibited almost no axonal abnormalities over a 5 mm length. Furthermore, the number of layer V cortical neurons was decreased in intact cerebral hemispheres in mice with EAE compared to healthy controls. Cross-modality correlations revealed a direct relationship between cortical volume loss and spinal cord end bulb number, but not ovoid number. We conclude that cortical atrophy is strongly associated with axonal transection, but not axonal injury, in the spinal cord in EAE.
OP02-02 MYELIN DISRUPTION LEADS TO TARGETED BEHAVIORAL DEFICITS Elizabeth Gould, Nicolas Busquet, Diego Restrepo, Wendy Macklin University of Colorado, Denver, Cellular and Developmental Biology, Aurora, USA Myelin is required for proper nerve conduction and has an important role in normal axonal function. Motor function and learning depend on myelination. Alteration in myelin structure and quantity is observed in various neurodegenerative diseases. However, little is known about the neuronal and behavioral consequences of myelin disruption alone. To address the contribution of myelin function to behavioral and cognitive deficits, we used a mouse line lacking the myelin proteolipid protein (PLP). These mice generate myelin but they exhibit progressive myelin dysfunction. We tested 8 month-old PLP knockout PLP(-/Y) male mice in a battery of behavioral tests. As expected from published studies, PLP(-/Y) mice performed similarly to age-matched controls __________________ 2015 Transactions of the American Society for Neurochemistry ®
in the Rotarod, a classical test of motor function. Exploration of the center of an Open Field arena was reduced in PLP(-/Y) mice (159±23 seconds vs. 239±25 seconds, p