36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH...

72
36 th Annual Conference of the North American Section of ISHR Hilton Riverside Hotel May 30 - June 2 2017 New Orleans, Louisiana International Society for Heart Research North American Section

Transcript of 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH...

Page 1: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

36 th Annual Conference of the

North American Section of ISHR

Hilton Riverside Hotel May 30 - June 2

2017 New Orleans, Louisiana

International Society for Heart Research North American Section

Page 2: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

2

XXXVI North American Section New Orleans, Louisiana

TABLE OF CONTENTS

LOCAL ORGANIZING & ISHR COMMITTEE 3

WELCOME LETTER 4

PROGRAM AGENDA

TUESDAY, MAY 30th 5

WEDNESDAY, MAY 31st 10

THURSDAY, JUNE 1st 14

FRIDAY, JUNE 2nd 18

POSTER SESSION I 21

POSTER SESSION II 27

Page 3: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

3

36th Annual Conference of the North American Section of ISHR

Local Organizing Committee

Program Chair David J. Lefer, Ph.D.

Program Co-Chairs

Richard S. Vander Heide, M.D., Ph.D., Daniel Kapusta, Ph.D., and Kurt J. Varner, Ph.D.

ECI Program Chairs Traci T. Goodchild, Ph.D. and Imran N. Mungrue, Ph.D.

Social Chairman

Jeffery D. Molkentin, Ph.D.

ISHR Officers and Council Members President: Gary D. Lopaschuk President Elect: Peipei Ping Past President: Elizabeth Murphy Secretary: Susan Howlett Treasurer: Evangelia Kranias Recording Secretary: Thomas M. Vondriska Executive Secretary: Leslie Lobaugh Council Members 2012-2018: Chris Baines, Asa Gustafson, Joan Heller Brown, Timothy Kamp, Pieter de Tombe, Jun Sadoshima, Walter Koch, Gary Lopaschuk

2015-2021: Dale Abel, Burns Blaxall, John Elrod, Joseph Hill, Tim O'Connell, Brian O'Rourke, Monte Willis and Rick Vanderheide

Page 4: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

4

North American Section www.american.ishrworld.org

Office of the President:

Gary Lopaschuk, Ph.D.

Dear Colleagues,

It is my pleasure to welcome you to the 36th Annual Conference of the North American Section of the International Society for Heart Research in New Orleans, Louisiana. The meeting is being hosted by Dr. David Lefer and his colleagues, who have put together a wonderful scientific program that addresses a number of cutting edge topics in the cardiovascular research arena. The meeting will also provide both established and young investigators an ideal venue for exchange of scientific ideas. Dr. Lefer and his colleagues have also put together a wonderful social program that will expose conference registrants to the rich culture that New Orleans has to offer. My sincerest thanks go out to Dr. Lefer and his colleagues for the tremendous time and effort in putting together what I am sure will be an excellent congress.

I welcome you to New Orleans, and hope you enjoy the congress.

Sincerely,

Dr. Gary D. Lopaschuk President, NAS- ISHR

Page 5: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

5

N

TUESDAY

30th May

36th Annual ISHR-NAS Conference Early Career

Investigator Event

TIME ROOM & LOCATION ACTIVITY

ECI Registration opens.

ECI Symposium Chair: Samarjit Das, John Hopkins, Baltimore Co-Chair: Randi Parks, NHLBI/NIH, Bethesda

7:30 am Registration Desk

8:00am - 10:30am

Quarter Deck Room

1. COMPARTMENTALIZED Gaq-SIGNALING IN ADULT CARDIAC MYOCYTE Erika Dahl, University of Minnesota, Madison 2. THE CONTRIBUTION OF FATTY ACID AND KETONE BODY OXIDATION TO ENERGY PRODUCTION INCREAS- ES IN THE FAILING HEART AND IS ASSOCIATED WITH A DECREASE IN CARDIAC EFFICIENCY Kim Ho, University of Alberta, Alberta

3. miR-181c REGULATES MITOCHONDRIAL CALCIUM IN- FLUX BY TARGETING CYTOCHROM C OXIDASE SUBU- NIT 1 Samarjit Das, Johns Hopkins, Baltimore

4. THE W792R MUTATION IN CARDIAC MYOSIN BINDING PROTEIN-C REDUCES THE C6 Fnlll DOMAIN STABILITY AND CAUSES HYPERTROPHIC CARDIOMYOPATHY THROUGH HAPLOINSUFFICIENCY Dan Smelter, University of Wisconsin, Madison

5. GRK2-S670A MICE REVEAL CARDIOPROTECTION POST ISCHEMIA-REPERFUSION Priscila Sato, Temple University, Philadelphia

6. MECHANO-CHEMO-TRANSUCTION IS ATTENUATED I A RABBIT MODEL OF HEART FAILURE Rafael Shimkunas, University of California, Davis

7. MITOCHONDRIAL MEMBRANE PROTEIN SIGMAR1 REGULATES MITOCHONDRIAL DYNAMICS AND FUNC- TION Shafiul Alam, Louisiana State University Health Science Cen- ter, Shreveport

8. REGIONAL CARDIAC DENERVATION PRODUCES SU- PERSENSITIVITY OF MYOCARDIAL Ca2+ HANDLING TO B-ADRENERGIC STIMULATION Srinivas Tapa, University of California, Davis

9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

Page 6: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

6

TUESDAY

30th May

36th Annual ISHR-NAS Conference Early Career

Investigator Event

TIME ROOM & LOCATION ACTIVITY 10. CARDIOPROTECTION IN MICE WITH A KNOCK-IN MU-

TATION IN CYCLOPHILIN D (CypD-C202S): A SITE OF S- NITROSYLATION Georgios Amanakis, NHLBI/NIH, Bethesda

11. A ROLE FOR PPARα IN SEX DIFFERENCES IN CAR- DIAC HYPERTROPHY Natasha Fillmore, NIH, Bethesda

10:30am - 11:00am

Foyer Coffee Break.

11:00am - 12:00pm

Quarter Deck

Room 1

Career Development Panel: Session A : Graduate Stu- dents Moderator: Catherine Makarewich, University of Texas, Southwestern Medical Center, Dallas Panelists: Alice Kane, Dalhousie University, Halifax Edward Lau, Stanford University, Stanford Randi Parks, NHLBI/NIH, Bethesda Phillip Bidwell, University of Cincinnati, Cincinnati

11:00am - 12:00pm

Quarter Deck

Room 2

Career Development Panel: Session B: Post Doctoral Fel- lows & Junior Faculty Moderator: Stephen Lange, University of California, San Die- go Panelists: Joan Heller Brown, University of California, San Diego Jeffery Robbins, University of Cincinnati, Cincinnati Sarah Franklin, University of Utah, Salt Lake City Mark Kohr, Johns Hopkins University, Baltimore

11:00am- 12:00pm

Compass Room ISHR American Section: Council Meeting #1

12:15pm - 2:00pm

Drago’s Seafood Restaurant

ECI Luncheon Invited Guests: Sarah Franklin, University of Utah, Salt Lake City Joan Heller Brown, University of California, San Diego Jeffery Robbins, University of Cincinnati, Cincinnati Susan Howlett, Dalhousi University, Halifax Mark Sussman, San Diego State University, San Diego Asa Gustafsson, University of California, San Diego Junichi Sadoshima, Rugters University, New Brunswick Brian O’Rourke, Johns Hopkins University, Baltimore Tom Vondriska, University of California, Los Angeles Mark Kohr, Johns Hopkins University, Baltimore Christopher Baines, University of Missouri, Columbia

Page 7: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

7

TIME ROOM & LOCATION ACTIVITY

2017 ISHR NA SECTION YOUNG INVESTIGATOR AWARDS Annual Young Investigator Awards (YIAs) were established by the North American Section of the ISHR to recognize out- standing research in the field of cardiovascular science by junior Investigators. From applications received for this meet- ing, finalists in each of the two categories, Graduate Students and Early Postdoctoral Fellows (within 4 years of completing their degree) and Senior Postdocs and Early Assistant Profes- sors (between 4 and 10 years after completing their degree), were selected by a Committee to present their work at our meeting. A panel of judges will make the final award (presented at the banquet) based on the scientific merit of a submitted unpublished manuscript, quality of the oral presen- tation, and responses to questions asked during the discus- sion period. The finalists, their presentation titles and their mentors are listed below. Young Investigator Competition (Junior Science) Chair: Peipei Ping, UCLA, Los Angeles

2:30pm - 5:00pm

Quarter Deck Room

2:30pm 4:00pm

2:30pm 2:50pm

1. CANONICAL TRANSIENT RECEPTOR POTENTIAL 6 CHANNEL AMELIORATES INCREASED CARDIAC SNITRO- SYLATION IN DUCHENNE MUSCULAR DYST Heaseung Sophia Chung, John Hopkins School of Medicine, Baltimore Mentor: Jenny Van Eyk

2. The SPHINGOSINE-1-PHOSPHATE RECEPTOR MODU- LATOR, FTY720, REVERSES DIASTOLIC DYSFUNCTION AND HYPERTROPHY IN HYPERTROHIC CARDIOMYOPA- THY David M. Ryba, University of Illinois, Chicago Joint Mentor: John Solaro and Beata Wolska

3. PEDIATRIC DILATED CARDIOMYOPATHY HEARTS DIS- PLAY A GENE EXPRESSION PROFILE CONSISTENT WITH PLURIPOTENCY AND DEDIFFERNTIATION Kathleen Woulfe, University of Colorado, Denver Joint Mentor: Carmen Sucharov and Brian Stauffer

2:50pm - 3:10pm

3:10pm - 3:30pm

36th Annual ISHR-NAS

Conference Early Career Investigator Event

TUESDAY

30th May

Page 8: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

8

TIME ROOM & LOCATION ACTIVITY

3:30pm - 3:50pm

Quarter Deck Room 4. THE IMPACT OF AGE AND FRAILTY ON VENTRICULAR STRUCTURE AND FUNCTION IN C57BL/6 MICE Hirad Feridooni, Dalhousie University, Halifax Mentor: Susan Howlett

4:00pm - 5:30pm

Young Investigator Competition(Senior Science) Chair: Susan Howlett, Dalhousie University, Halifax

4:00pm - 4:20pm

1. ENHANCED ACTIVATION OF INFLAMMASOME PRO- MOTES ATRIAL FIBRILLATION Na Li, Baylor College of Medicine, Houston Mentor: Xander H.T. Wehrens

2. ENDOGENOUS HAX-1 REGULATES SERCA ACTIVITY AND OXIDATION DEPENDENT STABLITY Philip A Bidwell, University of Cincinnati, Cincinnati Mentor: Evangelia G. Kranias

3. GLUCOSE PROMOTES CELL GROWTH BY SUPPRESS- ING BRANCHED-CHAIN AMINO ACID DEGRADATION Dan Shao, University of Washington, Seattle Mentor: Rong Tian

4:20pm - 4:40pm

4:40pm – 5:00pm

36th Annual ISHR-NAS Conference Early Career

Investigator Event

TUESDAY

30th May

Page 9: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

9

TIME ROOM & LOCATION ACTIVITY

5:30pm- 6:30pm

Quarter Deck Room Opening Keynote Lecture Chair: Jeffery D. Molkentin, Cincinnati Children’s Hospital Medical Center, Cincinnati

5:30pm- Eric Olson, University of Texas, Southwestern Medical Center “THE MOLECULES AND MECHANISMS OF HEART DEVELOPMENT, DISEASE AND REGENERATION”

7:15pm - 9:00pm

River/Port/Starboard Room

Welcome Reception

36th Annual Conference of the

North American Section of ISHR

TUESDAY

30th May

Page 10: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

10

TIME ROOM & LOCATION ACTIVITY

7:00am- 8:00am

Compass Room Women In Science Breakfast The UCLA Cardiovascular Theme is proud to sponsor the Women in Science Breakfast at ISHR 2017.

7:30am Registration Desk Registration

8:00am- 9:00am

River/Port/Starboard Room

Research Achievement Award: Chair: Gary Lopaschuk, University of Alberta, Edmonton Co-chair: Metin Avkiran, King’s College London

8:00am Rong Tian, University of Washington “Navigating the Metabolic Maze: Do we see the light?”

9:00am- 10:40am

River/Port/Starboard Room

Session 1: Post-Translational Modification of the Cardiac Proteome - Sponsored by JMCC Chair: Traci T. Goodchild, Louisiana State University Health Sciences Center, New Orleans Co-chair: Frederica del Monte, Beth Isreal Deaconess Medical Center - Harvard, Boston

9:00am 1. PROTEIN DYNAMICS IN HEART Peipei Ping, University of California, Los Angeles

9:25am 2. O-GLCNACYLATION IN HEART FAILURE Steven Jones, University of Louisville, Louisville

9:50am 3. ACETYLATION OF TRANSCRIPTION FACTORS IN THE HEART Ravichandran Ramasamy, New York University, New York

10:15am- 10:40am

4: S-NITROSYLATION: INSIGHTS FROM THE FEMALE HEART Mark Kohr, John Hopkins University, Baltimore

9:00am- 10:40am

Quarter Deck Room Session 2: New Targets for Heart Failure Chair: Lea Delbridge, University of Melbourne, Australia Co-chair: Livia Hool, University of Western Australia, Perth

9:00am 1. EPIGENETIC CONTROL OF HEART FAILURE Joe Hill, University of Texas Southwestern, Dallas

9:25am 2. RENAL DENERVATION TO TREAT HEART FAILURE David Polhemus, Louisiana State University, New Orleans

36th Annual Conference of the

North American Section of ISHR

WEDNESDAY

31st May

Page 11: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

11

TIME ROOM & LOCATION ACTIVITY 9:50am 3. AMINO ACID CATABOLISM REPROGRAMMING IN

HEART FAILURE Yibin Wang, University of California, Los Angeles

10:15am- 10:40am

4. TARGETING MITOCHONDRIAL IRON Hossein Ardehali, Northwestern University, Chicago

10:40am -11:00am

Foyer Coffee Break.

11:00am- 12:40pm

River/Port/Starboard Room

Session 3: Stem Cells and Stem Cell Derived Factors Chair: Thomas Eschenhagen, Medical Center Hamburg- Eppendorf, Germany Co-chair: Yi Zhu, Tianjin Medical University, China

11:00am 1. DECONSTRUCTING CELL THERAPY FOR HEART DIS- EASE Ahmed Ibrahim, Capricor, Beverly Hills

11:25am 2. REPEATED CELL THERAPY: A NEW PARADIGM Roberto Bolli, University of Louisville, Louisville

11:50am 3. LINEAGE TRACING OF CARDIAC PROGENITOR CELLS Jeff Molkentin, Cincinnati Children’s Hospital Medical Center, Cincinnati

12:15pm- 12:40pm

4. IPSCS FOR PRECISION CARDIOVASCULAR MEDICINE Joseph Wu, Stanford Medicine, Stanford

11:00am -12:40pm

Quarter Deck Room Session 4: Mitochondrial Quality Control in Health and Disease Chair: Christopher P. Baines, University of Missouri Co-Chair: Shenu Bhuiyan, Louisiana State University Health Science Center , Shreveport

11:00am 1. MITOCHONDRIAL DYNAMICS Gerald Dorn, Washington University, St. Louis

2. MITOCHONDRIAL REMODELING IN HEALTH AND DISEASE Dan Kelly, Sanford Burnham Prebys Medical Discovery Institute, Lake Nona

3. NON-CANNONICAL MITOPHAGIC PATHWAYS Asa Gustafsson, University of California, San Diego

4. MITOCHONDRIAL QUALITY CONTROL Jonathan Burman, National Institutes of Health

11:25am

11:50am

12:15pm- 12:40pm

36th Annual Conference of the

North American Section of ISHR

WEDNESDAY

31st May

Page 12: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

12

TIME ROOM & LOCATION ACTIVITY

12:00pm- 2:00pm

Compass Room ISHR-International Council Meeting

12:45pm- 2:00 pm

Chart Room Lunch and Poster Session I Posters 054-105

2:00pm- 3:40pm

River/Port/Starboard Room

Session 5: Novel Targets in Cell Death and Survival Chair: Evripidis Gavathiotis, Albert Einstein College of Medicine, Bronx Co-chair: Lorrie Kirshenbaum, University of Manitoba, Canada

2:00pm 1. MITOCHONDRIAL PERMEABILITY TRANSITION- MOLECULAR MAKEUP AND THERAPEUTIC TARGET Paolo Bernardi, University of Padova, Italy

2. 3. NOVEL THERAPEUTIC APPROACHES IN CELL DEATH Richard Kitsis, Albert Einstein College of Medicine, New York

4. REDOX SIGNALING IN CARDIAC RENEWAL Hesham Sadek, University of Texas Southwestern, Dallas

2:25pm

2:50pm

3:15pm-

3:40pm

2:00pm- 3:40pm

Quarter Deck Room Session 6: Signaling Mechanisms in Heart Failure Chair: Imran Mungrue, Louisiana State University Health Sciences Center, New Orleans Co-chair: Stephen Lange, University of California, San Diego

2:00pm 1. DJ-1 SIGNALING IN HEART FAILURE John W. Calvert, Emory University, Atlanta

2:25pm 2. CAM KINASE II REGULATION OF INFLAMMATION AND FIBROSIS Joan Heller Brown, University of California, San Diego

2:50pm 3. MACROPHAGE MEDIATED SIGNALING Sumanth Prabhu, University of Alabama, Birmingham

3:15pm- 3:40pm

4. RELAXIN SIGNALING AND MYOCARDIAL PROTECTION Fadi Salloum, Virginia Commonwealth University, Richmond

3:40pm - 4:00pm

Foyer Coffee Break.

36th Annual Conference of the

North American Section of ISHR

WEDNESDAY

31st May

Page 13: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

13

TIME ROOM & LOCATION ACTIVITY

4:00pm- 5:40pm

River/Port/Starboard Room

Session 7: Stem Cell Mediated Myocardial Regeneration Chair: Luiz Sampaio, Texas Heart Institute, Houston Co-chair: Timothy J. Kamp, University of Wisconsin - Madison, Madison

4:00pm 1. C-KIT STEM CELL MEDIATED MYOCARDIAL REPAIR Jop Van Berlo, University of Minnesota, Minneapolis

4:25pm 2. PLOIDY: WE ARE SO MUCH MORE THAN DIPLOID Mark A. Sussman, San Diego State University, San Diego

4:50pm 3. MESENCHYMAL STEM CELLS Joshua Hare, University of Miami Health System, Miami

5:15pm- 5:40pm

4. Computational Modeling of Stem Cells and Exosomes Michael E. Davis, Emory University, Atlanta

4:00pm- 5:40pm

Quarter Deck Room Session 8: Cardiac Myofilament Proteins - Sponsored by JMCC Chair: Pieter DeTombe, Loyola University, Chicago Co-Chair: Jill Tardiff, University of Arizona, Tucson

4:00pm 1. DIASTOLIC DYSFUNCTION IN INHERITED AND ACQUIRED CARDIOMYOPATHIES Richard Moss, University of Wisconsin, Madison

2. TITIN AND CARDIOMYOPATHIES Henk Granzier, University of Arizona , Tucson 3. 4. REGULATION OF TROPONIN IN HEAT FAILURE Jian-Ping Jin, Wayne State University, Detroit

4:25pm

4:50pm

5:15pm-

5:40pm

5:45pm - 7:15pm

Chart Room Reception and Poster Session II. Posters 002-053

7:30pm - 9:30pm

Napoleon House ECI Social Event

36th Annual Conference of the

North American Section of ISHR

WEDNESDAY

31st May

Page 14: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

14

TIME ROOM & LOCATION ACTIVITY

7:30am Registration Desk Registration 8:00am- 9:00am

River/Port/Starboard Room

Peter Harris Award Lecture Chair: David Eisner, University of Manchester, UK Co-chair: Monte Willis, University of North Carolina, Chapel Hill

8:00am R. John Solaro, University of Illinois, Chicago “SARCOMERES AS HUBS OF SIGNALING IN THE HEART”

9:00am- 10:40am

River/Port/Starboard Room

Session 9: Proteotoxicity and Cardiovascular Disease Chair: Johannes Backs, Heidelberg University Hospital, Germany Co-Chair: Suresh Palaniyandi, Henry Ford Hospital, Detroit

9:00am 1. PROTEIN AGGREGATES AND AUTOPHAGY IN HEART FAILURE Jeffrey Robbins, Cincinnati Children’s Hospital Medical Center, Cincinnati

9:25am 2. AMYLOID CARDIOMYOPATHIES Ronglih Liao, Harvard University, Boston

9:50am 3. PROTEIN DEGRADATION IN HEART FAILURE X.J. Wang, University of South Dakota, Vermillion

10:15am- 10:40am

4. PTMS AND CARDIOVASCULAR DISEASE Jenny Van Eyk, Cedars-Sinai, Los Angeles

9:00am- 10:40am

Quarter Deck Room Session 10: Calcium Regulation in Heart Failure Chair: Salvatore Pepe, Murdoch Children’s Research Institute, Australia Co-chair: Don Bers, University of California, Davis

9:00am 1. CAMKII SIGNALING Mark Anderson, Johns Hopkins University, Baltimore 2. MICROPEPTIDE CONTROL OF CALCIUM SIGNALING Douglas M. Anderson, University of Rochester Medical Center, Rochester 3. DYNAMIC MITOCHONDRIAL CA2+ AND ROS SIGNALING Shey-Shing Sheu , Thomas Jefferson University, Philadelphia 4. MOLECULAR REGULATION OF MITOCHONDRIAL CA2+ IN DISEASE John Elrod, Temple University, Philadelphia

9:25am

9:50am

10:15am- 10:40am

36th Annual Conference of the

North American Section of ISHR

THURSDAY

1st June

Page 15: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

15

TIME ROOM & LOCATION ACTIVITY

10:40am- 11:00am

Foyer Coffee Break

11:00am- 12:40pm

River/Port/Starboard Room

Session 11: Epigenetic Regulation in Cardiovascular Disease Chair: Thomas Vondriska, University of California, Los Angeles Co-chair: Susan Howlett, Dalhousie University, Canada

11:00am 1. CHROMATIN BIOLOGY IN CARDIAC STRESS Sarah Franklin, University of Utah, Salt Lake City

11:25am 2. GENOME WIDE MAPPING OF CARDIAC AND FIBROBLAST ENHANCERS Izhak Kehat, Technion - Israel Institute of Technology, Israel, Haifa

11:50am 3. HEMATOPOIETIC CELL REGULATION OF CARDIOVASCULAR DISEASE Kenneth Walsh, Boston University, Boston

12:15pm- 12:40pm

4. NOVEL CARDIOPROTECTIVE SIGNALS Kika Sucharov, University of Colorado, Denver

11:00am- 12:40pm

Quarter Deck Room Session 12: Myocardial Protection Chair: Dan Kapusta, Louisiana State University Health Sciences Center , New Orleans Co– chair: Kurt Varner, Louisiana State University Health Sci- ences Center, New Orleans

11:00am 1. AMPK MEDIATED CARDIOPROTECTION Lawrence Young, Yale School of Medicine, New Haven

11:25am 2. REMOTE ISCHEMIC POSTCONDITIONING Karin Przyklenk, Wayne State University, Detroit

11:50am 3. CAVEOLIN REGULATION OF METABOLISM: IMPLICA- TIONS FOR CARDIAC STRESS ADAPTATION IN MULTIPLE PATHOLOGIES Hemal Patel, University of California, San Diego

12:15pm- 12:40pm

4. MICRO RNAS, MITOCHONDRIA, AND MYOCARDIAL SALVAGE Charles Steenbergen, John Hopkins University, Baltimore

12:00pm- 1:30pm

Compass Room ISHR-American Section: Council Meeting #2

36th Annual Conference of the

North American Section of ISHR

THURSDAY

1st June

Page 16: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

16

TIME ROOM & LOCATION ACTIVITY

12:40pm Foyer Lunch -2:00pm 2:00pm-

3:40pm

River/Port/Starboard Room

Session 13: Heart Failure with Preserved Ejection Fraction - Sponsored by JMCC Chair: Jeff Madwed, MT-Pharma-US, New Jersey Co-chair: Thomas E. Sharp, Temple University, Philadelphia

2:00pm 1. WHAT IS HFpEF AND WHY DO WE CARE? Stephen R. Houser, Temple University, Philadelphia

2. HFpEF ANIMAL MODELS: DESIGNING AND PERFORM- ING STUDIES LIKE CLINICAL TRIALS Kersten Small, MT-Pharma-US, New Jersey

3. PDE SIGNALING IN HFpEF David A. Kass, Johns Hopkins University, Baltimore

4. VASCULAR RAREFACTION IN HFPEF Frank W. Smart, Louisiana State University Health Sciences Center, New Orleans

2:25pm

2:50pm

3:15pm- 3:40pm

2:00pm- 3:40pm

Quarter Deck Room Session 14: Hypertrophy and Cardiomyopathy Chair: Chen Gao, University of California, Los Angeles Co-chair: Yoshihiko Saito, Nara Medical University, Japan

2:00pm 1. MALADAPTIVE SIGNALING IN CARDIAC HYPERTROPHY Jun Sadoshima, Rutgers New Jersey Medical School, Newark

2:25pm 2. COMMON CARDIOMYOPATHY SIGNALING PATHWAYS Jeffrey A. Towbin, Le Bonheur Children’s Hospital, Memphis

2:50pm 3. SARCOMERES AND CARDIAC HYPERTROPHY Leslie A. Leinwand, University of Colorado, Boulder

3:15pm- 3:40pm

4. HYPERTROPHIC CARDIOMYOPATHY MUTATIONS IN IPSCS J. Carter Ralphe, University of Wisconsin Health, Madison

3:40pm- 4:00pm

Foyer Coffee Break

4:00pm- 5:40pm

River/Port/Starboard Room

Session 15: Non-Myocytes in Heart Failure Chair: Jason Gardner, Louisiana State University Health Sci- ences Center, New Orleans Co-chair: Lisandra de Castro Brás, Brody School of Medi- cine, Greenville

36th Annual Conference of the

North American Section of ISHR

THURSDAY

1st June

Page 17: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

17

TIME ROOM & LOCATION ACTIVITY

4:00pm 1. MYOFIBROBLASTS IN CARDIAC DISEASE Jennifer Davis, University of Washington, Seattle

4:25pm 2. NOVEL STRATEGIES TO LIMIT FIBROBLAST ACTIVATION Timothy McKinsey, University of Colorado, Denver

4:50pm 3. THERAPEUTIC TARGETING OF ORGAN FIBROSIS Burns Blaxall, Cincinnati Children’s Hospital Medical Center, Cincinnati

5:15pm- 5:40pm

4. POLYUNSATURATED FATTY ACIDS AND FIBROSIS Tim O’Connell, University of Minnesota, Minneapolis

4:00pm- 5:40pm

Quarter Deck Room Session 16: Cardiac Gene Therapy Chair: Michael Kapiloff, University of Miami Health System, Miami Co-chair: Md Abdur Razzaque, Louisiana State University Health Science Center, New Orleans,

4:00pm 1. SERCA2 GENE THERAPY Roger Hajjar, Icahn School of Medicine at Mount Sinai, New York

2. AC6 GENE TRANSFER FOR HEART FAILURE H. Kirk Hammond, University of California, San Diego, San Diego

4:25pm

4:50pm 3. GENETIC CORRECTION STRATEGIES FOR MYOPATHIES Elizabeth McNally, Northwestern University, Chicago

4.

5:15pm- 5:40pm 6:00pm Hilton Riverside Front

Entrance Bus transportation to Banquet at Champion Square locat- ed at the Mercedes - Benz Superdome

6:30pm - 8:30pm

Mercedes - Benz Superdome, Champion Square Encore Room

Dinner Reception and Awards Banquet

8:30pm - 11:30pm

Club XLIV Social Celebration

11:30pm Back Entrance of Encore Room

Bus transportation returning to Hilton Riverside Hotel

36th Annual Conference of the

North American Section of ISHR

THURSDAY

1st June

Page 18: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

18

TIME

7:30am 8:00am- 9:00am

8:00am

9:00am- 10:40am

9:00am

9:25am

9:50am

10:15am- 10:40am

9:00am- 10:40am

9:00am

9:25am

ACTIVITY

Registration Plenary Lecture - Pfeiffer Distinguished Lecture Chair: Richard Vander Heide, Louisiana State University Health Sciences Center, New Orleans Co-chair: Martin Vila - Petroff, University of Melbourne, Australia Tesuji Miura, Sapporo Medical University School of Medicine, Japan “DIABETIC CARDIOMOPATHY -- ADAPATATION AND MALA-DAPTATION OF PRO-SURVIVAL AND METABOLISM” Session 17: Cardiac Metabolism Chair: Jianyi Zhang, University of Alabama, Birmingham Co- chair: E. Douglas Lewandowski, Sanford Burnham Prebys Medical Discovery Institute, Orlando 1. LIPOTOXICITY AND MITOCHONDRIAL DYNAMICS E. Dale Abel, University of Iowa, Carver College of Medicine Iowa City 2. CARDIAC TOXICITY OF DUAL PPARα/γ ACTIVATION Konstantinos Drosatos, Temple University, Philadelphia 3. METABOLIC REGULATION OF CARDIAC GROWTH Bradford Hill, University of Louisville, Louisville 4. ROLE OF MG53 IN CARDIAC METABOLIC DISEASE Rui-Ping Xiao, Peking University, Institute of Molecular Medicine, China Session 18: Non-coding and Extracellular RNAs Chair: Catherine Makarewich, University of Texas Southwest- ern Medical Center, Dallas Co-chair: Gangjian Qin, University of Alabama, Birmingham

1. REGULATION OF RNAS BY IL-10 Raj Kishore, Temple University, Philadelphia 2. IMPACT OF THE TRANSCRIPTION FACTOR GATA2 IN ENDOTHELIAL CELLS ON CARDIAC FAILURE THROUGH REGULATION OF TWO SECRETED LONG NON-CODING RNAS Joerg Heineke, Hannover Medical School, Germany

36th Annual Conference of the North American Section of ISHR

FRIDAY

2nd June

ROOM & LOCATION Registration Desk River/Port/Starboard Room

River/Port/Starboard Room

Quarter Deck Room

Page 19: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

19

TIME ROOM & LOCATION ACTIVITY

9:40am 3. LONG NONCODING RNAS REGULATE ENERGY METAB- OLISM IN MICE AND HUMANS Haiming Cao, NHLBI, National Institutes of Health

10:15am- 10:40am

4. THERAPEUTIC POTENTIAL OF NON-CODING RNAS IN HEART FAILURE Rusty Montgomery, miRagen Therapeutics, Boulder

10:40am- 11:00am

Foyer Coffee Break.

11:00am- 12:40pm

River/Port/Starboard Room

Session 19: Cardiac Signaling Pathways Chair: Randi Parks, NIH National Heart, Lung, and Blood Insti- tute, Maryland Co-chair: Edward Glasscock, Louisiana State University Health Science Center, Shreveport

11:00am 1. POST-TRANSCRIPTIONAL REGUALTION OF CARDIAC INFLAMMATION Federica Accornero, Ohio State University, Columbus

11:25am 2. NITRIC OXIDE SIGNALING PATHWAYS Sruti Shiva, University of Pittsburgh, Pittsburgh

11:50am 3. EXOSOME MEDIATED CARDIOPROTECTION Geoffrey De Couto, Cedars-Sinai, Los Angeles

12:15pm- 12:40pm

4. MITOCHONDRIAL SIGNALING AND CELL SURVIVAL Brian O’Rourke, John Hopkins School of Medicine, Baltimore

11:00am- 12:40pm

Quarter Deck Room Session 20: Aging and Heart Failure Chair: Mohsin Khan, Temple University, Philadelphia Co-chair: Emma Monte, Stanford School of Medicine, Stan- ford

11:00am 1. DYSTROPHIN AND AGE-RELATED CARDIAC FAILURE Joseph Metzger, University of Minnesota, Minneapolis

11:25am 2. AGING OF THE HEART Marcello Rota, New York Medical College, Valhalla

11:50am 3. CARDIAC PROGENITOR CELL THERAPY AND AGING Annarosa Leri, Swiss Institute for Regenerative Medicine, Lu- gano

12:15pm- 12:40pm

4. MITOCHONDRIAL DYSFUNCTION IN THE AGED HEART Edward Lesnefsky, Virginia Commonwealth University Medical Center, Richmond

FRIDAY

2nd June

36th Annual Conference of the North American Section of ISHR

Page 20: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

20

TIME ROOM & LOCATION ACTIVITY

12:40pm - 1:40pm

River/Port/Starboard Room

Outstanding Investigator Award Chair: Litsa Kranias, University of Cincinnati College of Medi- cine, Cincinnati Co-chair: Elizabeth Murphy, NIH/NHLBI, Bethesda

12:40pm Xander Wehrens, Baylor College of Medicine, Houston “CALCIUM RELEASE UNIT DEFECTS- SOURCE OF MANY CARDIAC EVILS?”

1:40pm Meeting Adjournment.

FRIDAY

2nd June

36th Annual Conference of the North American Section of ISHR

Page 21: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

21

POSTER SESSIONS

Posters are located in the Chart Room.

Poster Session I Wednesday, May 31 12:30-2:00 PM Posters P054 to P105

Poster Session II Wednesday, May 31 5:30-7:00 PM Posters P002 to P053

Presenting author underlined

P# POSTER SESSION I: Wednesday, May 31 12:30-2:00 PM

54 Regional cardiac denervation produces supersensitivity of myocardial Ca2+ handling to β- adrenergic stimulation Srinivas Tapa1, Lianguo Wang1, Samantha Francis Stuart1, Crystal Ripplinger1, 1University of

California, Davis, Davis, CA, USA

55 GRK2-S670A Mice reveal cardioprotection post ischemia-reperfusion Priscila Sato1, J Kurt Chuprun1, Laurel Grisanti1, Meryl Woodall1, CJ Traynham1, Anna Maria Lucchese1, Ancai Yuan1, Jessica Ibetti1, Doug Tilley1, Erhe Gao1, Walter Koch1, 1Temple University School of Medicine, Philadelphia, PA, USA

56 Model establishment for cardiovascular evaluation using the novel Stellar TSE’s Type

PPBTA-XL Telemetry Transmitter in the Vervet (St. Kitts green monkey) Shervin Liddie1, Aryamitra Banerjee1, David Moddrelle1, Xavier Morton1, Matthew Lawrence1, 1RxGen Inc, New Haven, CT, USA

57 Dose-dependent effect of Hyperglycemia during Cardiac Development Amelia Cephas1,2, Madhumita Basu2, Vidu Garg1,2, 1The Ohio State University, Columbus, OH, USA, 2Nationwide Children's Hospital, Columbus, OH, USA

58 Radiofrequency Renal Denervation Decreases Renal Fibrosis in Spontaneously Hypertensive Rats (SHR) Juan Gao1, Ian Denys1, Liang Xiao2, David Polhemus1, Frank Smart1, Traci Goodchild1, David Lefer1, David Harrison2, Daniel Kapusta1, 1LSUHSC, New Orleans, LA, USA, 2Vanderbilt University, Nashville, TN, USA

59 Renal Sympathetic Denervation Reverses Diastolic Dysfunction in a Rodent Model of Heart

Failure with Preserved Ejection Fraction (HFpEF) Hiroshi Koiwaya1, David Polhemus1, Rishi Trivedi1, David Lefer1, 1Cardiovascular Center of

Excellence, Louisiana State University Health Science Center, New Orleans, LA, USA

60 High-Throughput Screen Identifies Novel Small Molecule Stress Regulator That Confers Cardioprotection During Ischemia-Reperfusion Injury Erik Blackwood1, Lars Plate2, Ryan Paxman2, Kyle Malter1, Luke Wiseman2, Jeff Kelly2, 1San Diego State University Heart Institute and Department of Biology, San Diego, CA, USA, 2Departments of Chemistry, Molecular and Experimental Medicine, Chemical Physiology - The Scripps Research Institute, La Jolla, CA, USA

Page 22: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

22

61 Multi-omics approach to identify disease signatures in cardiac remodeling Edward Lau1,2, Maggie P.Y. Lam1, Peipei Ping1, 1University of California, Los Angeles, Los Angeles, CA, USA, 2Stanford University, Palo Alto, CA, USA

62 Readmission rates after Acute Decompensated Heart Failure Waqas Siddiqui1, Andrew Kohut1, Syed Hasni1, Jesse Goldman1, Benjamin Silverman1, Ellie Kelepouris1, Howard Eisen1, Sandeep Aggarwal1, 1Drexel University College of Medicine, Philadelphia, PA, USA, 2Hahnemann University Hospital, Philadelphia, PA, USA

63 Pacemaker syndrome; an often overlooked diagnosis in patients with pacemakers Awais Arif1, Rizwan Khan1, Nicole Tran1, 1University Of Oklahoma, Oklahoma, USA

64 Tuberculous aortitis, an unusual presentation of tuberculosis Rizwan Khan1, Awais Arif1, Nicole Tran1, 1University Of Oklahoma, Oklahoma, USA

65 Recurrent episodes of loss of consciousness; autoimmune autonomic ganglionopathy with features of postural orthostatic tachycardia syndrome treated with plasmapheresis: a prospective case study Rizwan Khan1, Murtaza Mazhar1, Christian Kaufman1, 1University Of Oklahoma, Oklahoma City,

Oklahoma, USA

66 Glucose Promotes Cell Growth by Suppressing Branched-chain Amino Acid Degradation Dan Shao1, Outi Villet1, Zhen Zhang1, Sung Won Choi1, Jie Yan2, Haiwei Gu1, Danijel Djukovic1, Danos Christodoulou2, Julia Ritterhoff1, Stephen C Kolwicz Jr1, Daniel Raftery1, Rong Tian1, 1University of Washington, Seattle, WA, USA, 2Brigham and Women’s Hospital, Boston, MA, USA

67 The Sphingosine-1-Phosphate Receptor Modulator, FTY720, Reverses Diastolic Dysfunction

and Hypertrophy in Hypertrophic Cardiomyopathy David M. Ryba1, Chad M. Warren1, Chehade N. Karam1, Robert T. Davis, 3rd1, Shamim A. K.

Chowdhury1, Manuel G. Alvarez1, David F. Wieczorek2, R. John Solaro1, Beata M. Wolska1, 1Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA, 2Department of Molecular Genetics, Biochemistry, & Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA, 3Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, USA

68 Effect of moderate exercise training and continuous normobaric hypoxia on postinfarction

heart failure in rats Jaroslav Hrdlicka1, Jan Neckar1, Frantisek Papousek1, Jana Vasinova1, Petra Alanova1, Frantisek Kolar1, 1Institute of Physiology CAS, Prague, Czech Republic

69 Cardioprotective adaptation to chronic hypoxia stimulates the ROS-dependent/cytosolic phospholipase A2a pathway in rat heart Marketa Hlavackova1,2, Petra Micova2, Klara Hahnova2, Barbora Elsnicova2, Anna Chytilova1, Kristyna Holzerova1, Jiri Novotny2, Jitka Zurmanova2, Jan Neckar1, Olga Novakova1, Frantisek Kolar1, 1Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic, 2Department of Physiology, Faculty of Science, Charles University in Prague, Prague, Czech Republic

70 Renal Sympathetic Denervation Improves Left Ventricular Function and Vascular Reactivity

in Heart Failure David Polhemus1, Rishi Trivedi1, Zhen Li1, Hiroshi Koiwaya1, Traci Goodchild1, Juan Gao1, Daniel Kapusta1, David Lefer1, 1LSU Health Sciences Center, New Orleans, LA, USA

71 Cardiosphere-Derived Cells Combined with Renal Sympathetic Denervation Improves Ventricular Function and Remodeling Following Ischemic Injury David Polhemus1, Rishi Trivedi1, Traci Goodchild1, Geoffrey De Couto2, Eduardo Marban2, David

Page 23: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

23

Lefer1, 1LSU Health Sciences Center, New Orleans, LA, USA, 2Cedars Sinai Heart Institute, Los Angeles, CA, USA

72 A Novel Hydrogen Sulfide Donor, JK1, Protects the Heart Against Pressure Overload Induced Heart Failure A Novel Hydrogen Sulfide Donor, JK1, Protects the Heart Against Pressure Overload Induced Heart Failure Zhen Li1, Chelsea Organ1, David Polhemus1, Rishi Trivedi1, Jianming Kang2, Ming Xian2, David

Lefer1, 1LSU Health Sciences Center, New Orleans, USA, 2Washington State University, Pullman, USA

73 A Novel Hydrogen Sulfide Prodrug, SG-1002, Augments Angiogenesis and Coronary

Vascular Tone in a Swine Model of Critical Limb Ischemia Amanda Rushing1, Erminia Donnarumma1, Amy Scarborough1, Sarah Boisvert1, Rishi Trivedi1, David Polhemus1, Zhen Li1, Kevin Au2, Sam Victoria2, Jeffrey Schumacher3, David Lefer1, Traci Goodchild1, 1Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA, 2Department of Vascular Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA, 3Department of Animal Care, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA

74 A Novel Histone Deacetylase (HDAC) Inhibitor Attenuates Cardiac Dysfunction in the Setting

of Pressure Overload Heart Failure Chelsea Organ1, 2, Zhen Li1, 2, Craig Zibilich1, Traci Goodchild1,2, Shubing Wang3, Kersten M. Small4, Jeffrey Madwed4, Jian Liu4, Joseph Kozlowski4, David J. Lefer1, 2, 1Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA; 2Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, USA; 3Merck Research Labs, Rahway, NJ, USA; 4Merck Research Laboratories, Kenilworth, NJ, USA

75 Hypercholesterolemic LDLr Knockout Swine as a Clinically Relevant Model of Hypertension

Amanda Rushing1, Amy Scarborough1, James Stephen Jenkins2, John Reilly2, Seena Khosravi1, Rishi Trivedi1, David Polhemus1, Traci Goodchild1, David Lefer1, 1Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA, 2Ochsner Interventional Cardiology, Ochsner Medical Center, New Orleans, LA 70121, USA

76 Genetic inhibition of the UPR gene Chac1 preserves cardiac function in a murine model of pressure overload induced heart failure. Zhen Li1,2, Lisa O Nguyen1, Chelsea L Organ1,2, David J Lefer1,2, Imran N Mungrue1, 1Lsu Health- Dept Pharmacology, New Orleans, USA, 2LSU Health- Cardiovascular Center of Excellence, New Orleans, USA

77 Ischemic vs. Non-Ischemic Dilated Cardiomyopathy: a Comparative Study in Stem Cell

Therapy Efficacy Bryon A. Tompkins1, Angela C. Rieger1, Victoria Florea1, Makoto Natsumeda1, Evan D. Nigh1, Ana

Marie Landin1, Gianna M. Rodriguez1, Konstantinos E. Hatzistergos1, Ivonne H. Schulman1, Joshua M. Hare1, 1Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA

78 Regulation of cardiac excitation-contraction coupling by fibroblasts in health and disease

Brian Wang1, Cesare Terracciano1, Kenneth Macleod1, 1Imperial College London, London, UK

79 DWORF overexpression prevents heart failure in an experimental mouse model of dilated cardiomyopathy Cat Makarewich1,2, Svetlana Bezprozvannaya1,2, Rhonda Bassel-Duby1,2, Eric Olson1,2, 1Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA, 23Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, USA

Page 24: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

24

80 CaMKII oxidation causes increased atrial fibrillation in diabetic mice

Olurotimi Mesubi1, Adam Rokita2, Neha Abrol1, Yuejin Wu1, Biyi Chen2, Qinchuan Wang1, Jonathan Granger1, Elizabeth Luczak1, Lars Maier4, Xander Wehrens5, Joel Pomenrantz3, Long-Sheng Song2, Gerald Hart3, Mark Anderson1, 1Division of Cardiology, Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA, 2Division of Cardiovascular Medicine and Cardiovascular Research Center, Carver College of Medicine, Iowa City, IA, USA, 3Department of Biological Chemisty, The Johns Hopkins University School of Medicine, Baltimore, MD, USA, 4Division of Cardiology and Pneumology, German Heart Center, University Hospital Goettingen, Goettingen, Germany, USA, 5Department of Molecular Physiology & Biophysics and Medicine (Cardiology), Baylor College of Medicine, Houston, TX, USA

81 LCZ696, the First-in-Class Angiotensin Receptor Neprilysin Inhibitor, Improves Vascular

Reactivity in the Setting of Heart Failure Rishi K. Trivedi1, Zhen Li1, David J. Polhemus1, Daniel Yoo1, Hiroshi Koiwaya1, Traci T. Goodchild1,

David J. Lefer1, 1LSU Health Sciences Center - New Orleans, New Orleans, LA, USA

82 LCZ696 Reduces Myocardial Fibrosis in Hypertensive Rats in the Setting of Heart Failure Rishi K. Trivedi1, Zhen Li1, David J. Polhemus1, Daniel Yoo1, Hiroshi Koiwaya1, Traci T. Goodchild1, David J. Lefer1, 1LSU Health Sciences Center, New Orleans, LA, USA

83 Reversible expression of cardiac MyBP-C using an inducible tet-off system Jasmine Giles1, Adam Miller1, Daniel Fitzsimons1, Richard Moss1, 1University of Wisconsin, Madison, WI, USA

84 The hypertrophic cardiomyopathy-causing W792R and T1075 mutations in cardiac myosin

binding protein-C generate cardiac dysfunction in mice Thomas Lynch IV1, Jasmine Giles1, Elizabeth Iverson1, Daniel Fitzsimons1, Richard

Moss1, 1University of Wisconsin, Madison, WI, USA

85 Phosphorylation of cMyBP-C accelerates the rate of force relaxation in murine skinned myocardium Jitandrakumar Patel1, Daniel Fitzsimons1, Richard Moss1, 1University of Wisconsin, Madison, WI,

USA

86 Ablation of cMyBP-C eliminates the activation-dependence of unloaded shortening velocity at low levels of Ca2+ activation Daniel Fitzsimons1, Jitandrakumar Patel1, Richard Moss1, 1University of Wisconsin, Madison, WI, USA

87 A missense mutation within the C6-domain of cMyBP-C results in cardiac enlargement and

depressed ventricular function Jitandrakumar Patel1, Jasmine Giles1, Adam Miller1, Daniel Fitzsimons1, Richard Moss1, 1University of Wisconsin, Madison, WI, USA

88 The mAKAP complex orchestrates the dephosphorylation of MEF2D in muscle cells to

stimulate its activity Shania Aponte París1, Michael Kapiloff1, Kimberly Dodge-Kafka1, 1University of Connecticut,

Farmington, CT, USA

89 Integrated omics analysis of isoform switching under cardiac hypertrophy Maggie Lam1, Peipei Ping1, Yi Xing1, 1UCLA, Los Angeles, USA

90 MANF, a structurally unique redox-sensitive chaperone, restores ER-protein folding in the ischemic heart. Adrian Arrieta1, Erik Blackwood1, Winston T. Stauffer1, Michelle Santo Domingo1, Amber N.

Page 25: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

25

Pentoney1, Donna J. Thuerauf1, Shirin Doroudgar2,3, Christopher C. Glembotski1, 1San Diego State University, San Diego, CA, USA, 2Department of Cardiology, Angiology, and Pneumology, University Hospital Heidelberg, Innere Medizin III, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany, 3DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany

91 Loss of caveolin-1 alters cardiac mitochondrial function and increases susceptibility to stress Jan M Schilling1,2, Mehul Dhanani1,2, Kristofer J Haushalter1,2, Sarah A Howell1,2, Ravina Verma1,2, Ingrid R Niesman1, Alice E Zemljic-Harpf1,2, Hemal H Patel1,2, 1University of California, San Diego, CA, USA, 2VA San Diego Healthcare System, San Diego, CA, USA

92 Top-down proteomics for assessing the maturation of stem cell-derived cardiomyocytes

Wenxuan Cai1, Jianhua Zhang1, William J. de Lange1, Zachery Gregorich1, J. Carter Ralphe1, Timothy Kamp1, Ying Ge1, 1University of Wisconsin-Madison, Madison, WI, USA

93 Regulation of ryanodine receptor mediated perinuclear calcium by the mAKAP complex Moriah Gildart1, Michael Kapiloff2, Kimberly Dodge-Kafka1, 1University of Connecticut Health Center, Farmington, Connecticut, USA, 2University of Miami Miller School of Medicine, Miami, Florida, USA

94 Cardiac muscle function across the natural history of a genetic minipig model of

hypertrophic cardiomyopathy Marcus Henze1, Robert Anderson1, Fiona Wong1, Robert Weiss2, Abhay Divekar2, David Meyerholz2, Ferhaan Ahmad2, Eric Green0, 1MyoKardia, Inc., South San Francisco, CA, USA, 2University of Iowa, Iowa City, IA, USA

95 The muscle-specific ubiquitin ligase Atrogin-1 (MAFbx) inhibits age-associated cardiac fibrosis by enhancing MMP-9 levels in vivo Traci Parry1, Roberto Mota1, Monte Willis1, 1Univ of North Carolina, Univ of North Carolina, USA

96 Curation and Phenotyping of Cardiovascular Case Reports Achieved by ICD Based Index System and MeSH Supported Query Platform Yijiang Zhou1,2, David Liem1,2, Quan Cao1,2, Jessica Lee1,2, Wei Wang1,3, Alex Bui1,4, Karol

Watson1,2, Jiawei Han5, Peipei Ping1,2, 1The NIH BD2K Center of Excellence at UCLA, Los Angeles, California, USA, 2Departments of Physiology, Medicine/Cardiology, Bioinformatics, University of California at Los Angeles, Los Angeles, California, USA, 3Departments of Computer Science, University of California at Los Angeles, Los Angeles, California, USA, 4Departments of Radiology, University of California at Los Angeles, Los Angeles, California, USA, 5NIH BD2K KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA

97 Construction a Standardized Metadata Template to Extract Relevant Biomedical Insights from

Clinical Case Reports Yijiang Zhou1, David A. Liem1, Quan Cao1, Jessica Lee1, Wei Wang2, Alex Bui3, Karol Watson4, Jiawei Han5, Peipei Ping6, 1The NIH BD2K Center of Excellence at UCLA, Departments of Physiology, Los Angeles, California, USA, 21The NIH BD2K Center of Excellence at UCLA, Department of Computer Science, University of California at Los Angeles, CA 90095, USA, Los Angeles, California, USA, 31The NIH BD2K Center of Excellence at UCLA, Departments of Radiology, University of California at Los Angeles, CA 90095, USA, Los Angeles, California, USA, 41The NIH BD2K Center of Excellence at UCLA, Departments of Medicine/Cardiology, University of California at Los Angeles, CA 90095, USA, Los Angeles, California, USA, 5NIH BD2K KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA, Urbana-Champaign, Illinois, USA, 61The NIH BD2K Center of Excellence at UCLA, Departments of Physiology, Medicine/Cardiology, Bioinformatics, Computer Science, and Radiology, University of California at Los Angeles, CA 90095, USA, Los Angeles, California, USA

98 Identification of cardiomyocytes' characteristics responsible for dynamical changes in

calcium profile in response to mechano-chemo transduction

Page 26: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

26

Zana Coulibaly1, Rafael Shimkunas1, Bence Hegyi1, Zhong Jian1, Ye Chen-Izu1, Leighton Izu1, 1University of California, Davis, Davis, California, USA

100 Aortic acceleration as noninvasive index of left ventricular contractility

Anilkumar Reddy1,2, Celia Pena Heredia1, Thuy Pham1, George Taffet1, 1Baylor College of Medicine, Houston, Texas, USA, 2Indus Instruments, Webster, Texas, USA

101 Phrase Mining and Machine Learning in Textual Data to Uncover Distinct Protein Patterns in Cardiovascular Disease David A. Liem1, Vincent Kyi1, Yu Shi2, Fangbo Tao2, Jiawei Han2, Peipei Ping1, 1NIH BD2K Center

of Excellence at UCLA, Departments of Physiology, Medicine and Bioinformatics, UCLA School of Medicine, Los Angeles, CA 90095, USA, Los Angeles, California, USA, 2NIH BD2K KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA, Urbana-Champaign, Illinois, USA

102 Temporal Dynamics of Plasma Metabolites in ISO-induced Cardiac Remodeling in Mice

Quan Cao1,2, Howard Choi1,2, Ding Wang1,2, David Liem1,2, Chelsea Ju1,3, Jennifer Polson1,2, Wei Wang1,3, Peipei Ping1,2, 1NIH BD2K Center of Excellence at University of California, Los Angeles, Los Angeles, California, USA, 2Departments of Physiology, Medicine and Bioinformatics, UCLA, Los Angeles, California, USA, 3Departments of Computer Science, UCLA, Los Angeles, California, USA

103 The spatial distribution of the Na+/Ca2+ exchanger in cardiac mitochondria enhances the efficiency of mitochondrial Ca2+ signal generation Sergio De La Fuente1, Celia Fernandez-Sanz1, Jonathan Lambert2, John Elrod2, Shey-Shing Sheu1, Gyorgy Csordas1, 1Thomas Jefferson University, Philadelphia, PA, USA, 2Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA

104 Autologous bone marrow stem cell therapy in patients with ST-elevation myocardial

infarction: a systematic review and meta-analysis Maya M. Jeyaraman1,10, Rasheda Rabbani1,10, Leslie Copstein1, Wasan Sulaiman1, Farnaz

Farshidfar1, Hessam Kashani1, Sheikh M.Z. Qadar1, Qingdong Guan2,3, Becky Skidmore4, Elissavet Kardami5, John Ducas6, Samer Mansour7,8, Ryan Zarychanski1,9, Ahmed M. Abou- Setta1,10, 1George & Fay Yee Center for Healthcare Innovation, Winnipeg, Manitoba, Canada, 2Manitoba Center for Advanced Cell and Tissue Therapy, Winnipeg, Manitoba, Canada, 3Cellular Therapy Laboratory, CancerCare Manitoba, Winnipeg, Manitoba, Canada, 4Information Specialist Consultant, Ottawa, Ontario, Canada, 5Department of Human Anatomy and Cell Sciences, University of Manitoba, Winnipeg, Manitoba, Canada, 6Section of Cardiology, Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada, 7Centre Hospitalier de l’Université de Montreal, Montreal, Quebec, Canada, 8Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada, 9Department of Haematology and Medical Oncology, Cancer Care Manitoba, Winnipeg, Manitoba, Canada, 10Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada

105 Fractionation of embryonic cardiac progenitor cells and evaluation of their differentiation

potential Tiam Feridooni1, Kishore Pasumarthi1, 1Dalhousie University, Halifax, Nova Scotia, Canada

Page 27: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

27

P# POSTER SESSION II: Wednesday, May 31 5:30-7:00 PM

2 Chronic Inotrope Infusion: A Viable Option for Low Output Heart Failure Rizwan Khan1, Murtaza Mazhar1, Ammar Tahir2, Alya Ahsan2, Omer Iftikhar2, David Najman2, 1University of Oklahoma, Oklahoma City, OK, USA, 2The University of Chicago (NorthShore University HealthSystem), Evanston, IL, USA

3 CaMKII as a pathological mediator of inflammation, oxidative stress, ER stress, autophagy

and mitochondrial dysfunction in free fatty acids/hyperlipidemia-induced cardiac remodeling both in vitro and vivo Peng Zhong1,3, He Huang1,2, 1Department of Cardiology, Renming Hospital of Wuhan University,

Wuhan, China, 2Cardiovascular Research Institute, Wuhan University, Wuhan, China, 3Department of Cardiology, Johns Hopkins Medical Institute, USA

4 INAPPROPRIATE SINUS TACHYCARDIA – SYMPTOM AND HEART RATE REDUCTION WITH

IVABRADINE: POOLED ANALYSIS OF PROSPECTIVE STUDIES ST Mathew1, SS Po1,2, U Thadani1, 1Veterans Affairs Medical Center/University of Oklahoma,

Oklahoma City, OK, USA, 2Heart Rhythm Institute/University of Oklahoma, Oklahoma City, OK, USA

5 Study of Some Coronary Atherosclerotic Biomarkers Garmaa Nandin1, 1Mongolian National University of Medical Sciences., Ulaanbaatar, Mongolia

6 Decreased Aldehyde Dehydrogenase (ALDH)2 Activity Contributes to Coronary Endothelial Dysfunction in Diabetic Cardiomyopathy Guodong Pan1, Mandar Deshpande1, Suresh S. Palaniyandi1, 1Henry Ford Health System, Detroit, MI, USA

7 Dual interplay between desmin phosphorylation and dysregulated autophagy in heart failure

Marion Bouvet1, Emiliie Dubois-Deruy1, Paul Mulder2, Maggy Chwastyniak1, Arthur Dechaumes1, Olivia Beseme1, Philippe Amouyel1, Nicolas Lamblin1, Vincent Richard2, Florence Pinet1, 1Inserm U1167, Lille, France, 2Inserm U1096, Rouen, France

8 Cardioprotective biology of S-nitrosylated Hypoxia-inducible Factor 1α Kevin Casin1, Mark Kohr1,2, 1Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA, 2Johns Hopkins School of Medicine, Baltimore, MD, USA

9 Deficiency of miR-1954 promotes cardiac remodeling Sudhiranjan Gupta1, Ana Takano1, Rakeshwar Guleria1, 1Texas A&M University, Temple, TX- 76504, USA

10 NF-kB-miR-23a-miR-27a: a critical modulator in post myocardial infarction remodeling

Sudhiranjan Gupta1, Li Li1, 1Texas A&M University, Temple, TX, USA

11 Mitochondrial superoxide in myocardial lysophosphatidic acid signaling Manikandan Panchatcharam1, Mini Chandra1, Diana Escalante-Alcalde2, Wayne Orr1, Christopher Kevil1, Shenuarin Bhuiyan1, Joseph Wu3, Sumitra Miriyala1, 1Louisiana State University Health Sciences Center, Shreveport, LA, USA, 2Universidad Nacional Autónoma de México, México DF, Mexico, Mexico, 3Stanford Cardiovascular Institute, Stanford, CA, USA

12 Abcg2-Expressing cells fuse with existing cardiomyocytes. Amritha Yellamilli1, Yi Ren1, Ron McElmurry1, Jop van Berlo1, 1Universsity of Minnesota, Minneapolis, Minnesota, USA

13 Cardiac hypertrophy suppresses glucose oxidation in newborns with congenital heart

defects Sonia Rawat1, Arata Fukushima1,2, Liyan Zhang1, Alda Huqi1,3, Tariq Altamimi1, Cory Wagg1, Lisa

Page 28: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

28

Hornberger1, Paul Kantor1, Ivan Rebeyka1, Gary Lopaschuk1, 1Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada, 2Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan, 3Cardio Thoracic and Vascular Department, University of Pisa, Pisa, Tuscany, Italy

14 Cardiac Progenitor Cell Lineage Tracing During Embryonic Cardiomyogenesis

Bingyan Wang1, Alvin Muliono1, Roberto Alvarez Jr.1, Mark Sussman1, 1San Diego State University, San Diego, CA, USA

15 Takotsubo Cardiomyopathy-Associated Ventricular Standstill in a Peripartum Patient

Nelson Lee1,2, Kevin Lee1,2, Joseph Banta1,2, Matthew D'Ambrosio1,2, Benyamin Hannallah2, Michael Benz2, Apostolos Voudouris2, 1Rowan University School of Osteopathic Medicine, Stratford, NJ, USA, 2Christ Hospital, Jersey City, NJ, USA

16 P2Y14 nucleotide receptor overexpression: Letting blind cardiac progenitor cells 'see' again

Farid Khalafalla1, Waqas Kayani1, Arwa Kassab1, Kelli Ilves1, Roberto Alvarez1, Monica Chavarria1, Benjamin Norman1, Mark Sussman1, 1San Diego State University, San Diego, California, USA

17 Membrane delimited estrogen receptor activation protects heart against ischemic- reperfusion injury in mice with cardiac but not endothelial specific ablation of ERα Junhui Sun1, Sara Menazza1,6, Swathi Appachi1, Ken Chambliss5, Sung-Hoon Kim2, Angel Aponte1, Sohaib Khan3, John Katzenellenbogen2, Benita Katzenellenbogen4, Philip Shaul5, Elizabeth Murphy1, 1Systems Biology Center, NHLBI/NIH, Bethesda, MD, USA, 2Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA, 3Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA, 4University of Cincinnati Cancer Center, Cincinnati, OH, USA, 5Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA, 6Department of Biomedical Sciences, University of Padova, Padova, Italy

18 Cardioprotection in mice with a knock- in mutation in Cyclophilin D (CypD- C202S): a site of S- nitrosylation Georgios Amanakis1, Junhui Sun1, Jennifer Boylston1, Elizabeth Murphy1, 1NHLBI/NIH, Bethesda,

MD, USA

19 Generation of MnSOD cardiomyocyte-specific knockout: Role in Heart failure development and progression Sumitra Miriyala1, Mini Chandra1, Wayne Orr1, Christopher Kevil1, Shenuarin Bhuiyan1, Joseph Wu2, Manikandan Panchatcharam1, 1Louisiana State University Health Sciences Center, Shreveport, LA, USA, 2Stanford Cardiovascular Institute, Stanford, CA, USA

20 Pediatric dilated cardiomyopathy hearts display a gene expression profile consistent with pluripotency and dedifferentiation Kathleen C. Woulfe1, Phillip D. Tatman1, Anis Karimpour-Fard1, Danielle A. Jeffrey1, Karin Nunley1, Matthew R. G Taylor1, Shelley D. Miyamoto1, Brian L. Stauffer1,2, Carmen C. Sucharov1, 1University of Colorado School of Medicine, Aurora, CO, USA, 2Denver Health and Hospital Authority, Denver, CO, USA

21 Cardioprotection in the mouse heart: acute protective effects of an estrogen receptor agonist

Anjali Ghimire1, Susan Howlett1, 1Dalhousie University, Halifax, Canada

22 The W792R mutation in cardiac myosin binding protein-C reduces the C6 FnIII domain stability and causes hypertrophic cardiomyopathy through haploinsufficiency Dan Smelter1, Willem de Lange1, J. Carter Ralphe1, 1University of Wisconsin, Madison, WI, USA

23 A role for PPARα in sex differences in cardiac hypertrophy Natasha Fillmore1, Josephine Harrington1, Shouguo Gao1, Yanqin Yang1, Xue Zhang1, Poching Liu1, Andrea Stoehr1, Danielle Springer1, Jun Zhu1, Xujing Wang1, Elizabeth Murphy1, 1National Heart

Page 29: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

29

Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA

24 Deletion of the Z-disc Protein Enigma Homolog Depresses Cross-bridge Cycling Kinetics in Mouse Myocardium Zachery Gregorich1, Jitandra Patel1, Wenxuan Cai1, Rachel Heurer1, Ziqing Lin1, Richard Moss1, Ying Ge1, 1University of Wisconsin-Madison, Madison, WI 53705, USA

25 Postconditioning with H2S donors: effect on reperfusion-induced ventricular arrhythmias Qutuba G Karwi1,2, Matt Whiteman3, Mark Wood4, Gary Baxter1, 1School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK, 2College of Medicine, University of Diyala, Diyala, Iraq, 3Medical School, University of Exeter, Exeter, UK, 4School of Biosciences, University of Exeter, Exeter, UK

26 CypD-mediated regulation of the permeability transition pore is altered in mice lacking the

mitochondrial calcium uniporter Randi Parks1, Sara Menazza1, Angel Aponte1, Paolo Bernardi2, Toren Finkel1, Elizabeth Murphy1, 1NHLBI, NIH, Bethesda, MD, USA, 2University of Padova, Padova, PD, Italy

27 Blocking/Knocking out Smad3 Alleviates Doxorubicin Effects on Endothelial Cells Jill Schriewer1, Melissa Cobb1, Meera Raghavan1, Eugene Konorev1, 1Kansas City University, Kansas City, MO, USA

28 The contribution of fatty acid and ketone body oxidation to energy production increases in

the failing heart and is associated with a decrease in cardiac efficiency Kim Ho1,2, Cory Wagg1,2, Liyan Zhang1,2, John Ussher2,3, Gary Lopaschuk1,2, 1Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada, 2Cardiovascular Research Centre, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada, 3Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada

29 The role of Midkine in Children with Dilated Cardiomyopathy

Xuan Jiang1, Kathleen Woulfe1, Anis Karimpour-Fard1, Keith Koch1, Brian Stauffer1,2, Shelley Miyamoto3, Carmen Sucharov1, 1University of Colorado-Denver, Aurora, CO, USA, 2Denver Health and Hospital Authority, Denver, CO, USA, 3Children’s Hospital Colorado, Aurora, CO, USA

30 Compartmentalized Gαq-Signaling in Adult Cardiac Myocytes Erika Dahl1, Steven Wu1, Chastity Healy1, Timothy O'Connell1, 1University of Minnesota, Minneapolis, MN, USA

31 Endogenous HAX-1 Regulates SERCA Activity and Oxidation Dependent Stability

Philip Bidwell1, Guan-Sheng Liu1, Narayani Nagarajan2, Kobra Haghighi1, George Gardner1, Junichi Sadoshima2, Evangelia Kranias1, 1University of Cincinnati, Cincinnati, OH, USA, 2Rutgers New Jersey Medical School, Newark, NJ, USA

32 Estrogen-independent female resilience in dystrophic models of heart failure

Tatyana Meyers1, Jackie Heitzman1, Lauren Aufdembrink1, Aimee Krebsbach1, DeWayne Townsend0, 1University of Minnesota, Minneapolis, USA

33 Contactless Particle Image Velocimetry (PIV) Method of Screening Drugs Using Human iPSC- derived Cardiomyocytes Sheeja Rajasingh1, Andras Czirok1, Dona Greta Isai1, Saheli Samanta1, Zhigang Zhou1, Buddhadeb Dawn1, Johnson Rajasingh1, 1University of Kansas Medical Center, Kansas City, USA

34 Influence of an ACE inhibitor on frailty and cardiac function in old male C57BL/6 mice Alice Kane1, Kailtyn Keller1, Susan Howlett1, 1Dalhousie University, Halifax, NS, Canada

35 Transient Receptor Potential Vanilloid 1 Mediates Laporotomy and Opioid-induced Infarct

Page 30: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

30

Size Reduction in Rats Eric Gross1, Yun Wu1, Helen Heymann1, Garrett Gross2, 1Stanford University, Stanford, CA, USA, 2Medical College of Wisconsin, Milwaukee, WI, USA

36 Circulating Factors Contribute to PDE5-Mediated Pathological Myocardial Remodeling in Single Ventricle Congenital Heart Disease Anastacia Garcia1,2, Stephanie Nakano1,2, Anis Karimpour-Fard1, Brian Stauffer1,3, Carmen Sucharov1, Shelley Miyamoto1,2, 1University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA, 2Children’s Hospital Colorado, Aurora, CO, USA, 3Denver Health and Hospital Authority, Denver, CO, USA

38 Function Beyond RNA Splicing for RBFox Family Members in Heart

Chen Gao1,2, Jing Hu3, Chaoliang Wei4, Yunhua Esther Hsiao5, Shuxun Ren1,2, Yuanchao Xue3, Yu Zhou3, Jianlin Zhang6, Ju Chen6, Xinshu Xiao5, Xiang-Dong Fu3,5, Yi Xing7, Yibin Wang1,2, 1Division of Molecular Medicine, Cardiovascular Research Laboratories, University of California, Los Angeles, Los Angeles, California, USA, 2Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA, 3Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, California, USA, 4Department of Cell Biology and Medical Genetics, School of Medicine, Shenzhen University, Shenzhen, China, 5Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California, USA, 6Department of Medicine, University of California, San Diego, San Diego, California, USA, 7Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA

39 Enhanced Activation of Inflammasome Promotes Atrial Fibrillation

Chunxia Yao1, Larry Scott Jr.1, Tina Veleva2, Frank U. Müller3, Stanley Nattel4, Dobromir Dobrev2, Xander H.T. Wehrens1, Na Li1, 1Baylor College of Medicine, Houston, TX, USA, 2University Duisburg-Essen, Essen, Germany, 3University of Münster, Münster, Germany, 4Montreal Heart Institute, Montreal, QC, Canada

40 Mechano-chemo-transduction is attenuated in a rabbit model of heart failure

Rafael Shimkunas1, Bence Hegyi1, Zhong Jian1, Zana Coulibaly1, Kit S. Lam1, Kenneth S. Ginsburg1, Julie Bossuyt1, Donald M. Bers1, Leighton T. Izu1, Ye Chen-Izu1, 1University of California, Davis, Davis, CA, USA

41 Protein tyrosine phosphatase 1B is a regulator of microRNA-mediated gene silencing and

cardiac hypertrophy Benoit Boivin1,2, 1SUNY Polytechnic Institute, Albany, New York, USA, 2Montreal Heart Institute, Montreal, Quebec, Canada

42 THE IMPACT OF AGE AND FRAILTY ON VENTRICULAR STRUCTURE AND FUNCTION IN

C57BL/6 MICE Hirad Feridooni1, Alice Kane1, Omar Ayaz1, Ali Boroumandi2, N Polidovitch2, Robert Tsushima2, Robert Rose1, Susan Howlett1, 1Dalhousie University, Halifax, Canada, 2York University, Toronto, Canada

43 Oroxylin A reduces angiotensin II- induced hypertrophy and mitochondrial dysfunction by

activating sirtuin 3 in cardiac myocytes Niria Treviño Saldaña2,3, Gerardo de Jesús García Rivas2,3, Luz Leticia Elizondo Montemayor2,3, 1Centro de Investigación Biomédica de la Escuela Nacional de Medicina, Monterrey, Nuevo León, Mexico, 2Instituto de Cardiología y Medicina Vascular del Centro Médico Zambrano Heullion, Monterrey, Nuevo León, Mexico

44 The Roles of Dopamine Receptor 3 in Age- and Sex-dependent Left Ventricular Remodeling

Gabriel Grilo1, Patti Shaver1, Stefan Clemens1, Lisandra de Castro Brás1, 1East Carolina University, Greenville, NC, USA

Page 31: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

31

45 In vivo reduction of mitochondrial oxidative stress abolishes spontaneous arrhythmic sudden cardiac death (SCD) in non-ischemic heart failure (HF) Swati Dey1, Deeptankar DeMazumder1, Brian O'Rourke1, 1Johns Hopkins University, Baltimore, MD, USA

46 Molecular function of Sigma-1 receptor in obesity-induced metabolic dysfunction

Chowdhury S. Abdullah1, Shafiul Alam1, Richa Aishwarya2, Jonette M. Green1, A. Wayne Orr1, Matthew D. Woolard3, Aimee E. Vozelinek3, Norman R. Harris2, Randa S. Eshaq2, Christopher G. Kevil1, Md. Shenuarin Bhuiyan1, 1Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA, 2Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center- Shreveport, Shreveport, LA, USA, 3Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA

47 Evidence for Reductive Stress in the Heart Failure Patients

Thiagarajan Sairam1, Gobinath Shanmugam2, Madhusudhanan Narasimhan3, Meenu Subramanian1, Amit N. Patel4, Rajendran Gopalan1, Ramalingam Sankaran1, Rajasekaran Namakkal Soorappan1,2, 1PSG Institute of Medical Sciences & Research, , Coimbatore, Tamil Nadu, India, 2Department of Pathology/Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL., USA, 3Texas Tech University Health Sciences Center, Lubbock, TX, USA, 4University of Miami – Miller School of Medicine, Miami, FL, USA

48 Interleukin-1a Blockade Reduce Acute Myocardial Ischemic Injury In The Mouse Adolfo Mauro1, Eleonora Mezzaroma1, Juan Torrado1, Salvatore Carbone1, Benjamin Vantassel1, Antonio Abbate1, Stefano Toldo1, 1Virginia Commonwealth University, Richmond, USA

49 Constitutive Activation of Nrf2 Causes Hyper-Reductive State and Heart Failure Gobinath Shanmugam1, Madhusudhanan Narasimhan2, Silvio H. Litovsky1, Jolyn Fernandes3, Kevin Whitehead4, John R. Hoidal4, Thomas W. Kensler5, Dean P. Jones3, E. Dale Abel6, Namakkal- Soorapppan Rajasekaran1,4, 1Cardiac Aging & Redox Signaling Laboratory, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA, 2Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA, 3Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory Univeristy, Atlanta, GA, USA, 4Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA, 5Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, USA, 6Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA

50 Mitochondrial membrane protein Sigmar1 regulates mitochondrial dynamics and function

Shafiul Alam1, Chowdhury S. Abdullah1, Richa Aishwarya2, Jonette M. Green1, A. Wayne Orr1, Sumitra Miriyala3, Manikandan Panchatcharam3, Hanna Osinska4, John N. Lorenz5, Jeffrey Robbins4, Md. Shenuarin Bhuiyan1, 1Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA, 2Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center- Shreveport, Shreveport, LA, USA, 3Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA, 4Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Centre, Cincinnati, OH, USA, 5Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA

51 miR-181c Regulates Mitochondrial Calcium Influx by targeting Cytochrome C Oxidase

subunit 1 Soroosh Solhjoo1, Sangeetha Kannan1,2, Deepthi Ashok1, Brian O'Rourke1, Charles Steenbergen1, Samarjit Das1, 1Johns Hopkins University, Baltimore, MD, USA, 2B.S.Abdur Rahman University, Chennai, Tamil Nadu, India

52 Dual optical mapping of the innervated Langendorff-perfused heart reveals novel insights

into acute electrophysiological responses to sympathetic stimulation Lianguo Wang1, Srinivas Tapa1, Samantha Stuart1, Rachel Myles2, Kieran Brack3, Andre Ng3,

Page 32: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

32

Donald Bers1, Crystal Ripplinger1, 1University of California Davis, Davis, USA, 2University of Glasgow, Glasgow, UK, 3University of Leicester, Leicester, UK

53 Effect of thymoquinone on high fructose diet-induced metabolic syndrome in rats Pankaj Prabhakar1, KH Reeta1, Subir K Maulik1, Amit K Dinda1, Yogendra K Gupta1, 1All India Institute of Medical Sciences, New Delhi, India

Page 33: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

33

Author Index With Poster Numbers

Author Index

Abbate, Antonio 048

Abdullah, Chowdhury S 046, 050

Abel, E Dale 049

Abou-Setta, Ahmed M 104

Abrol, Neha 080

Aggarwal, Sandeep 062

Ahmad, Ferhaan 094

Ahsan, Alya 002

Aishwarya, Richa 046, 050

Alam, Shafiul 046, 050

Alanova, Petra 068

Altamimi, Tariq 013

Alvarez Jr., Roberto 014

Alvarez, Manuel G 067

Alvarez, Roberto 016

Amanakis, Georgios 018

Amouyel, Philippe 007

Anderson, Mark 080

Anderson, Robert 094

Aponte, Angel 017, 026

Aponte París, Shania 088

Appachi, Swathi 017

Arif, Awais 063, 064

Arrieta, Adrian 090

Ashok, Deepthi 051

Aufdembrink, Lauren 032

Au, Kevin 073

Ayaz, Omar 042

Banerjee, Aryamitra 056

Banta, Joseph 015

Bassel-Duby, Rhonda 079

Basu, Madhumita 057

Baxter, Gary 025

Benz, Michael 015

Bernardi, Paolo 026

Bers, Donald M 040, 052

Beseme, Olivia 007

Bezprozvannaya, Svetlana 079

Bhuiyan, Shenuarin 011, 019, 046, 050

Bidwell, Philip 031

Blackwood, Erik 060, 090

Boisvert, Sarah 073

Boivin, Benoit 041

Boroumandi, Ali 042

Bossuyt, Julie 040

Bouvet, Marion 007

Boylston, Jennifer 018

Brack, Kieran 052

Bui, Alex 096, 097

Cai, Wenxuan 024, 092

Cao, Quan 096, 097, 102

Carbone, Salvatore 048

Casin, Kevin 008

Cephas, Amelia 057

Chambliss, Ken 017

Chandra, Mini 011, 019

Chavarria, Monica 016

Chen, Biyi 080

Chen-Izu, Ye 040, 098

Chen, Ju 038

Choi, Howard 102

Choi, Sung Won 066

Chowdhury, Shamim A K 067

Christodoulou, Danos 066

Chuprun, J Kurt 055

Chwastyniak, Maggy 007

Chytilova, Anna 069

Clemens, Stefan 044

Cobb, Melissa 027

Copstein, Leslie 104

Page 34: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

34

Coulibaly, Zana 040, 098

Csordas, Gyorgy 103

Czirok, Andras 033

Dahl, Erika 030

D'Ambrosio, Matthew 015

Das, Samarjit 051

Davis, 3rd, Robert T 067

Dawn, Buddhadeb 033

de Castro Brás, Lisandra 044

Dechaumes, Arthur 007

De Couto, Geoffrey 071

De La Fuente, Sergio 103

de Lange, Willem 022, 092

DeMazumder, Deeptankar 045

Denys, Ian 058

Deshpande, Mandar 006

Dey, Swati 045

Dhanani, Mehul 091

Dinda, Amit K 053

Divekar, Abhay 094

Djukovic, Danijel 066

Dobrev, Dobromir 039

Dodge-Kafka, Kimberly 088, 093

Donnarumma, Erminia 073

Doroudgar, Shirin 090

Dubois-Deruy, Emiliie 007

Ducas, John 104

Eisen, Howard 062

Fernandes, Jolyn 049

Fernandez-Sanz, Celia 103

Fillmore, Natasha 023

Finkel, Toren 026

Fitzsimons, Daniel 083, 084, 085, 086, 087

Florea, Victoria 077

Francis Stuart, Samantha 054

Fukushima, Arata 013

Fu, Xiang-Dong 038

Gao, Chen 038

Gao, Erhe 055

Gao, Juan 058, 070

Gao, Shouguo 023

Garcia, Anastacia 036

García Rivas, Gerardo de Jesús

043

Gardner, George 031

Garg, Vidu 057

Ge, Ying 024, 092

Ghimire, Anjali 021

Gildart, Moriah 093

Giles, Jasmine 083, 084, 087

Ginsburg, Kenneth S 040

Glembotski, Christopher C 090

Goldman, Jesse 062

Goodchild, Traci T 058, 070, 071, 073, 074, 075, 081, 082

Gopalan, Rajendran 047 Elizondo Montemayor, Luz 043

Leticia Granger, Jonathan 080

Elrod, John 103 Green, Eric 094

Elsnicova, Barbora 069 Green, Jonette M 046, 050

Escalante-Alcalde, Diana 011 Gregorich, Zachery 024, 0092

Eshaq, Randa S 046 Grilo, Gabriel 044

Farshidfar, Farnaz 104 Grisanti, Laurel 055

Feridooni, Hirad 042 Gross, Eric 035

Feridooni, Tiam 105 Gross, Garrett 035

Page 35: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

35

Guan, Qingdong 104

Gu, Haiwei 066

Guleria, Rakeshwar 009

Gupta, Sudhiranjan 009, 010

Gupta, Yogendra K 053

Haghighi, Kobra 031

Hahnova, Klara 069

Han, Jiawei 096, 097, 101

Hannallah, Benyamin 015

Hare, Joshua M 077

Harrington, Josephine 023

Harris, Norman R 046

Harrison, David 058

Hart, Gerald 080

Hasni, Syed 0062

Hatzistergos, Konstantinos E 077

Haushalter, Kristofer J 091

Healy, Chastity 030

Hegyi, Bence 040, 098

Heitzman, Jackie 032

Henze, Marcus 094

Heurer, Rachel 024

Heymann, Helen 035

Hlavackova, Marketa 069

H. Litovsky, Silvio 049

Ho, Kim 028

Holzerova, Kristyna 069

Hornberger, Lisa 013

Howell, Sarah A 091

Howlett, Susan 021, 034, 042

Hrdlicka, Jaroslav 068

Hsiao, Yunhua Esther 038

Huang, He 003

Ilves, Kelli 016

Isai, Dona Greta 033

Iverson, Elizabeth 084

Izu, Leighton T 040, 098

Jeffrey, Danielle A 020

Jenkins, James Stephen 075

Jeyaraman, Maya M 104

Jiang, Xuan 029

Jian, Zhong 040, 098

Ju, Chelsea 102

Kamp, Timothy 092

Kane, Alice 034, 042

Kang, Jianming 072

Kannan, Sangeetha 051

Kantor, Paul 013

Kapiloff, Michael 088, 093

Kapusta, Daniel 058, 070

Karam, Chehade N 067

Kardami, Elissavet 104

Karimpour-Fard, Anis 020, 029, 036

Karwi, Qutuba G 025

Kashani, Hessam 104

Kassab, Arwa 016

Katzenellenbogen, Benita 017

Katzenellenbogen, John 017

Kaufman, Christian 065

Kayani, Waqas 016

Kelepouris, Ellie 062

Keller, Kailtyn 034

Kelly, Jeff 060

Kevil, Christopher G 011, 019, 046

Khalafalla, Farid 016

Khan, Rizwan 002, 063, 064, 065

Khan, Sohaib 017

Khosravi, Seena 075

Kim, Sung-Hoon 017

Hu, Jing 038

Huqi, Alda 013

Ibetti, Jessica 055

Iftikhar, Omer 002

Page 36: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

36

Lopaschuk, Gary 013, 028

Lorenz, John N 050

Lucchese, Anna Maria 055

Luczak, Elizabeth 080

Lynch IV, Thomas 084

Macleod, Kenneth 078

Madwed, Jeffrey 074

Maier, Lars 080

Makarewich, Cat 079

Malter, Kyle 060

Mansour, Samer 104

Marban, Eduardo 071

Mathew, ST 004

Maulik, Subir K 053

Mauro, Adolfo 048

Mazhar, Murtaza 002, 065

McElmurry, Ron 012

Menazza, Sara 017, 026

Mesubi, Olurotimi 080

Meyerholz, David 094

Meyers, Tatyana 032

Mezzaroma, Eleonora 048

Micova, Petra 069

Miller, Adam 083, 087

Miriyala, Sumitra 011, 019, 050

Miyamoto, Shelley D 020, 029, 036

Moddrelle, David 056

Morton, Xavier 056

Moss, Richard 024, 083, 084, 085, 086, 087

Mota, Roberto 095

Mulder, Paul 007

Muliono, Alvin 014

Müller, Frank U 0039

Mungrue, Imran N 076

Murphy, Elizabeth 017, 018, 023, 026

Koch, Keith 029

Koch, Walter 055

Kohr, Mark 008

Kohut, Andrew 062

Koiwaya, Hiroshi 059, 070, 081, 082

Kolar, Frantisek 068, 069

Kolwicz Jr, Stephen C 066

Konorev, Eugene 027

Kranias, Evangelia 031

Kozlowski, Joseph 074

Krebsbach, Aimee 032

Kyi, Vincent 101

Lambert, Jonathan 103

Lamblin, Nicolas 007

Lam, Kit S 040

Lam, Maggie PY 061, 089

Landin, Ana Marie 077

Lau, Edward 061

Lawrence, Matthew 056

Lee, Jessica 096, 097

Lee, Kevin 015

Lee, Nelson 015

Lefer, David J 058, 059, 070, 071, 072, 073, 074, 075, 076, 081, 082

Liddie, Shervin 056

Liem, David A 096, 097, 101, 102

Li, Li 010

Li, Na 039

Lin, Ziqing 024

Liu, Guan-Sheng 031

Liu, Jian 074

Liu, Poching 023

Li, Zhen 070, 072, 073, 074, 076, 081, 082

Page 37: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

37

Myles, Rachel 052 Paxman, Ryan 060

Nagarajan, Narayani 031 Pena Heredia, Celia 100

Najman, David 002 Pentoney, Amber N 090

Nakano, Stephanie 036 Pham, Thuy 100

Namakkal 047 Pinet, Florence 007 Soorappan, Rajasekaran Ping, Peipei 061, 089, 096, Nandin, Garmaa 005 097, 101, 102

Narasimhan, Madhusudhana 047, 049 P. Jones, Dean 049 n

Natsumeda, Makoto 077

Nattel, Stanley 039

Neckar, Jan 068, 069

Plate, Lars 060

Polhemus, David J 058, 059, 070, 071, 072, 073,

Ng, Andre 052

Nguyen, Lisa O 076

Niesman, Ingrid R 091

Nigh, Evan D 077

Norman, Benjamin 016

Novakova, Olga 069

Novotny, Jiri 069

N. Patel, Amit 047

Nunley, Karin 020

O'Connell, Timothy 030

Olson, Eric 079

Organ, Chelsea L 072, 074, 076

O'Rourke, Brian 045, 051

Orr, A Wayne 011, 019, 046, 050

Osinska, Hanna 050

Palaniyandi, Suresh S 006

Panchatcharam, Manikandan 011, 019, 050

Pan, Guodong 006

Papousek, Frantisek 068

Parks, Randi 026

Parry, Traci 095

Pasumarthi, Kishore 105

Patel, Hemal H 091

Patel, Jitandra 024

Patel, Jitandrakumar 085, 086, 087

075, 081, 082

Polidovitch, N 042

Polson, Jennifer 102

Pomenrantz, Joel 080

Po, SS 004

Prabhakar, Pankaj 053

Qadar, Sheikh MZ 104

Rabbani, Rasheda 104

Raftery, Daniel 066

Raghavan, Meera 027

Rajasekaran, Namakkal- Soorapppan

049

Rajasingh, Johnson 033

Rajasingh, Sheeja 033

Ralphe, J Carter 022, 092

Rawat, Sonia 013

Rebeyka, Ivan 013

Reddy, Anilkumar 100

Reeta, KH 053

Reilly, John 075

Ren, Shuxun 038

Ren, Yi 012

R. Hoidal, John 049

Richard, Vincent 007

Rieger, Angela C 077

Ripplinger, Crystal 052, 054

Ritterhoff, Julia 066

Page 38: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

38

Robbins, Jeffrey 050

Rodriguez, Gianna M 077

Rokita, Adam 080

Rose, Robert 042

Rushing, Amanda 073, 075

Ryba, David M 067

Sadoshima, Junichi 031

Sairam, Thiagarajan 047

Samanta, Saheli 033

Sankaran, Ramalingam 047

Santo Domingo, Michelle 090

Sato, Priscila 055

Scarborough, Amy 073, 075

Schilling, Jan M 091

Schriewer, Jill 027

Schulman, Ivonne H 077

Schumacher, Jeffrey 073

Scott Jr., Larry 039

Shanmugam, Gobinath 047, 049

Shao, Dan 066

Shaul, Philip 017

Shaver, Patti 044

Sheu, Shey-Shing 103

Shimkunas, Rafael 040, 098

Shi, Yu 101

Siddiqui, Waqas 062

Silverman, Benjamin 062

Skidmore, Becky 104

Small, Kersten 074

Smart, Frank 058

Smelter, Dan 022

Solaro, R John 067

Solhjoo, Soroosh 051

Song, Long-Sheng 080

Springer, Danielle 023

Stauffer, Brian 029, 036

Stauffer, Brian L 020

Stauffer, Winston T 090

Steenbergen, Charles 051

Stoehr, Andrea 023

Stuart, Samantha 052

Subramanian, Meenu 047

Sucharov, Carmen C 020, 029, 036

Sulaiman, Wasan 104

Sun, Junhui 017, 018

Sussman, Mark 014, 016

Taffet, George 100

Tahir, Ammar 002

Takano, Ana 009

Tao, Fangbo 101

Tapa, Srinivas 052, 054

Tatman, Phillip D 020

Taylor, Matthew R G 020

Terracciano, Cesare 078

Thadani, U 004

Thuerauf, Donna J 090

Tian, Rong 066

Tilley, Doug 055

Toldo, Stefano 048

Tompkins, Bryon A 077

Torrado, Juan 048

Townsend, DeWayne 032

Tran, Nicole 063, 064

Traynham, CJ 055

Treviño Saldaña, Niria 043

Trivedi, Rishi K 059, 070, 071, 072, 073, 075,

081, 082

Tsushima, Robert 042

Ussher, John 028

van Berlo, Jop 012

Vantassel, Benjamin 048

Vasinova, Jana 068

Veleva, Tina 039

Page 39: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

39

Verma, Ravina 091 Xiao, Liang 058

Victoria, Sam 073 Xiao, Xinshu 038

Villet, Outi 066 Xing, Yi 038, 089

Voudouris, Apostolos 015 Xue, Yuanchao 038

Vozelinek, Aimee E 046 Yang, Yanqin 023

Wagg, Cory 013, 028 Yan, Jie 066

Wang, Bingyan 014 Yao, Chunxia 039

Wang, Brian 078 Yellamilli, Amritha 012

Wang, Ding 102 Yoo, Daniel 081, 082

Wang, Lianguo 052, 054 Yuan, Ancai 055

Wang, Qinchuan 080 Zarychanski, Ryan 104

Wang, Shubing 074 Zemljic-Harpf, Alice E 091

Wang, Wei 096, 097, 102 Zhang, Jianhua 092

Wang, Xujing 023 Zhang, Jianlin 038

Wang, Yibin 038 Zhang, Liyan 013, 028

Warren, Chad M 067 Zhang, Xue 023

Watson, Karol 096, 097 Zhang, Zhen 066

Wehrens, Xander HT 039, 080 Zhong, Peng 003

Wei, Chaoliang 038 Zhou, Yijiang 096, 097

Weiss, Robert 094 Zhou, Yu 038

Whitehead, Kevin 049 Zhou, Zhigang 033

Whiteman, Matt 025 Zhu, Jun 023

Wieczorek, David F 067 Zibilich, Craig 074

Willis, Monte 095 Zurmanova, Jitka 069

Wiseman, Luke 060

W. Kensler, Thomas 049

Wolska, Beata M 067

Wong, Fiona 094

Woodall, Meryl 055

Wood, Mark 025

Woolard, Matthew D 046

Woulfe, Kathleen C 020, 029

Wu, Joseph 011, 019

Wu, Steven 030

Wu, Yuejin 080

Wu, Yun 035

Xian, Ming 072

Page 40: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

40

002 Chronic Inotrope Infusion: A Viable Option for Low Output Heart Failure Rizwan Khan1, Murtaza Mazhar1, Ammar Tahir2, Alya Ahsan2, Omer Iftikhar2, David Najman2, 1University of Oklahoma, Oklahoma City, OK, USA, 2The University of Chicago (NorthShore University HealthSystem), Evanston, IL, USA

Introduction: The use of inotropes in low cardiac output heart failure (HF) has been controversial. While widely used clinically, randomized trials have had conflicting data. There is insufficient evidence to support long-term inotrope use secondary to the increased mortality seen in patients with advanced heart failure. We present an interesting case of a female with low cardiac output heart failure who has been on a stable regimen of chronic dobutamine and milrinone treatment for over 2 years. Case description: Our case is about a 78 year old female with an extensive past medical history, including ischemic cardiomyopathy post bypass surgery, systolic HF (NYHA Class III), atrial fibrillation, ventricular tachycardia and subsequent biventricular pacemaker, venous thrombosis, mitral valve replacement, end stage renal disease with hemodialysis 3 days a week. Given all of her co- morbidities, she was deemed to not be a candidate for LVAD or transplant. She continued to have multiple admissions for congestive HF. Treatment options were limited due to hypotension, and she was unable to tolerate ace inhibitors, hydralazine, isosorbide dinitrate and spironolactone. She was started on continuous dobutamine infusion and intermittent milrinone infusion with hemodialysis sessions. The patient has now been on this regimen for over 2 years, and has been tolerating well, with decreased hospital admissions for congestive heart failure. Discussion: While the use of inotropic agents chronically continues to be controversial, it is important to note that many of these studies were performed on patients without low output HF limited by hypotension. Recent studies have shown that for patients with low output HF, milrinone and dobutamine, the two inotropes approved for use in the U.S., can improve NYHA class, may reduce the number in hospitalizations, and may not affect overall mortality. Our case demonstrates a situation where the use of chronic inotropes has been clearly beneficial for our patient. Conclusion: Outpatient chronic inotrope infusions may be an effective form of therapy for selected patients with severe, low output congestive failure who are unable to take conventional HF medications secondary to hypotension.

003 CaMKII as a pathological mediator of inflammation, oxidative stress, ER stress, autophagy and mitochondrial dysfunction in free fatty acids/hyperlipidemia-induced cardiac remodeling both in vitro and vivo Peng Zhong1,3, He Huang1,2, 1Department of Cardiology, Renming Hospital of Wuhan University, Wuhan, China, 2Cardiovascular Research Institute, Wuhan University, Wuhan, China, 3Department of Cardiology, Johns Hopkins Medical Institute, USA

Rationale: The cellular mechanisms of obesity/hyperlipidemia-induced cardiac remodeling are multiple and not completely elucidated. Ca2+/calmodulin-dependent protein kinase II (CaMKII), a multifunctional serine/threonine kinase, has been reported to be involved in a variety of cardiovascular diseases. However, its role in obesity/hyperlipidemia-induced cardiac remodeling is still unknown. Objective: The objective of this study was to demonstrate the role of CaMKII in the pathogenesis of obesity/ hyperlipidemia-induced cardiac remodeling both in vitro and in vivo. Methods and Results: In cardiac-derived H9C2 cell model, palmitate treatment induced cell apoptosis coupled with activation of mitochondrial apoptotic pathway, cell hypertrophic and fibrotic response, while all these alterations were significantly inhibited by pharmacological inhibition of CaMKII with two specific inhibitors with Myr-AIP and KN93, respectively. In addition, increased inflammatory response coupled with activated MAPKs and NF-κB signaling pathway, exaggerated oxidative stress, ER stress and autophagy were also observed in palmitate-treated H9C2 cells, while pretreatment with CaMKII inhibitors significantly inhibited these pathological signaling. Furthermore, we also demonstrated TLR4 as the upstream signaling of CaMKII in palmitate- treated H9C2 cells. In APOE-/- mice fed a high fat diet (HFD) for 16 weeks, serum lipid profiles (FFAs, TG, TC) and glucose level were significantly increased compared with mice fed a normal diet. In addition, apparent cardiac hypertrophy, fibrosis and apoptosis, associated with increased inflammation, ER stress and autophagy were also observed in HFD-induced heart. However, all these changes were significantly reversed by 8-weeks peritoneal injection of KN93. And KN93 also increased antioxidant defence as evidenced by increased expression of Nrf2 system in HFD-induced heart. Conclusions: Taken together, our results demonstrated a critical role of CaMKII in the pathogenesis of obesity/hyperlipidemia-induced cardiac remodeling and TLR4 may be an upstream signaling of Cardiac CaMKII under hyperlipidemia conditions. These results suggested that CaMKII has the potential to be a therapeutic target in preventing obesity/hyperlipidemia-induced cardiac remodeling.

004 Inappropriate sinus tachycardia – symptom and heart rate reduction with ivabradine: pooled analysis of prospective studies ST Mathew1, SS Po1,2, U Thadani1, 1Veterans Affairs Medical Center/University of Oklahoma, Oklahoma City, OK, USA, 2Heart Rhythm Institute/University of Oklahoma, Oklahoma City, OK, USA

Objective: Inappropriate sinus tachycardia (IST), is a debilitating condition despite currently available treatment. Ivabradine, a selective sinus node blocker, has been used off-label to treat IST. We therefore systematically reviewed the literature and analyzed the quality of existing data and calculated sample size estimates for adequately powered studies. Methods: We included all available trials, open label prospective studies studies with ivabradine, specifically for IST among subjects with no structural heart disease with a minimum follow-up period of > 2 weeks. Heart rate and symptom reduction with ivabradine were estimated based on results of subjective change in symptoms assessed by data instruments used in the studies. All studies were assessed for quality using two validated checklists scoring the quality of studies. Sample size estimates for future studies were calculated based on the magnitude of symptom reduction encountered after treatment with ivabradine. Results: Nine studies met study criteria, culminating in 145 patients pooled with the majority being women (> 70%). All studies were small, most inadequately powered. All studies reported a decrease in maximum and/or mean resting heart rate with ivabradine. Symptom reduction demonstrated either complete or considerable amelioration. Most studies were at least of moderate quality. Excellent consistency of study quality was gauged by both data-quality tools and narrow limits of agreement between these tools. Calculated sample size estimates for adequately powered studies with a placebo effects of 30 and 50% was 56 and 242 patients respectively. Conclusions: Although ivabradine is effective in reducing heart rate and symptoms in IST patients, none of the studies were adequately powered to account for the expected placebo effect on symptoms. A multicenter-randomized, placebo controlled active comparative study with a beta-blocker is needed to confirm these findings. This is relevant given the ivabradine’s potential teratogenic effect as many IST patients are females of childbearing potential.

005 Study of Some Coronary Atherosclerotic Biomarkers Garmaa Nandin1, 1Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia

Background: Myocardial ischemia disease is explained by atherosclerosis, structural and functional failure of lipid metabolism, blood circulation, endothelial cell and cytokines, complement system, adhesion molecules, growth factors. There is no complete work on atheros pathogenesis. But it is still updating in different ways. Contemporary studies on atherosclerosis are rarely found in Mongolia. Thus it was served as the background of the recent study. Purpose: Determine the serum levels of some adhesive molecules and growth factors, estimate their pathogenetic roles in coronary atherosclerosis, and conduct correlation study on the structural changes of coronary walls. Methods and materials: The study was conducted by the case control model. Serum of 354 persons served as the materials in DuoSet® ELISA Development Systems to determine the levels of biomarkers such as ICAM1, VCAM1, LOX1, and VEGFA. A total of 54 cadavers’ coronary artery tissues were used in the immunohistochemical study to detect Anti-VCAM, LOX, and VEGFA antibody. Results: We compared the case and control groups: the level of serum VCAM1, ICAM1, LOX1, VEGFA increased in the case group. According to the high occlusion level of vessels the serum level of VCAM1, LOX1 declined in the early stages, and the VEGFA increased in the advanced atherosclerosis changes. According to the morphological study the intima was thickened, extracellular lipid accumulation formed, collagen fibers increased and the scarring process started robustly because of the infiltration of the inflammatory cells. After all fibro-atheroma, atheroma, and neovascularization were shaped. In this case the VCAM1 detection was VCAM1++. In the advanced case endothelial cells felt of the wall and it leads to the thrombosis while the plaque calcified. The ulceration was strongly detected. The periphery of the lesions demonstrated neovascularizationa and muscular layer. In this case VEGFA was VEGFA+. Conclusions: In coronary atherosclerosis the level of serum VCAM1, ICAM1, LOX1, VEGFA increased compared with the control group. The serum level of VCAM1, LOX decreased in the early changes accordingly with the high occlusion levels, but the VEGFA increased in the advanced cases. Immunohistochemical tests showed that in the early stage, the VCAM1 was determined as VCAM1++, and the VEGFA – VEGFA+. The number of VCAM1 positive cells decreased in the early coronary atherosclerosis changes and 0-II occlusion levels. But the VEGFA positive cells increased during the advanced changes and III-IV occlusion levels. Serum and histological tests shows that VCAM1 and VEGFA

Page 41: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

41

expressions served as the biomarkers during the early and advanced atherosclerosis changes, respectively.

006 Decreased Aldehyde Dehydrogenase (ALDH)2 Activity Contributes to Coronary Endothelial Dysfunction in Diabetic Cardiomyopathy Guodong Pan1, Mandar Deshpande1, Suresh S. Palaniyandi1, 1Henry Ford Health System, Detroit, MI, USA

Background: Around 8% of Americans acquire diabetes mellitus (DM). Diabetics cause micro and macrovascular complications to develop, that lead to end-organ damage. However, microvascular damage is understudied in diabetic cardiomyopathy (DCM), despite needing extensive coronary perfusion. Hyperglycemia-mediated reactive aldehydes, like 4-hydroxy-2- nonenal (4HNE) are associated with cardiac damage. Aldehyde dehydrogenase (ALDH) 2, a mitochondrial enzyme which detoxifies 4HNE, is implicated in endothelial cell function in vasculature. Thus, we hypothesize that DM-mediated 4HNE-induced coronary endothelial cell (CEC) injury potentiates DCM, which is further augmented by low ALDH2 activity. Methods and results: Type-1 DM was induced in C57BL/6 wild-type (WT-DM) and ALDH2*2 mice (with intrinsic low ALDH2 activity due to E487K mutation; ALDH2*2-DM). After 3 weeks, in comparison to WT-DM, we found decreased fractional shortening (%FS), an index of cardiac function (55±4 vs 40±11%, p<0.05) and decreased area of fibrosis (6±0.7 vs 9.5±2%, p<0.05) in ALDH2*2-DM. This indicates the augmentation of DCM in ALDH2*2-DM compared to WT-DM. Similarly, we found reduced CD31+ coronary endothelial cells (CECs): 2119±277 vs 478±62 cells/mm2, p<0.001, increased coronary perfusion: 77±7 vs 94±7 mmHg, p<0.01, and reduced eNOS levels in CECs in WT-DM vs ALDH2*2-DM respectively, which indicates more coronary endothelial dysfunction in DCM in ALDH2*2-DM. Furthermore, to substantiate our findings on role of ALDH2 in endothelial dysfunction, we subjected cultured mouse CECs to high-glucose (HG, 33mM D-Glucose) stress with ALDH2 inhibition by disulfiram (DSF+) or without DSF (DSF-). We found DSF augmented HG-induced 4HNE adducts: 1.4±0.2 vs 4±0.8 folds of Ctrl, p<0.05, mitochondrial ROS: 1.6±0.2 vs 3.3±0.4 folds of Ctrl, p<0.05, cell death: 28.8 ±4.2 vs 57.5± 8%, p<0.01, and decreased angiogenesis: 35.6±3.3 vs 16.7±3 branches/mm2, p<0.05, in CECs (in DSF- vs DSF+ respectively). Conclusion: ALDH2 plays an important role in protecting CECs from hyperglycemic stress. Decreased ALDH2 activity deteriorates coronary endothelial dysfunction in diabetic cardiomyopathy.

007 Dual interplay between desmin phosphorylation and dysregulated autophagy in heart failure Marion Bouvet1, Emiliie Dubois-Deruy1, Paul Mulder2, Maggy Chwastyniak1, Arthur Dechaumes1, Olivia Beseme1, Philippe Amouyel1, Nicolas Lamblin1, Vincent Richard2, Florence Pinet1, 1Inserm U1167, Lille, France, 2Inserm U1096, Rouen, France

Post-translational modifications (PTMs) of sarcomeric proteins could participate to left ventricular (LV) remodeling and contractile dysfunction leading in advanced heart failure (HF) with altered ejection fraction.Using an experimental rat model of HF (ligation of left coronary artery) and phosphoproteomic analysis, we highlighted an increase of phosphorylated desmin on serine residues in LV of 2 month-HF rats, dependent of PKCζ and GSK3β. We verified the desmin phosphorylation profile by Phos-Tag™ gel and observed that desmin is mainly expressed in insoluble fraction of LV of HF rats. Electron microscopy shows alteration of sarcomere in LV of HF rats due to desmin filaments desorganisation and immunofluorescence accumulation of desmin aggregates that could be toxic in cardiomyocytes. We then analyzed whether the autophagic pathways (macro-autophagy and chaperone-mediated autophagy (CMA)) may be involved in phosphorylated desmin clearance in our experimental model of HF. We found a decrease of macroautophagy processes (LC3II/LC3I, p62, beclin-1) throughout LV remodeling that may participate to hyperphosphorylated desmin accumulation. We identified that CMA is activated throughout the LV remodelling post-MI with increased CMA markers (Lamp-2a, hsc70, hsp90) in LV of HF rats suggesting that CMA may be involved in the clearance of aggregates of phosphorylated desmin. Bioinformatical analysis identified desmin as a new CMA substrate with 4 KFERQ motifs conserved in the aminoacid sequence of rat, mouse, bovine, pig, danio Rerio and human species. We then developed an in vitro model of CMA induction in neonate cardiomyocytes with geldanamycin. We observed increased levels of hsc70 and hsp90 and colocalization of desmin and Lamp2a that suggests desmin lysosomal sequestration by PLA technique. CMA activation did not induce changes in insoluble desmin but decreased levels of soluble desmin and phosphorylated soluble desmin. Conversely, preliminary data on inhibition of CMA with Lamp2a siRNA only induced increase of soluble desmin and phosphorylated soluble desmin. Our data suggest for the first time that CMA is able to clear

monomeric phosphorylated soluble desmin but is not sufficient for intracellular clearance of phosphorylated desmin aggregates in the experimental model of ischemic HF. We could speculate that induction of early CMA activation in post-infarction might be a therapeutic approach to fight against desmin aggregates in order to maintain cardiomyocyte survival.

008 Cardioprotective biology of S-nitrosylated Hypoxia-inducible Factor 1α Kevin Casin1, Mark Kohr1,2, 1Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA, 2Johns Hopkins School of Medicine, Baltimore, MD, USA

Ischemia-reperfusion (IR) injury is the leading cause of death in the United States. While cardioprotective therapies are effective at reducing injury in pre- clinical models, they have failed to reduce mortality in human populations. Our group has shown that increased protein S-nitrosylation (SNO), a nitric oxide- mediated protein modification, can reduce IR-dependent injury. The genetic ablation of S-nitrosoglutathione (GSNO) reductase (GSNO-R), a regulator of SNO, leads to an increase in SNO and cardioprotection. Interestingly, the SNO-mediated stabilization of HIF-1α was also noted with GSNO-R knockout mice, and was thought to underlie the protection observed in these mice. As a transcription factor, HIF-1α is responsible for orchestrating the cellular response to low oxygen by activating specific cell survival genes, but studies have yet to investigate the protective function of SNO-HIF-1α. We hypothesize that SNO alters the traditional function of HIF-1α as a transcription factor. HEK293 cells were either subjected to 3% oxygen, or treated with GSNO during normoxia. HIF-1α stabilization was observed under both conditions via western blot. We next investigated gene expression with RT-qPCR under these conditions, and found that hypoxia up-regulated several classic HIF-1α target genes, such as VEGFA (G: -1.29; H: 0.93-fold), EPO (G: -0.51; H: 3.42-fold), and NOS2 (G: 0.22; H: 3.25-fold). Interestingly, treatment with GSNO only up-regulated GLUT1 (G: 0.67; H: 1.06-fold) and HK2 (G: 0.48; H: 0.70-fold). We are now beginning to examine HIF-1α in GSNO-R knockout mice, and have confirmed protection from IR injury in a Langendorff model (WT: 40.2 +/- 3.09 % infarct, GSNO-R KO: 27.3 +/- 5.28 % infarct; p<0.05). We have also visualized HIF-1α stabilization in nuclear fractions of these hearts via western blot and will next examine transcriptional differences. In conclusion, our results suggest that SNO has the potential to mediate HIF-1α activity, and this may be important in cardioprotection.

009 Deficiency of miR-1954 promotes cardiac remodeling Sudhiranjan Gupta1, Ana Takano1, Rakeshwar Guleria1, 1Texas A&M University, Temple, TX-76504, USA

Cardiac remodeling due to hemodynamic overload is associated with significant morbidity and mortality. In response to stress, cardiomyocyte (CM) become hypertrophied whereas cardiac fibroblasts convert into myofibroblasts. The phenomenon leads to the development of cardiac hypertrophy, fibrosis and impair cardiac function. Previously, we have shown the pivotal role of miRNA (a new class of post-transcriptional regulators) in cardiac remodeling, but, loss of miRNA contributing to the onset of cardiac remodeling remains elusive. Using next generation miRNA sequencing, we discovered a panel of novel dysregulated miRNAs from read-data, secondary structure and miRPara classification score analysis in wild-type mice (WT) infused with Angiotensin II (Ang II). Among them, one was identified as miR- 1954, a novel miRNA which was significantly reduced in Ang II-infusion and transverse aortic constriction (TAC). Following an unbiased approach, we confirmed that Sp1-Gata4-Col I-Tsp1-axis is the bona-fide targets. Our hypothesis is that deficiency of miR-1954 exacerbates cardiac remodeling leading to hypertrophy and fibrosis through paracrine mechanism; and overexpression of miR-1954 mitigates the cardiac damage and abrogates remodeling by modulating Sp1-Gata4-Col I-Tsp1-axis. Our data demonstrated that depletion of miR-1954 in CM triggers hypertrophic response by modulating Sp1 and Gata4; releases soluble factors (Tgfβ1) that triggers cardiac fibroblasts proliferation; upregulation of thrombospondin 1 (Tsp1) and collagen I (Col I). Overexpression of miR-1954 in CM reverses these processes implicated a cellular cross-talk. Cardiac-specific overexpression of pre-miR-1954 transgenic mice (miR-1954 Tg) showed reduced cardiac mass and improved function compared to WT littermate after Ang II treatment. Inhibition of miR-1954 by locked nucleic acid of anti-miR-1954 exacerbates cardiac hypertrophy and fibrosis. Our findings provide evidence that loss of miR-1954 promotes cardiac remodeling by targeting Sp1-Gata4-Col I-Tsp1- axis and, overexpression of miR-1954 reverses the process. We conclude that miR-1954 could be a triggering factor in cardiac remodeling and providing new mechanistic information for therapeutic benefit.

Page 42: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

42

010 NF-kB-miR-23a-miR-27a: a critical modulator in post myocardial infarction remodeling Sudhiranjan Gupta1, Li Li1, 1Texas A&M University, Temple, TX, USA

Myocardial infarction (MI) is one of the leading causes of death around the world. Myocardial ischemia contributes to the development of MI and initiates cardiomyocyte (CM) apoptosis, myofibroblasts differentiation and extracellular matrix protein deposition and promotes left ventricle (LV) remodeling eventually leading to cardiac dysfunction. Previously, we have demonstrated the direct involvement of miRNAs (a new class of post-translational regulator) and NF-κB in LV remodeling. However, NF-κB-mediated miRNA modulation and miRNA/gene networks in post-MI remodeling remain elusive. Furthermore, with an emerging interest to combine cell transplantation with gene therapy, mesenchymal stem cells (MSC) are considered most beneficial of the stem cells in regenerative medicine. We have a paucity of data in genetically modified MSC on their mobilization within infarcted myocardium. We identified a panel of novel dysregulated miRNAs in the wild type (WT) mice subjected to MI using conventional miRNA array. These miRNAs were restored in IκBα triple-mutant (3M) mice indicated NF-κB-dependent regulation. Among them, miR-23a and miR-27a were significantly upregulated in MI. Our study has identified PPARγ-GSK3β-Bcl2-axis, a bona-fide target for miR-23a/-27a after unbiased in vitro screening. Our hypothesis is that NF-κB- dependent miR-23a/-27a is a pathogenic niche, exacerbates post-MI remodeling through several target pathways; and inhibition of NF-κB, miR- 23a/-27a and transplantation of 3M-MSC mitigates the cardiac damage and abrogates the cardiac remodeling by restoring PPARγ-GSK3β-Bcl2-axis. Inhibition of NF-κB normalized miR-23a/-27a expression and reduced CM death in hypoxia/ reoxygenation (H/R) model. Our data indicated that miR-23a is a key regulator in simulated H/R-induced injury. Inhibition of miR-23a also showed reduction of cell death and improved mitochondrial function im G/R model. Interestingly, 3M-MSC showed significant reduction in apoptosis, inflammation and oxidative stress in H/R-induced CM, compared to WT-MSC. Our findings provide evidence that miR-23a/-27a could be a triggering factor in post-MI remodeling and providing new mechanistic information for therapeutic benefit.

011 Mitochondrial superoxide in myocardial lysophosphatidic acid signaling Manikandan Panchatcharam1, Mini Chandra1, Diana Escalante-Alcalde2, Wayne Orr1, Christopher Kevil1, Shenuarin Bhuiyan1, Joseph Wu3, Sumitra Miriyala1, 1Louisiana State University Health Sciences Center, Shreveport, LA, USA, 2Universidad Nacional Autónoma de México, México DF, Mexico, Mexico, 3Stanford Cardiovascular Institute, Stanford, CA, USA

The bioactive lysophosphatidic acid (LPA) plays a well-known role in atherosclerotic disease, whereas, its role in myocardial function remains virtually unexplored. Following acute myocardial infarction, serum LPA concentration rises by six-fold over control human subjects, suggesting LPA may contribute to the pathogenesis of myocardial infarction. LPA production involves hydrolysis of lysophosphatidylcholine by the secreted enzyme autotaxin, whereas lipid phosphate phosphatase-3 (LPP3) catalyzes LPA dephosphorylation to generate lipid products that are not receptor active. We present the first evidence that cardiac ischemia/reperfusion (I/R) injury enhances myocardial autotaxin levels and decreases myocardial LPP3 expression, and this is associated with increased serum LPA levels. Upon reperfusion, reactive oxygen species production arises as a burst of superoxide from mitochondria following I/R injury. The redox-sensitive transcription factor NFAT has been shown to bind to the autotaxin promoter and induce its expression. Therefore, we looked at the autotaxin and LPP3 regulation in mice following I/R injury in the myocardium. After 1h ligation followed by 3h reperfusion in the myocardium, we observed a 3 fold increase in the autotaxin protein levels, whereas LPP3 protein levels were significantly downregulated as observed through Western blot analysis in these myocardial ischemic tissues. Autotaxin and miR-92a mRNA expression levels were significantly upregulated, whereas KLF2 and LPP3 mRNA expressions were significantly downregulated following I/R injury at 24 hours. Western blot analysis showed a 3 fold increase autotaxin protein levels and immunohistochemistry of human infarct tissues at 24 hours showed disruption of the sarcomere with decreased LPP3 staining. We found that I/R injury transactivates miR-92a, and inhibit KLF2, an upstream activator of LPP3. Taken together, our in vivo data, from the myocardial I/R injury and human infarct tissues, suggest that regulation of autotaxin and LPP3 activity might cause the rise in serum LPA levels as reported with acute myocardial infarct patients.

012 Abcg2-Expressing cells fuse with existing cardiomyocytes. Amritha Yellamilli1, Yi Ren1, Ron McElmurry1, Jop van Berlo1, 1Universsity of Minnesota, Minneapolis, Minnesota, USA

Cardiac side population cells (cSPCs) were the first group of progenitor cells identified in the heart; however, their progenitor cell properties have only been established in cell culture and after transplantation. To determine whether cSPCs possess progenitor cell properties in vivo, we generated an Abcg2- driven, lineage-tracing mouse model. In this model, 75.8 ± 10.87% of cSPCs are labeled with GFP. Over a four-week chase period, there is a five-fold increase in cardiomyocyte-labeling from 0.17 ± 0.15% to 0.84 ± 0.24%. Surprisingly, 90.7 ± 2.66% of labeled cardiomyocytes arise from fusion events and not direct differentiation from cSPCs based on labeling in Abgc2MCM/+

R26mTmG/+ mice. Given the extensive labeling of bone marrow and endothelial cells in our model, we hypothesized that the increase in cardiomyocyte labeling over the four-week chase period arises from fusion of unlabeled cardiomyocytes with GFP-labeled bone marrow cells or endothelial cells. To test this hypothesis, we irradiated Myh7Cre/+ R26tdTomato/+ mice, transplanted them with hematopoietic stem (LSK) cells isolated from Abgc2MCM/+ R26GFP/+

and pulsed them with tamoxifen. At the end of a four-week chase period, a similar percentage of bone marrow side population cells, LSK cells and differentiated lineages were labeled with GFP. More importantly, only 0.02 ± 0.03% of cardiomyocytes were labeled with both GFP and tdTomato. No cardiomyocytes were labeled with only GFP. Next, we evaluated whether fusion with endothelial cells could account for the increase in cardiomyocyte labeling we observed. Using an endothelial-specific, lineage-tracing mouse model, Cdh5CreER/+ R26mTmG/+, we found no cardiomyocytes labeled at the end of a four-week chase period. Our results demonstrate that the primary mechanism of cardiomyocyte labeling in our Abcg2-driven, lineage-tracing mouse model arises from fusion of cardiomyocytes with lineage-traced cells, but not with bone marrow or endothelial cells. These results indicate that fusion in the heart may occur at much higher rates than previously assumed, and may play important roles in homeostatic maintenance of cardiac function.

013 Cardiac hypertrophy suppresses glucose oxidation in newborns with congenital heart defects Sonia Rawat1, Arata Fukushima1,2, Liyan Zhang1, Alda Huqi1,3, Tariq Altamimi1, Cory Wagg1, Lisa Hornberger1, Paul Kantor1, Ivan Rebeyka1, Gary Lopaschuk1, 1Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada, 2Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan, 3Cardio Thoracic and Vascular Department, University of Pisa, Pisa, Tuscany, Italy

Introduction: Cardiac fatty acid oxidation in the newborn dramatically increases shortly after birth, while glucose oxidation remains low until weaning. In newborn rabbit hearts, volume overload hypertrophy delays this increase in fatty acid oxidation, while keeping glucose oxidation low. This results in a decreased capacity to produce energy in the heart and increases the susceptibility to ischemic injury. The presence of cardiac hypertrophy in patients with congenital heart defects also decreases the maturation of fatty acid oxidation, but it is not clear what happens to glucose oxidation. We therefore determined what happens to the control of glucose oxidation in hypertrophied human newborn hearts. Method: Human right ventricular biopsy samples were collected during corrective heart surgery from neonates aged 101-200 days (an age group with increased fatty acid oxidation enzyme activity compared to 0-100 day old hearts). Samples were grouped based on the presence or absence of right ventricular hypertrophy, as assessed by echocardiography. Results: Pyruvate dehydrogenase (PDH), the rate-limiting enzyme of glucose oxidation, had significantly increased phosphorylation in hypertrophied vs. non-hypertrophied hearts (1.08±0.09 vs. 0.81±0.06 arbitrary units, respectively, n=6, p<0.05), resulting in inactivation of PDH. Concomitantly, expression of the PDH kinases, PDK2 and PDK4, were significantly increased in hypertrophied hearts. Transcription factors PPARα and ERRα, which are known to regulate PDH kinases, showed no changes between groups, while E2F1, a transcriptional regulator of PDK2 and PDK4, was increased. Components of the E2F1 pathway were also upregulated in the hypertrophied group, namely P-cyclin D1 and CDK4. Conclusion: Hypertrophy of the newborn heart upregulates E2F1-mediated transcription of PDH kinases, thereby phosphorylating and inhibiting PDH and reducing glucose oxidation rates. This may contribute to compromised energetics in the newborn heart.

Page 43: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

43

014 Cardiac Progenitor Cell Lineage Tracing During Embryonic Cardiomyogenesis Bingyan Wang1, Alvin Muliono1, Roberto Alvarez Jr.1, Mark Sussman1, 1San

Diego State University, San Diego, CA, USA

Background: Stem cell therapy represents great promises to myocardium regeneration. Multipotent c-Kitpos cardiac progenitor cells (CPCs) are able to differentiate into endothelial cells, smooth muscle cells, and cardiomyocytes. However, fundamental knowledge of CPC biology remains incomplete. Studies in rodent myocardial infarction model revealed that CPCs have poor long-term survival and engraftment after adoptive transfer, perhaps due to severely damaged host environment. Therefore, it is critical to understand how CPCs interface with recipient environment following transfer in order to enhance their true regenerative potentials. Hypothesis: Adoptively transferred stem cells are thought to survive and engraft best in an environment closely resembling their original habitat. Thus, we hypothesized that embryonic environment provides optimal spatiotemporal conditions to promote CPCs engraftment and commitment to cardiac fate. Methods: CPCs isolated from adult mouse hearts were expanded, fluorescence-tagged, and injected into blastocysts at E3.75 and in utero at E15.5. Embryos were analyzed following cardiogenesis by immunofluorescence for presence of CPC-derived tissues. Additionally, CPCs were injected intramyocardially at various stages from P0 to P7, to follow long-term adoptive transfer and assess CPCs lineage commitment. Results and Conclusions: At 48 hours post injection, donor CPCs were found anchoring in blastocoel and trophoblasts at E5.5, and were detected within host myocardium at E17.5 predominantly at perivascular regions (n=4). Interestingly, CPCs also integrated into aminochorionic sac, indicating a novel non-cardiogenic fate of CPCs (n=5). CPCs injected at P3 stably engrafted into left ventricular myocardium by 14 days post injection (n=4), sharing gap junction proteins (ZO-1, Connexin-43) with neighboring cells. In conclusion, this study provides vivid evidence for the first time of CPC engraftment and survival in vivo under homeostasis during cardiogenesis. Future studies will assess permissive environmental conditions and cardiogenic potential of CPCs, which may optimize their use in therapeutic applications and provide fundamental insights on CPCs biology.

015 Takotsubo Cardiomyopathy-Associated Ventricular Standstill in a Peripartum Patient Nelson Lee1,2, Kevin Lee1,2, Joseph Banta1,2, Matthew D'Ambrosio1,2, Benyamin Hannallah2, Michael Benz2, Apostolos Voudouris2, 1Rowan University School of Osteopathic Medicine, Stratford, NJ, USA, 2Christ Hospital, Jersey City, NJ, USA

Takotsubo cardiomyopathy (tako tsubo translates to “octopus pot” in Japanese) is a stress-related cardiomyopathy in which weakening of the cardiac musculature leads to left ventricular failure with apical akinesis and ballooning. Takotsubo Cardiomyopathy is hypothesized to be triggered by emotional stress leading to a catecholamine surge and symptoms that can mimic acute coronary syndrome. It has been well documented that Takotsubo cardiomyopathy can lead to several fatal ventricular arrhythmias. What we describe here is the first reported case of symptomatic ventricular standstill in Takotsubo Cardiomyopathy. We noted persistent failure of the conduction system leading to degeneration of the circuit and ventricular standstill six (6) days after initial presentation. It should be noted that in Takotsubo cardiomyopathy, symptoms typically resolve in approximately 24-48 hours. We suspect that ventricular ballooning impairs the cardiac conduction system such that the AV node and His-Purkinje system can no longer effectively communicate electrical impulses to the remainder of the electrical circuit. In our case, a previously healthy 24-year-old, six-month-postpartum, Egyptian female presented to the ED with complaints consistent with acute coronary syndrome. Multiple syncopal episodes were witnessed and a new right bundle branch block was noted with intermittent complete atrioventricular blocks progressing to a ventricular standstill. Cardiac catheterization and serology was used to rule out other underlying etiologies of this patient’s cardiac symptoms. An emergent transvenous pacemaker was then inserted due to the instability of the patient’s clinical condition leading to placement of a dual- chambered pacemaker. The patient also expressed concerns for returning to breastfeeding her infant son during the course of her hospital stay. During follow-up, the patient has reported no further syncopal episodes and has had no complications since pacemaker placement. Our findings highlight the medical management for prolonged ventricular standstill of the cardiac conduction system in a symptomatic postpartum patient with Takotsubo cardiomyopathy.

016 P2Y14 nucleotide receptor overexpression: Letting blind cardiac progenitor cells 'see' again Farid Khalafalla1, Waqas Kayani1, Arwa Kassab1, Kelli Ilves1, Roberto Alvarez1, Monica Chavarria1, Benjamin Norman1, Mark Sussman1, 1San Diego State University, San Diego, California, USA

Heart failure (HF) is a leading cause of death due to limited regenerative capacity of adult mammalian heart following injury. Autologous stem cell therapy holds promise for promoting cardiac regeneration. However, stem cells derived from aged/diseased organs exhibit poor growth and survival capabilities. Empowering cardiac progenitor cells (CPC) with prosurvival genes has been attempted. Nonetheless, the molecular mechanisms by which stem cells initially detect stress signals to stimulate appropriate regenerative responses are poorly understood. This study aims to explore the physiological responses mediated by purinergic receptors, which represent a major detector for extracellular nucleotides released during injury/stress, with a focus on P2Y14 nucleotide receptor (P2Y14R) activated by extracellular UDP-conjugated sugars. P2Y14R mediates proliferation of keratinocytes and chemotaxis of neutrophils and hematopoietic stem cells (HSCs). In addition, P2Y14R enhances HSC resistance to stress-induced senescence and maintains regenerative capacity after injury. However, the physiological roles of P2Y14R in CPCs are largely unknown. Preliminary data show striking correlations between P2Y14R expression in human CPCs derived from HF patients (hCPCs) and patients’ ejection fraction (EF), where low EF corresponds to low P2Y14R expression hCPCs. Moreover, hCPCs with relatively slower growth kinetics and enhanced senescence exhibit dramatic decreases in P2Y14R expression compared to fast-growing hCPCs. P2Y14R overexpression improves hCPC proliferation, migration, survival under stress stimuli and reverses senescent-associated phenotypes. Furthermore, P2Y14R- overexpressing hCPCs show remarkable upregulation in the expression of paracrine factors critical for cardiac repair. Preliminary studies will be extended in vivo to assess whether P2Y14R overexpression in hCPCs improves their reparative potential for injured mouse myocardium. Overall, this study introduces a novel interventional molecular approach to improve the therapeutic outcome of hCPCs by enhancing their capability to detect stress- induced extracellular nucleotides and initiate proper regenerative responses through augmenting P2Y14R expression.

017 Membrane delimited estrogen receptor activation protects heart against ischemic-reperfusion injury in mice with cardiac but not endothelial specific ablation of ERα Junhui Sun1, Sara Menazza1,6, Swathi Appachi1, Ken Chambliss5, Sung-Hoon Kim2, Angel Aponte1, Sohaib Khan3, John Katzenellenbogen2, Benita Katzenellenbogen4, Philip Shaul5, Elizabeth Murphy1, 1Systems Biology Center, NHLBI/NIH, Bethesda, MD, USA, 2Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA, 3Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA, 4University of Cincinnati Cancer Center, Cincinnati, OH, USA, 5Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA, 6Department of Biomedical Sciences, University of Padova, Padova, Italy

Steroid hormone receptors, such as estrogen receptors (ER) function as ligand-regulated transcription factors. However, recent data indicate that estrogen also elicits effects through binding to estrogen receptors (ER-α, ER- β and GPER) at the plasma membrane and initiating signaling. In this study, ovariectomized C57BL/6J mice were treated with estradiol (6 µg/day) or estrogen-dendrimer conjugate (EDC, a membrane delimited ER modulator) for two weeks. Ischemia-reperfusion injury was evaluated in Langendorff perfused hearts. Similar to estradiol-treated hearts, EDC treatment significantly decreased infarct size and improved post-ischemic functional recovery. EDC treatment also resulted in an increase in protein S-nitrosylation (SNO), consistent with previous studies showing a SNO role in cardioprotection. In further support of a role for SNO, inhibition of nitric oxide synthase but not soluble guanylyl cyclase blocked EDC-mediated protection. ICI182,780 (antagonist of classic ER but agonist of GPER) significantly blocked the EDC-mediated cardioprotection, suggesting EDC-induced protection is mediated by classic ER. Cardiac- or endothelial-specific (cs- or es-) ERα knockout (KO) mice were generated to test tissue-specific ERα involved in EDC protection. In cs-ERαKO mice, EDC treatment still significantly decreased infarct size and improved functional recovery similar to that found in wildtype, while protection was lost in es-ERαKO mice, suggesting an important role for endothelial ERα. In contrast to wildtype hearts, EDC treatment did not increase SNO in es-ERαKO mice. ERα but not ERβ was found significantly decreased in cs-ERαKO mice, while opposite results were

Page 44: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

44

found in es-ERαKO mice. Thus, loss of EDC-induced protection and SNO increase in es-ERαKO mice could also be due to significant down-regulation of cardiac ERβ/SNO signaling. In conclusion, these results demonstrated a role of activation of membrane delimited ER in cardioprotection, suggesting that EDC-like compounds could be used clinically to provide cardiovascular benefit without the classical steroid hormone side effects, such as stimulation of uterine and breast cancer.

018 Cardioprotection in mice with a knock- in mutation in Cyclophilin D (CypD- C202S): a site of S- nitrosylation Georgios Amanakis1, Junhui Sun1, Jennifer Boylston1, Elizabeth Murphy1, 1NHLBI/NIH, Bethesda, MD, USA

Our previous study in mouse embryonic fibroblasts showed that cysteine 202 of cyclophilin D (CyPD) is necessary for redox stress-induced activation of the mitochondrial permeability transition pore (mPTP). To further investigate the essential function of this cysteine residue in situ, we used CRISPR to develop a knock-in mouse model (C57BL/6N strain), where CyPD cysteine 202 was mutated to a serine (C202S-KI). The amount of total CyPD expressed in the CyPD C202S-KI did not differ compared to the wild-type (WT). However, the CyPD C202S-KI mouse hearts elicit a significant cardioprotective effect against ischemia-reperfusion (I/R) injury in the Langendorff perfused heart model. After 20 min of global ischemia followed by 90 min of reperfusion, the post-ischemic recovery of rate pressure product (RPP= heart rate x LVDP) was 45.0±4.2% in CyPD WT and 59.6±4.0% in CyPD C202S-KI mice (p=0.0455). Myocardial infarct size was decreased in CyPD C202S-KI mouse hearts versus CyPD WT mice (24.5±4.7% vs 49.8±2.7%, p=0.0095). Isolated heart mitochondria from CyPD C202-KI mice had a higher calcium retention capacity compared to CyPD WT mice (213.3±16.67 vs 140.0±20.82 umol Ca+2/g protein, p=0.0371). However, in contrast to CyPD knockout mice which exhibit more pronounced cardiac hypertrophy in response to pressure overload stimulation than control mice, CyPD C202S-KI mice developed a comparable level of hypertrophy to their WT littermate in an angiotensin II- induced hypertrophy model delivered by implanted osmotic minipumps. In conclusion, these results show that mutated CyPD C202S affords cardioprotection against I/R injury, suggesting that the redox-modification of cysteine 202 might play an important role in the regulation of CyPD and its downstream targets such as mPTP.

019 Generation of MnSOD cardiomyocyte-specific knockout: Role in Heart failure development and progression Sumitra Miriyala1, Mini Chandra1, Wayne Orr1, Christopher Kevil1, Shenuarin Bhuiyan1, Joseph Wu2, Manikandan Panchatcharam1, 1Louisiana State University Health Sciences Center, Shreveport, LA, USA, 2Stanford Cardiovascular Institute, Stanford, CA, USA

Manganese Superoxide Dismutase (MnSOD), an antioxidant enzyme that catalyzes the conversion of superoxide radicals (O2•-) in mitochondria. Constitutive activation mitochondrial reactive oxygen species (ROS) has been implicated in both the pathogenesis and the progression of cardiovascular disease. The absence of SOD2 (a gene that encodes MnSOD) is found to be embryonic lethal in animal models due to impairment of mitochondrial function, most noticeably in the heart. In our earlier investigation, we have shown that the MnSOD mimetic, MnTnBuOE-2-PyP5+ distributes 3-fold more in mitochondria than in cytosol. The exceptional ability of MnTnBuOE-2- PyP5+ to quench O2•- parallels its ability to reduce ONOO− and CO3−. Based on our earlier reports, we have generated mice that specifically lack MnSOD in cardiomyocytes (Mhy6-SOD2Δ). These mice showed early mortality ~4 months due to cardiac mitochondrial dysfunction. FACS analyses using Mito-Tracker Green indicated that the mass of mitochondria per cell was slightly decreased in the Mhy6-SOD2Δ to the wild type. We then examined Oxidative phosphorylation (OXPHOS) levels in Mhy6-SOD2Δ v.s. wild type using a Seahorse XF analyzer. The rate of oxygen consumption per cells was significantly lower in Mhy6-SOD2Δ cardiomyocytes than that in wild type. The most noticeable difference in the O2 consumption was found in the presence of FCCP (H+ ionophore/uncoupler). Remarkably, while the FCCP treatment increased O2 consumption in wild-type, the treatment showed no effect on the O2 consumption in the Mhy6-SOD2Δ cardiomyocytes. The result indicated that the low basal OXPHOS activity in Mhy6-SOD2Δ was due to unusually low OXPHOS potential. We examined glycolysis in these cells by measuring extracellular acidification (ECAR) and the pattern exactly opposite to that of oxygen consumption rate (OCR) was observed for glycolysis rates between Mhy6-SOD2Δ and wild type.

020 Pediatric dilated cardiomyopathy hearts display a gene expression profile consistent with pluripotency and dedifferentiation Kathleen C. Woulfe1, Phillip D. Tatman1, Anis Karimpour-Fard1, Danielle A. Jeffrey1, Karin Nunley1, Matthew R. G Taylor1, Shelley D. Miyamoto1, Brian L. Stauffer1,2, Carmen C. Sucharov1, 1University of Colorado School of Medicine, Aurora, CO, USA, 2Denver Health and Hospital Authority, Denver, CO, USA

Background: Our previous work has shown several myocellular differences in pediatric and adult dilated cardiomyopathy (DCM). However, a thorough characterization of the molecular pathways involved in pediatric DCM does not exist, limiting the development of age-specific therapies. To better characterize this patient population, we investigated the transcriptome profile of pediatric patients. Methods: RNA-seq from 7 DCM and 7 non-failing (NF) explanted pediatric left ventricles (LV) was performed. Changes in gene expression were confirmed by RT-PCR in 36 DCM and 21 NF pediatric hearts, and in 20 DCM and 10 NF adult hearts. The degree of myocyte hypertrophy was investigated in 4 DCM and 7 NF pediatric hearts, and in 4 DCM and 9 NF adult hearts. Neonatal ventricular myocytes (NRVMs) were treated with pluripotency-inducing factors, and changes in gene expression were determined by RT-PCR. Results: Changes in gene expression were identified in cytokine signaling, signal transduction, and transcription. Interestingly, these changes were age- dependent and are associated with dedifferentiation and pluripotency in cardiac progenitor cells. Importantly, myocytes from adult DCM hearts showed an increase in cell size that was not observed in pediatric hearts. Furthermore, treatment of NRVMs with pluripotency-inducing factors recapitulated changes in gene expression observed in the pediatric DCM heart. Conclusions: Pediatric DCM is characterized by unique changes in gene expression that suggest maintenance of an undifferentiated state. These changes are accompanied by lack of myocyte hypertrophy.

021 Cardioprotection in the mouse heart: acute protective effects of an estrogen receptor agonist Anjali Ghimire1, Susan Howlett1, 1Dalhousie University, Halifax, Canada

Background: High calcium levels in ischemia promote contractile dysfunction and cell death in myocytes from aged or ovariectomized female mice. This suggests that low estrogen levels alter myocyte calcium homeostasis promoting ischemia and reperfusion (I/R) injury. As I/R injury occurs during cardiac surgery, we propose to determine whether the G-protein coupled estrogen receptor (GPER) agonist, G1, enhances the benefits of a cardioplegic solution designed to protect hearts from ischemic damage. Methods: Hearts were isolated from adult female mice (7-9 mos, n=10) for Langendorff perfusion experiments and perfused with Krebs-Henseleit buffer (37°C; 80 mmHg) for 15 min. Perfusion was then interrupted and St. Thomas’ 2 cardioplegic solution was delivered (7-9°C) either with G1 (110 or 500 nM), or with vehicle alone for 6 min. This was followed by 90 mins global ischemia (22-24°C) and 30 min reperfusion (37°C). Results: Post-ischemic functional recovery in hearts perfused with cardioplegia + G1 (500 nM) was significantly better than cardioplegia alone. Left ventricular developed pressure (LVDP) recovered to 53.0 ± 21.5% for control vs. 78.5 ± 11.3% for G1 (p<0.05). There was, however, no significant difference compared to control with a lower concentration of G1 (110 nM; LVDP=62.0 ± 2.3%). Similar results were seen when the rates of pressure development (+dp/dt) and decay (-dp/dt) were assessed. Values for recovery of +dp/dt were 55.8 ± 22.5% vs. 84.2 ± 12% and for –dp/dt were 56.3 ± 22.3% vs. 86.3 ± 10.6% for control and G1 (500 nM), respectively. By contrast, lower concentrations of G1 had no effect on +dp/dt or –dp/dt. Conclusion: These results demonstrate that addition of 500 nM G1 enhances cardioprotective properties of a standard cardioplegic solution and significantly improves functional recovery after ischemia in female mouse hearts. These results have the potential to improve outcomes during cardiac surgery.

022 The W792R mutation in cardiac myosin binding protein-C reduces the C6 FnIII domain stability and causes hypertrophic cardiomyopathy through haploinsufficiency Dan Smelter1, Willem de Lange1, J. Carter Ralphe1, 1University of Wisconsin, Madison, WI, USA

Cardiac myosin binding protein-C (cMyBP-C) is a functional sarcomeric protein that regulates contractility in response to contractile demand. Mutations in cMyBP-C that produce a truncated protein are postulated to cause hypertrophic cardiomyopathy (HCM) through a mechanism of haploinsufficiency. Missense mutations predicted to produce full-length cMyBP-C likely cause HCM through more complex mechanisms. To gain

Page 45: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

45

insight into the effect of cMyBP-C missense mutations on contractile function, we expressed the pathogenic W792R mutation in cMyBP-C-/- mouse cardiomyocytes and studied the functional effects using a three-dimensional engineered cardiac tissue (mECT) construct. Based on the predicted structure and residue charge change, we hypothesized that this mutation affects folding of the C6 FnIII-domain and causes destabilization of the mutant protein. Following adenoviral transduction of the wild-type (WT) and W792R cDNA, we achieved equivalent mRNA transcript abundance but could not achieve equivalent protein levels compared to WT controls (26±5% W792R vs. 100% WT, p<0.001). mECTs expressing W792R demonstrated significantly abnormal contractile kinetics compared to WT (time to peak force: 38.4±1.2ms vs. 44.6±1.1ms, p=0.003; time to 50% relaxation: 32.1±0.5ms vs. 41.0±1.4ms, p<0.001). We next studied whether common pathways of protein degradation are responsible for the rapid degradation of W792R cMyBP-C. Inhibition of the ubiquitin-proteasome pathway was unable to increase full-length mutant protein abundance to WT equivalence. Bacterial expression of the WT and W792R C6 domains demonstrated decreased stability of the mutant W792R C6 domain. These data suggest that the W792R mutation destabilizes the C6 FnIII domain of cMyBP-C resulting in decreased protein expression and causing disease through haploinsufficiency. The contractility data indicate a phenotype similar to complete loss of cMyBP-C with accelerated contractile kinetics and increased force amplitude. This study highlights the vulnerability of FnIII-like domains to mutations that alter their stability, and reveals that missense mutations in cMyBP-C can cause disease through a mechanism of haploinsufficiency.

023 A role for PPARα in sex differences in cardiac hypertrophy Natasha Fillmore1, Josephine Harrington1, Shouguo Gao1, Yanqin Yang1, Xue Zhang1, Poching Liu1, Andrea Stoehr1, Danielle Springer1, Jun Zhu1, Xujing Wang1, Elizabeth Murphy1, 1National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA

Heart failure remains a leading cause of death worldwide and treatment is complicated by sex differences in the development of this disease. While sex differences in cardiac hypertrophy are well documented, the mechanisms involved are poorly understood. The purpose of this study is to better understand the mechanisms that contribute to sex differences in cardiac hypertrophy. Male and female mice were treated with vehicle or Angiotensin II (AngII; 1.5 mg•kg-1•day-1) to induce cardiac hypertrophy and a systems biology analysis was performed on RNAseq data to identify pathways central to sex differences in cardiac hypertrophy. Cardiac hypertrophy was observed after 2 weeks of AngII and sex differences became apparent after 3 weeks. After 3 weeks of treatment ejection fraction (EF) in females was not different from control values (54% vs 56%). In males, however, EF dropped from 55% to 37%, which was significantly lower than EF in females after 3 weeks of treatment. RNA sequencing was performed on hearts and sex differences in mRNA expression at baseline and following hypertrophy were observed along with differences between baseline and hypertrophy within a sex. Sex differences in mRNA were substantial at baseline and reduced somewhat with hypertrophy, as the hypertrophic differences tended to overwhelm the sex differences. We selected genes that were significant for the sex-disease interaction, and mapped them to the protein-protein interaction network constructed using STRING data. This identified a network centered on PPARα. To examine the role of PPARα in sex differences in cardiac hypertrophy, we treated male and female mice with a PPARα inhibitor (GW6471; (4 mg•kg-1•day-1)) along with vehicle or AngII. The inhibitor blunted the development of hypertrophy in male hearts (+5% AngII+GW6471 vs +20% AngII), blocking sex differences in cardiac hypertrophy. These results suggest that PPARα contributes to sex differences in cardiac hypertrophy.

024 Deletion of the Z-disc Protein Enigma Homolog Depresses Cross-bridge Cycling Kinetics in Mouse Myocardium Zachery Gregorich1, Jitandra Patel1, Wenxuan Cai1, Rachel Heurer1, Ziqing Lin1, Richard Moss1, Ying Ge1, 1University of Wisconsin-Madison, Madison, WI 53705, USA

Background: The Enigma Homolog (ENH) protein is a member of the Enigma subfamily of the PDZ/LIM family of proteins. ENH isoforms are abundantly expressed in the heart and loss of protein expression results in cardiac dysfunction and the development of dilated cardiomyopathy. Despite the fact that ENH isoforms have been shown to bind and modulate the activity of a variety of kinases involved in signalling to contractile proteins, the function of ENH in cardiac muscle remains unclear. In this study, we sought to determine whether ENH can modulate cardiac contractile function, possibly by influencing contractile protein phosphorylation. Methods: Right ventricular trabeculae were isolated from the hearts of ENH+/+, ENH+/-, and ENH-/- mice, skinned, and subjected to mechanical measurements. SDS-PAGE analysis

was used to assess the expression of myosin heavy chain (MHC) isoforms and other major contractile proteins. Top-down proteomics was utilized to analyze contractile protein phosphorylation. Results: The steady-state mechanical properties of trabeculae isolated from ENH+/+, ENH+/-, and ENH-/-

mice did not differ, but a significant depression in cross-bridge cycling kinetics was observed in ENH+/- and ENH-/- trabeculae. While increased expression of β-MHC partially explains decreased cross-bridge cycling kinetics in ENH-/-

trabeculae, the relative expression of β-MHC was not different between ENH+/+ and ENH+/- trabeculae despite depressed cross-bridge cycling kinetics in the latter. The expression of other major contractile proteins were similar between trabeculae from ENH+/+, ENH+/-, and ENH-/- animals. Although the phosphorylation of cardiac troponins and the myosin regulatory light chain were significantly decreased in ENH-/- mouse myocardium, contractile protein phosphorylation in ENH+/- mouse myocardium was not significantly different from that in ENH+/+ mouse myocardium. Conclusions: Collectively, this information suggests that loss of ENH protein expression depresses cross- bridge cycling kinetics in mouse myocardium independent of changes in contractile protein expression or phosphorylation.

025 Postconditioning with H2S donors: effect on reperfusion-induced ventricular arrhythmias Qutuba G Karwi1,2, Matt Whiteman3, Mark Wood4, Gary Baxter1, 1School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK, 2College of Medicine, University of Diyala, Diyala, Iraq, 3Medical School, University of Exeter, Exeter, UK, 4School of Biosciences, University of Exeter, Exeter, UK

BACKGROUND: We have reported that slow-releasing and mitochondrial- targeted H2S donors limit infarct size when given prior to reperfusion following coronary occlusion in vivo. In order to assess potential anti-arrhythmic effects of H2S donors, we have undertaken a comprehensive analysis of electrocardiograms recorded during the early phase of reperfusion following H2S postconditioning in a rat infarct model. METHODS: Inactin®-anesthetized Sprague-Dawley rats underwent 30 min regional myocardial ischemia and 120 min reperfusion. Drug interventions were given 10 min before reperfusion. We used the following H2S donors and non-H2S releasing negative control compounds: GYY4137 and its control Depleted-GYY4137; the mitochondria targeting donors AP39 and AP123 and their respective controls AP219 and HTB. We also examined ADT-OH, the H2S releasing moiety of AP39. We also used ischaemic preconditioning (IPC, 2 x 3 min) as a positive control. Lead II electrocardiograms were examined to identify ventricular premature beats (VPBs), ventricular tachycardia (VT), sustained VT (>30 s) and ventricular fibrillation (VF) during the first 10 minutes of reperfusion in the presence and absence of H2S donors. RESULTS: Postconditioning with AP39 significantly attenuated the occurrence of VPBs during early reperfusion by 56% compared to the vehicle group (Figure.1,p<0.01) while GYY4137 and AP123 reduced the incidence of VPBs more modestly by 41% and 31%, respectively (not significant). AP39 also limited the incidence of reperfusion-induced sustained VT and VF by 69% and 67% respectively compared to the control (Table.1,p<0.05). None of the control compounds or AP123 produced any detectable effects on ventricular arrhythmias. CONCLUSION: These results show for the first time the potential inhibitory effect of selective mitochondrial delivery of H2S with AP39 (but not AP123) on reperfusion-induced ventricular arrhythmia in vivo. Characterising the mechanism of AP39’s anti-arrhythmic effect is the subject of ongoing investigation.

026 CypD-mediated regulation of the permeability transition pore is altered in mice lacking the mitochondrial calcium uniporter Randi Parks1, Sara Menazza1, Angel Aponte1, Paolo Bernardi2, Toren Finkel1, Elizabeth Murphy1, 1NHLBI, NIH, Bethesda, MD, USA, 2University of Padova, Padova, PD, Italy

Knockout (KO) of the mitochondrial Ca2+ uniporter (MCU) abrogates rapid mitochondrial Ca2+ uptake and permeability transition pore (PTP) opening. However, hearts from global MCU-KO mice were not protected from ischemic injury. Furthermore, MCU-KO hearts were resistant to protection by cyclosporin A (CsA), a cyclophilin D (CypD)-dependent pore desensitizer. This study investigates the hypothesis that the lack of protection in MCU-KO may be explained by alterations in PTP opening due to compensatory changes in CypD signaling. To investigate whether pore opening can occur in MCU-KO, Ca2+ uptake and swelling were measured in isolated mitochondria in the presence of the Ca2+ ionophore ETH129 to permit Ca2+ entry into the matrix. With ETH129, MCU-KO mitochondria took up Ca2+ and underwent pore opening similar to WT. To investigate the Ca2+ sensitivity of PTP in MCU-KO, basal Ca2+ was set to the same level in mitochondria from KO and WT prior to measuring Ca2+ uptake. MCU-KO underwent PTP opening before WT, suggesting that PTP Ca2+-sensitivity is altered in the absence of MCU.

Page 46: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

46

To determine whether CypD-mediated regulation of PTP may be different following global MCU deletion, experiments were performed to examine the interaction between CypD and the proposed PTP component ATP synthase. WT and MCU KO cardiac mitochondria were incubated with an immunocapture antibody to pulldown ATP synthase. Interestingly, results suggest that there was more CypD associated with ATP synthase in MCU KO in comparison to WT (n=7,P=0.047). As phosphorylation of CypD has been proposed to enhance PTP opening, immunoprecipitation experiments were performed using an antibody for phosphorylated proteins. MCU KO mitochondria had an increase in the amount of phosphorylated CypD (n=9,P=0.027). These results suggest that absence of MCU may alter PTP opening such that less Ca2+ is required to trigger PTP, which may be due to compensatory changes in CypD-mediated pore regulation.

027 Blocking/Knocking out Smad3 Alleviates Doxorubicin Effects on Endothelial Cells Jill Schriewer1, Melissa Cobb1, Meera Raghavan1, Eugene Konorev1, 1Kansas City University, Kansas City, MO, USA

Recent studies have shown that patients with early-stage cancers are now more likely to die from complications of anticancer therapy than cancer. Doxorubicin (Dox), a drug used to treat numerous cancers, is known to cause delayed cardiomyopathy. We have previously reported reduced microvascular density and increased levels of TGFbeta related transcripts in Dox treated mouse hearts. We hypothesized that microvascular remodeling in Dox treated hearts is mediated by enhanced TGFbeta/Smad3 signaling. We observed the inhibition of human umbilical vein endothelial cell (HUVEC) proliferation and suppression of the formation of vessel-like structures by Dox. TGFbeta pathway inhibitor SB431542 (SB), which selectively blocks Smad2/3 signaling, alleviated the effects of Dox in vitro. In aortic ring experiments, Dox decreased the number of sprouts in aortas from wild type mice while aortic preparations from Smad3 knockout animals were more resistant to the deleterious effect of Dox and produced increased numbers of sprouts. In experiments with co- cultures of cardiac fibroblasts and HUVEC, we identified the detachment of endothelial cells as an additional putative mechanism of microvascular remodeling by Dox. Live imaging of co-cultures treated with Dox in the presence of caspase-3/7 substrate revealed that detaching endothelial cells were not undergoing apoptosis prior to detachment. SB prevented detachment and increased the number of endothelial cells in co-cultures treated with Dox. Treatment with Dox led to the reduced adhesion of endothelial cells to laminin, a major constituent of the endothelial basement membrane while SB improved adhesion of Dox treated cells to laminin. In conclusion, Dox causes inhibition of sprouting angiogenesis in aortic rings and detachment of endothelial cells in an in vitro co-culture model of formation of vessel-like structures. These deleterious processes appear to be mediated by TGFbeta/Smad3 signaling as they were alleviated by both a pathway inhibitor and a genetic Smad3 knockout.

028 The contribution of fatty acid and ketone body oxidation to energy production increases in the failing heart and is associated with a decrease in cardiac efficiency Kim Ho1,2, Cory Wagg1,2, Liyan Zhang1,2, John Ussher2,3, Gary Lopaschuk1,2, 1Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada, 2Cardiovascular Research Centre, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada, 3Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada

Background: The failing heart is energy-starved and inefficient due to perturbations in energy metabolism. Since recent evidence suggests that ketone body oxidation increases in the failing heart as an adaptive mechanism to counteract reductions in fatty acid oxidation, our aim was to assess overall cardiac metabolism in heart failure, to establish what metabolic alterations contribute to reduced cardiac efficiency. Methods: C57BL/6J male mice (12 wk old) underwent either sham surgery, or transverse aortic constriction (TAC) surgery to induce pressure overload hypertrophy over a 4 wk period. Isolated working hearts from these mice were then perfused with appropriately 3H or 14C labelled glucose (5 mM), palmitate (0.8 mM), and ß-hydroxybutyrate (0.6 mM) to assess oxidative metabolism and glycolysis. Results: A 45% reduction in %EF was seen in intact TAC mice and a 54% decrease in cardiac work was seen in isolated working hearts from TAC mice. However, normal cardiac Krebs Cycle acetyl CoA production was seen compared to sham mice, reflecting a reduction in cardiac efficiency. Correspondingly, absolute glucose oxidation rates decreased in TAC compared to sham hearts, whereas absolute rates of fatty acid and ketone body oxidation were similar. However, normalization to cardiac work revealed that glucose oxidation was not depressed in failing hearts, although glycolysis was increased. Conversely, both fatty acid and ketone body oxidation increased in TAC hearts when

normalized to cardiac work. This increased reliance on fatty acid oxidation challenges the current dogma suggesting that fatty acid oxidation is depressed in the failing heart. Conclusion: In the failing heart, a decreased cardiac efficiency is associated with increases in the contributions of ketone body and fatty acid oxidation to energy production. The latter observation suggests that normalizing excessive fatty acid oxidation in the failing heart may be a novel approach to improve cardiac efficiency.

029 The role of Midkine in Children with Dilated Cardiomyopathy Xuan Jiang1, Kathleen Woulfe1, Anis Karimpour-Fard1, Keith Koch1, Brian Stauffer1,2, Shelley Miyamoto3, Carmen Sucharov1, 1University of Colorado- Denver, Aurora, CO, USA, 2Denver Health and Hospital Authority, Denver, CO, USA, 3Children’s Hospital Colorado, Aurora, CO, USA

Introduction: While current medical therapies benefit adult heart failure (HF) patients, these therapies do not confer the same improvement in survival in children with HF. This difference suggests that the underlying cellular mechanisms in failing myocytes are uniquely regulated. We have found that serum circulating factors are important in cardiomyocyte remodelling in pediatric patients with Dilated Cardiomyopathy (DCM). In our preliminary results, we identified a specific heparin-binding growth factor, midkine (MDK), which is highly upregulated in the serum of pediatric DCM patients. The objective of this study is to investigate the relation between serum MDK levels and clinical outcome, and to define the effect of MDK in modulating pathologic responses. Methods and results: Our current serum bank contains 5 non- failing (NF) and 44 DCM samples from individual patients. The level of serum (circulating) MDK was determined in the banked serum from pediatric DCM patients by ELISA, and correlated with the patient’s clinical outcome. Our results show that MDK levels in serum was elevated in pediatric DCM patients who died or were transplanted in comparison with pediatric DCM patients who are clinically stable. The contribution of serum MDK to primary cardiomyocytes was investigated by treating neonatal rat ventricular myocytes (NRVMs) with human recombinant MDK for 72 hours. Our results show an increase in the expression of genes associated with pathologic remodelling in MDK-treated NRVMs. In addition, there is an increase in cell area and ANF expression in MDK-treated NRVMs compared to NF serum-treated cells. In our unique mouse model of pediatric cardiac pathologic growth (isoproterenol infusion –30mg/kg/day), MDK treatment (3mg/kg/week) resulted in increased heart/body weight. Conclusions: Our results suggest that MDK is detrimental to cardiac function. This finding combined with increased circulating MDK in end-stage pediatric DCM patients, suggests an important regulatory role for MDK in pediatric HF.

030 Compartmentalized Gαq-Signaling in Adult Cardiac Myocytes Erika Dahl1, Steven Wu1, Chastity Healy1, Timothy O'Connell1, 1University of Minnesota, Minneapolis, MN, USA

Gαq-coupled G-protein receptors (Gq-receptors), including α1-adrenergic and angiotensin receptors (α1-AR, AT-R), activate cardiac signaling pathways controlling hypertrophy, cell survival/death, and inotropy. Early studies in cell and animal models established the convention that Gq-signaling exacerbates heart failure (HF). However, clinically, AT-R antagonists improve HF outcomes, whereas α1-AR antagonists worsen HF, suggesting Gq-receptors are functionally unique. Our work indicates that α1-ARs are unique cardiac Gq-receptors by inducing cardioprotection and localizing to the nucleus in adult cardiac myocytes, suggesting that subcellular localization might form the mechanistic basis for unique Gq-receptor function. Here, we test the hypothesis that α1-ARs and AT-Rs localize to and initiate proximal signaling in distinct subcellular compartments. Using a novel fluorescent α1-ligand that improves the dynamics of defining receptor localization, we detected α1-ARs only at the nucleus in adult cardiac myocytes and isolated nuclei. Conversely, using a fluorescent AT-R ligand, we detected AT-Rs at the sarcolemma and the t-tubules. To measure proximal Gq-receptor signaling, we employed a fluorescent probe, GFP-PHD, that binds phosphatidylinositol-4,5- bisphosphate (PIP2) in the membrane and is released when phospholipase Cβ (PLCβ) cleaves PIP2. In adult cardiac myocytes, GFP-PHD localized to the sarcolemma and t-tubules, but not the nucleus, consistent with previous reports. Angiotensin, but not phenylephrine, induced movement of GFP-PHD off of the sarcolemma and t-tubules. The failure to detect GFP-PHD at the nucleus indicates that either PIP2 is excluded from nuclear membranes or that the probe fails to target the nucleus. Addition of a nuclear localization sequence to GFP-PHD (NLS-GFP-PHD) produced nuclear localization of GFP-PHD suggesting the presence of PIP2 in nuclear membranes in adult cardiac myocytes. Phenylephrine, and to a lesser extent, angiotensin induced movement of NLS-GFP-PHD off the nuclear membrane. In summary, our data demonstrate that α1-ARs and AT-Rs localize to and initiate proximal signaling in unique subcellular compartments.

Page 47: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

47

031 Endogenous HAX-1 Regulates SERCA Activity and Oxidation Dependent Stability Philip Bidwell1, Guan-Sheng Liu1, Narayani Nagarajan2, Kobra Haghighi1, George Gardner1, Junichi Sadoshima2, Evangelia Kranias1, 1University of Cincinnati, Cincinnati, OH, USA, 2Rutgers New Jersey Medical School, Newark, NJ, USA

HS-associated protein X-1 (HAX-1) has been identified as a novel binding partner of both SERCA2a and phospholamban (PLN) in the heart. We observed that the expression levels of HAX-1 are decreased in human failing hearts and experimental ischemia/reperfusion injury. To elucidate the impact of such reduction in endogenous HAX-1, a mouse model with inducible cardiac-specific ablation of HAX-1 was generated. Ablation of HAX-1 resulted in a hypercontractile phenotype in whole hearts and isolated cardiomyocytes. The underlying mechanisms included enhanced Ca-transient kinetics and SR Ca uptake affinity through relief of the PLN inhibitory effects. Despite the increases in Ca-cycling, HAX-1 ablation diminished contractile recovery and increased infarct size and apoptosis in hearts subjected to ex vivo ischemia/reperfusion. This was associated with enhanced degradation of SERCA2a by the calcium sensitive protease, calpain. Modulation of SERCA2a degradation by HAX-1 was also observed in isolated cardiomyocytes and SR microsomes. The ROS scavenger thioredoxin1 in cardiomyocytes and reducing conditions in SR microsomes attenuated degradation of SERCA2a and abolished the effects of HAX-1 deficiency, suggesting the involvement of a redox mechanism. Indeed, SERCA2a oxidation was assessed by two complementary free cysteine labelling methods and was found to be uniquely increased in HAX-1 deficient hearts. The SERCA2a inhibitor thapsigargin, which stabilizes a specific conformation of the enzyme, eliminated the HAX-1 variations in free cysteine labelling of SERCA2a. This suggests that HAX-1 alters the conformation of SERCA2a thereby changing the reactivity/availability of specific cysteines to oxidation. Additionally, HAX-1 levels inversely correlated to ROS production at the ER/SR compartment, quantified by genetically encoded HyPer indicators. These findings indicate that endogenous HAX-1 prevents SERCA2a cysteine oxidation, by a change in structure and/or a change in ROS, resulting in decreased susceptibility of SERCA2a to degradation. Thus, restoring the decreased HAX-1 levels in the stressed heart may hold therapeutic potential.

032 Estrogen-independent female resilience in dystrophic models of heart failure Tatyana Meyers1, Jackie Heitzman1, Lauren Aufdembrink1, Aimee Krebsbach1, DeWayne Townsend0, 1University of Minnesota, Minneapolis, USA

Background: Heart failure is a leading cause of death in both men and women. Female protection from heart disease is clearly documented in the literature, but its basis is not known. A major hurdle is understanding the role of sex hormones. While estrogen is clearly linked to important aspects of female protection from ischemic heart disease, post-menopausal women continue to show prolonged survival with severe heart failure. This indicates that protective estrogen-independent mechanisms significantly contribute to female resilience. Here we present evidence for striking estrogen-independent female resilience in a dystrophic mouse model of heart failure. Methods: To generate a heart failure model that combines genetic vulnerability with an adrenergic trigger, 10 mg/kg isoproterenol (Iso) injections were used to exacerbate the cardiac phenotype in β-sarcoglycan-deficient (β-SG-/-) mice. Delivery of 14 Iso injections over 5 days produced 89% mortality in β-sg-/-

male mice, but no deaths among female mice. Following the challenge, male hearts displayed extensive myocardial uptake of endogenous IgG, while female hearts showed a 4.5-fold reduction in acute myocyte injury. Ovariectomy did not reduce survival in female mice, indicating that their survival benefit does not depend on ovarian hormones. Male and female dystrophic hearts exhibited similarly high levels of myocyte damage 1 day after receiving a single dose of Iso. However, 1 month after one dose of Iso, males suffered 25% mortality and their hearts displayed extensive fibrosis and ventricular dilation, while female hearts showed normal geometry and comparatively reduced fibrosis. Conclusions: These results demonstrate estrogen-independent female protection from acute mortality after induction of heart failure, accompanied by resilience to long-term pathological remodeling following cardiac injury. The magnitude of these sex differences suggests that estrogen-indpendent mediators of cardiac resilience may be a significant source of clinically-observed female protection, and a promising target for future therapies.

033 Contactless Particle Image Velocimetry (PIV) Method of Screening Drugs Using Human iPSC-derived Cardiomyocytes Sheeja Rajasingh1, Andras Czirok1, Dona Greta Isai1, Saheli Samanta1, Zhigang Zhou1, Buddhadeb Dawn1, Johnson Rajasingh1, 1University of Kansas Medical Center, Kansas City, USA

Introduction: The development of cardiac arrhythmias as a pharmacological side effect has become the single most common cause of the withdrawal or restriction of previously marketed drugs. Currently available drug screening models are not efficient in detecting cardiac toxicity. Here we demonstrate in vitro Particle Image Velocimetry (PIV) assays followed by fourier spectrum and heatmap analyses for measuring the impact of different cardiac drugs on the rate and frequency of beating cardiomyocytes (CMCs). Hypothesis: Our PIV method of measuring CMCs contractility could be beneficial for drug testing, preclinical screening to detect cardio toxicity without jeopardizing the biology of cells. Methods and Results: The novelty of our system is that we can record the spatiotemporal maturation of human iPSC-derived CMCs at specific positions. We have evaluated the contractility of CMCs with the response to various chemical compounds such as Verapamil (Ca2+ channel blocker), Isoproterenol (Ca2+ channel activator), E-4031 (K+ channel inhibitor) and NS-1643 (K+ channel activator). Our data shows that the chronotropic effect of verapamil and E-4031 at the concentration of 1 µM and 500 nM respectively, has significantly reduced the beating frequency. Similarly, the chronotropic effect of Isoproterenol and NS-1643 at the concentration of 1 µM and 60 nM respectively, has meaningfully increased the beating frequency. Moreover, our fourier spectrum and heatmap analyses further confirmed the spatiotemporal changes occur due to the addition of chronotropic drugs. Conclusions: We report for the first time the applications of PIV assays for prospective cardiac drug efficacy screening by measuring the contractility of CMCs from the video image without jeopardizing the biology of cells.

034 Influence of an ACE inhibitor on frailty and cardiac function in old male C57BL/6 mice Alice Kane1, Kailtyn Keller1, Susan Howlett1, 1Dalhousie University, Halifax, NS, Canada

Background. Frailty is the accumulation of health deficits over the lifespan. Frailty results in increased risk of adverse outcomes including poor cardiovascular outcomes. Angiotensin converting enzyme (ACE) inhibitors improve exercise capacity in older adults without cardiovascular disease, and could be a potential frailty intervention. The aim of this study was to investigate whether chronic treatment with the ACE inhibitor enalapril would attenuate frailty in mice, and whether this occurs through changes to cardiac function. Methods. Male C57BL/6 mice (17 months) were fed enalapril containing chow (40 mg/kg/day; n=25) or control feed (n=13). After 6 weeks blood pressure (BP) was measured with a tail-cuff. Once the mice were 24 months old (7 months treatment), frailty was quantified with a validated mouse frailty index and in vivo cardiac function was measured using echocardiography. Results. Frailty index scores were significantly lower in the enalapril group than the control group after 7 months (0.24 ± 0.01 vs 0.29 ± 0.01, p<0.001). Enalapril treatment also resulted in lower systolic BP (85.0 ± 10.5 vs 98.1 ± 12.3 mmHg, p<0.01), with a trend towards decreased diastolic BP (65.4 ± 4.0 vs 75.8 ± 3.9 mmHg, p=0.08). Echocardiography showed changes to the left ventricle (LV) of the enalapril treated mice compared to control, including a thinner LV anterior wall in systole (1.38 ± 0.08 vs 1.60 ± 0.05 mm, p<0.05), smaller LV mass (127.2 ± 9.6 vs 180.7 ± 14.4 mg, p<0.01) and reduced stroke volume (36.5 ± 2.4 vs 48.5 ± 3.6 µl, p<0.01) and cardiac output (18.3 ± 1.2 vs 23.2 ± 1.7 ml/min, p<0.05). Conclusion. Enalapril treatment attenuated frailty in old male mice. As expected it also resulted in reduced blood pressure. Cardiac mechanisms contributing to the attenuation of frailty may include changes to the structure and function of the heart.

035 Transient Receptor Potential Vanilloid 1 Mediates Laporotomy and Opioid-induced Infarct Size Reduction in Rats Eric Gross1, Yun Wu1, Helen Heymann1, Garrett Gross2, 1Stanford University, Stanford, CA, USA, 2Medical College of Wisconsin, Milwaukee, WI, USA

Background: In light of an opioid epidemic in the United States, physicians are increasingly prescribing non-opioid analgesics. However, newer pain medications such as transient receptor potential vanilloid 1 (TRPV1) inhibitors may impede organ protection. We examined in rodents whether a laparotomy- and morphine-induced infarct size reduction is mediated by TRPV1. Further, we tested if inhibiting TRPV1 interferes with laparotomy- or morphine-induced cardioprotection. Methods: Male Sprague-Dawley rats were subjected to 30

Page 48: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

48

minutes of coronary occlusion followed by 120 minutes reperfusion. Before ischaemia, a laparotomy with or without application of capsaicin cream (0.1%, a TRPV1 activator), was performed. Additional sets of rats were given morphine (3 mg/kg) with or without capsaicin. Further, capsazepine (3 mg/kg, a classical TRPV1 inhibitor), and P5 (3 mg/kg, a peptide analgesic and TRPV1 inhibitor), were given prior to the laparotomy or morphine. Myocardial infarct size was determined. Results: A laparotomy or capsaicin reduced infarct size versus controls (to 44±2%* or 49±1%* vs. 66±1%, infarct size/area at risk %, n=6, *P<0.0001). When combining laparotomy and capsaicin, cardioprotection was not additive. Morphine also reduced infarct size (37±3%* vs. 61±2%, n=6, P<0.0001), with no additive effect when combined with capsaicin. The TRPV1 inhibitors capsazepine and P5 abolished cardioprotection induced by either a laparotomy (58±1%+ and 65±2%+ vs. 44±2%+, respectively) or morphine (62±3%+ and 58±2%+ vs. 37±3%, respectively, n=6, +P<0.0001). Conclusions: Infarct size reduction by a laparotomy or morphine is blocked by inhibiting TRPV1. Drugs given which inhibit TRPV1 may impede organ protection and this requires further study.

036 Circulating Factors Contribute to PDE5-Mediated Pathological Myocardial Remodeling in Single Ventricle Congenital Heart Disease Anastacia Garcia1,2, Stephanie Nakano1,2, Anis Karimpour-Fard1, Brian Stauffer1,3, Carmen Sucharov1, Shelley Miyamoto1,2, 1University of Colorado, Denver Anschutz Medical Campus, Aurora, CO, USA, 2Children’s Hospital Colorado, Aurora, CO, USA, 3Denver Health and Hospital Authority, Denver, CO, USA

Introduction: Single ventricle congenital heart disease (SV) is fatal without intervention, and maladaptive myocardial remodeling and eventual heart failure are a major cause of morbidity and mortality in this population. Our previous and current studies demonstrate gene expression changes indicative of pathological myocardial remodeling and increased phosphodiesterase-5 (PDE5) expression and activity in SV myocardium relative to non-failing (NF) myocardium. Interestingly, Sildenafil, a PDE5 inhibitor (PDE5i), is increasingly utilized in SV patients, to target the pulmonary vasculature. However, our data suggest potential myocardial effects of PDE5i. We are utilizing a cell-based model (neonatal rat ventricular myocytes, NRVMs) in combination with an ongoing human heart and blood bank to determine if PDE5 has a role in the induction of pathological gene expression changes seen in SV myocardium. Methods: PDE5 levels in NF and SV sera were measured using an ELISA. NRVMs were treated for 72 hours with sera or serum-derived exosomes from NF and SV patients +/- sildenafil. qRTPCR was performed for the targets of interest and RNAseq was performed on a subset of serum-treated NRVMs +/- sildenafil. Results: Circulating levels of PDE5 are significantly higher in SV patients compared to NF controls. Additionally, treatment of NRVMs with serum or exosomes from SV patients recapitulates the pathologic gene expression pattern seen in the human SV myocardium, and PDE5i blunts this response. Further, transcriptome analysis of serum-treated NRVMs +/- sildenafil revealed that SV circulating factors significantly impact the expression of 1,449 genes involved in multiple cellular processes including fibrosis, hypertrophy, lipid metabolism, and cell death, and PDE5i reverses the expression of 65% (1088) of these genes. Conclusion: Together, these data suggest that SV circulating factors contribute to PDE5-mediated myocyte remodeling, and in addition to effects on the pulmonary vasculature, PDE5i may be a direct myocardial target of therapy for the treatment of SV.

038 Function Beyond RNA Splicing for RBFox Family Members in Heart Chen Gao1,2, Jing Hu3, Chaoliang Wei4, Yunhua Esther Hsiao5, Shuxun Ren1,2, Yuanchao Xue3, Yu Zhou3, Jianlin Zhang6, Ju Chen6, Xinshu Xiao5, Xiang-Dong Fu3,5, Yi Xing7, Yibin Wang1,2, 1Division of Molecular Medicine, Cardiovascular Research Laboratories, University of California, Los Angeles, Los Angeles, California, USA, 2Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA, 3Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, California, USA, 4Department of Cell Biology and Medical Genetics, School of Medicine, Shenzhen University, Shenzhen, China, 5Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California, USA, 6Department of Medicine, University of California, San Diego, San Diego, California, USA, 7Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA

Background: RNA metabolism, from synthesis, processing, translation to degradation is an integrated part of gene regulation that ultimately determines the overall cardiac transcriptome complexity. The RBFox family members of RNA splicing regulator, including RBFox1 and 2, are dramatically downregulated in the diseased hearts and are found to have critical roles in cardiac development and pathological remodeling. In addition to expected

alternative RNA splicing and transcription regulation targeted by the nuclear RBFox1 and RBFox2, both genes also produce cytosolic localized splicing variants, raising interesting questions about additional roles in the cytosol for RNA processing. Goal and Methods: This study investigated the functional impact of two RBFox family members- RBFox1 and RBFox2 in cytosol in cardiomyocytes. Results: Cardiac specific KO of RBFox1 exacerbated pressure-overload induced heart failure and fibrotic remodeling while cardiac specific expression of the RBFox1 cytosolic isoform (RBFox1c) in mouse heart was sufficient to protect against pressure-overload induced cardiac dysfunction and fibrotic remodeling. RNA-seq analysis in cultured myocytes revealed a significant suppression of proinflammatory genes upon RBFox1c expression, and motif analysis identified significant enrichment of RBFox binding sites in the 3’UTR of the affected genes mRNA. By both mass-spec and co-IP analysis, we also detected specific interaction between RBFox1c and RNA degradation complex protein, implicating a molecular link between RBFox1c and targeted RNA degradation. For RBFox2, cardiac specific and inducible KO of RBFox2 in adult mouse hearts was sufficient to trigger dilated cardiomyopathy with disorganized T-tubules structure associated with targeted inhibition of Jph2 expression at protein level but not at mRNA level. Loss of RBFox2 repressed Jph2 protein expression due to competitive binding on the Jph2 3’-UTR with miR34a. Inactivating miR34a partially rescued heart failure induced by RBFox2 KO in heart by restoring Jph2 expression, normalizing cardiac function and T-tubule organization. Conclusion: Both RBFox1 and RBFox2 have potentially important function beyond splicing regulation in cardiac muscle cells. In cytosol, RBFox1 suppresses inflammatory gene expression and cardiac fibrosis by binding to 3’UTR, and potentially modulating target mRNA stability. In contrast, RBFox2 alters Jph2 protein expression by interacting with Jph2 3’UTR and competing with miR34a mediated gene suppression. Therefore, loss of RBFox1 and 2 in diseased heart contribute to different features of cardiac pathology via both nuclear and cytosolic function at different levels of RNA metabolism.

039 Enhanced Activation of Inflammasome Promotes Atrial Fibrillation Chunxia Yao1, Larry Scott Jr.1, Tina Veleva2, Frank U. Müller3, Stanley Nattel4, Dobromir Dobrev2, Xander H.T. Wehrens1, Na Li1, 1Baylor College of Medicine, Houston, TX, USA, 2University Duisburg-Essen, Essen, Germany, 3University of Münster, Münster, Germany, 4Montreal Heart Institute, Montreal, QC, Canada

Background: Inflammation has been associated with the progression of atrial fibrillation (AF) in patients, while the underlying molecular mechanisms remain elusive. It is known that a large multi-protein complex called the NACHT, LRR & PYD Domains-containing Protein 3 (NLRP3) inflammasome can activate caspase-1 and promote interlukin-1beta (IL-1β) release. Preliminary studies revealed NLRP3 activity is enhanced in atrial tissues of patients with either paroxysmal or long-lasting persistent AF, suggesting that AF could be influenced by NLRP3 inflammasome activation. However, the causal role of NLRP3 inflammasome in the pathogenesis of AF remains elusive. In this study, we tested the hypothesis that constitutive activation of NLRP3 in cardiomyocytes (CMs) promotes AF. Methods and Results: CM-specific knock-in mouse model expressing a gain-of-function mutation NLRP3 A350V (Myh6Cre;Nlrp3A350V/+, CM-KI) was established. At 3 months, 24-hour telemetry ECG recordings revealed 100% of CM-KI mice (n=5) developed premature atrial contractions (PACs), whereas only 25% of controls (n=4, P<0.05) had PACs. Moreover, CM-KI mice were more susceptible to pacing-induced AF (88.9%, n=9) than controls (20%, n=5, P<0.05). Echocardiography showed that the ventricular function in CM-KI was mildly reduced relative to control. Compared to the controls, CM-KI mice exhibited atrial hypertrophy, abnormal Ca2+-release via ryanodine receptor 2 (RyR2), and shortening of atrial effective refractory period and action potential duration, which were associated with upregulation of Mef2c, Ryr2, Kcna5, Kcnj 3 and Kcnj5. Importantly, genetic inhibition of NLRP3 prevented the onset of spontaneous AF and atrial remodelling in CREM-Tg mice (a murine model of spontaneous AF). Conclusion: Our data reveal that CM-specific activation of NLRP3 promotes both electrical and structural remodelling, permissive to the AF development. In addition to its canonical function, NLRP3 inflammasome may exhibit alternative function in regulating gene transcription. Our study establishes a mechanistic link between the inflammatory signalling and the pathogenesis of AF.

040 Mechano-chemo-transduction is attenuated in a rabbit model of heart failure Rafael Shimkunas1, Bence Hegyi1, Zhong Jian1, Zana Coulibaly1, Kit S. Lam1, Kenneth S. Ginsburg1, Julie Bossuyt1, Donald M. Bers1, Leighton T. Izu1, Ye Chen-Izu1, 1University of California, Davis, Davis, CA, USA

Page 49: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

49

Background: Excessive mechanical loading under pathological conditions such as hypertension, infarction and fibrosis can lead to arrhythmias and heart failure (HF). Important knowledge gaps exist, however, in the mechanisms of mechano-chemo-transduction (MCT) at cellular and molecular levels. Using our innovative Cell-in-Gel system to mechanically load single cardiomyocytes, we investigated MCT in healthy and HF hearts. Methods: Intact ventricular myocytes from healthy and pressure and volume overload-induced HF rabbits were embedded in crosslinked 3D hydrogels. Cytosolic and sarcoplasmic reticulum (SR) calcium concentrations were measured with Fura-2 and Fluo- 5N fluorescence, respectively. Cell contraction was measured by sarcomere length shortening. Cells in-gel were compared with load-free cells in solution. Results: Cardiomyocytes from healthy hearts contracting in-gel under mechanical load showed significantly increased systolic calcium transients than load-free cells (Fura-2 ratio 2.71 ± 0.07 and 2.08 ± 0.04, respectively), suggesting a compensatory MCT mechanism transduces mechanical load to increase calcium transients and enhance contractility. Furthermore, SR calcium content and fractional release were larger in-gel, demonstrating augmented SR calcium cycling in-gel. Inhibiting NO synthases (NOS) with 1 mM L-NAME attenuated SR calcium and cytosolic calcium transient in-gel to nearly load-free, revealing the importance of NOS-NO signaling in mediating MCT. Cells from HF hearts, however, exhibited severe reduction in cytosolic calcium transients in-gel and load-free, which was consistent with a similar reduction in SR fractional release. NOS inhibition did not affect calcium cycling, indicating MCT was reduced in HF. Attenuation of systolic calcium impaired cell contraction in HF. Conclusions: Our study elucidates the important MCT mechanism in healthy cells enhances systolic calcium to autoregulate contractility in response to increased mechanical loading. NOS inhibition diminishes MCT, while having no effect in HF, revealing MCT pathways in cardiomyocytes are significantly attenuated in HF. These results provide a mechanistic explanation of the Anrep effect and its impairment in disease.

041 Protein tyrosine phosphatase 1B is a regulator of microRNA-mediated gene silencing and cardiac hypertrophy Benoit Boivin1,2, 1SUNY Polytechnic Institute, Albany, New York, USA, 2Montreal Heart Institute, Montreal, Quebec, Canada

Transcriptional control of protein synthesis during cardiac hypertrophy is complex and protein expression levels do not reflect the rate of transcription of the corresponding genes: while the overall quantitative change in mRNA transcription occurs as a direct consequence of chronic stress on the heart, qualitative gene reprogramming is also tightly controlled by miRNA-mediated gene silencing. We have previously shown that the inactivating phosphorylation of Argonaute 2 (Ago2) on tyr-393 was an important switch regulating the loading of miRNAs onto Ago2 and shaping miRNA-mediated post-transcriptional regulation. Since we know that PTP1B regulates Ago2 tyr- 393 phosphorylation, we explored whether PTP1B activity was altered in hypertrophic hearts. We found that PTP1B was reversibly oxidized and that its substrate, Ago2, was phosphorylated on tyr-393 in hearts subjected to pressure-overload (PO). In order to study the role of PTP1B in miRNA- mediated gene silencing in hearts, we generated cardiomyocyte-specific PTP1B knockout (PTP1B CKO) mice. Subjecting PTP1B CKO mice to PO caused severe systolic and diastolic dysfunction when compared to control mice subjected to PO for the same period. Interestingly, characterization of several markers of hypertrophy revealed that the increased expression of β- MHC observed in control mice subjected to PO was compromised in PTP1B CKO-PO mice. Since miR-208 is known to regulate thyroid hormone- associated protein 1 (THRAP1) expression and Thyroid Receptor β1 (TRβ1)- mediated repression of β-MHC, we investigated whether this pathway was activated as a consequence of PTP1B inactivation. We found decreased association between THRAP1 mRNA and Ago2, and increased THRAP1 expression in PTP1B CKO-PO hearts, despite elevated expression of miR- 208. Finally, inhibiting this pathway in vivo by inducing hypothyroidism, rescued PTP1B CKO-induced cardiac dysfunction. We propose a novel mechanism whereby Ago2 inactivation and defective silencing of THRAP1 contributes to cardiac hypertrophy and heart failure upon ROS-mediated inhibition of PTP1B.

042 The impact of age and frailty on ventricular structure and function in C57BL/6 Hirad Feridooni1, Alice Kane1, Omar Ayaz1, Ali Boroumandi2, N Polidovitch2, Robert Tsushima2, Robert Rose1, Susan Howlett1, 1Dalhousie University, Halifax, Canada, 2York University, Toronto, Canada

Background: On average, cardiac hypertrophy and contractile dysfunction increase with age. Still, individuals age at different rates and their health status varies from fit to frail. We investigated the influence of frailty on age-

dependent ventricular remodeling. Methods: Frailty was quantified as deficit accumulation in adult (≈7 mos) and aged (≈27 mos) C57BL/6 mice by adapting a validated frailty index (FI) tool. Hypertrophy and contractile function were evaluated in Langendorff-perfused hearts; cellular correlates/mechanisms were investigated in ventricular myocytes. Results: FI scores increased with age. Mean cardiac hypertrophy increased with age, but values in the adult and aged groups overlapped. When plotted as a function of frailty, hypertrophy was graded by FI score (r=0.67-0.55, p<0.0003). Myocyte area also correlated positively with FI (r=0.34; p=0.03). Left ventricular developed pressure (LVDP) plus rates of pressure development (+dP/dt) and decay (-dP/dt) declined with age and this was graded by frailty (r=-0.51, p=0.0007; r=-0.48, p=0.002; r=-0.56, p=0.0002 for LVDP, +dP/dt and -dP/dt). Smaller, slower contractions graded by FI score were also seen in ventricular myocytes. Contractile dysfunction in cardiomyocytes isolated from frail mice was attributable to parallel changes in underlying Ca2+ transients. These changes were not due to reduced sarcoplasmic reticulum stores, but were graded by smaller Ca2+ currents (r=-0.40, p=0.008), lower gain (r=-0.37, p=0.02) and reduced expression of Cav1.2 protein (r=-0.68, p=0.003). Conclusions: These results show that cardiac hypertrophy and contractile dysfunction in naturally-ageing mice are graded by overall health and suggest that frailty, in addition to chronological age, can help explain heterogeneity in cardiac ageing. This model system can ultimately be used to test interventions designed to improve cardiovascular outcomes in frail older adults.

043 Oroxylin A reduces angiotensin II- induced hypertrophy and mitochondrial dysfunction by activating sirtuin 3 in cardiac myocytes Niria Treviño Saldaña2,3, Gerardo de Jesús García Rivas2,3, Luz Leticia Elizondo Montemayor2,3, 1Centro de Investigación Biomédica de la Escuela Nacional de Medicina, Monterrey, Nuevo León, Mexico, 2Instituto de Cardiología y Medicina Vascular del Centro Médico Zambrano Heullion, Monterrey, Nuevo León, Mexico

Cardiac hypertrophy and fibrosis occur in non-ischemic ventricular dysfunction, and angiotensin II (AngII) is a key molecule targeting myocytes and fibroblasts, and promoting both events by activating the ROS dependent pathway. Oroxylin A (OA) is a natural flavone derived from the root of Scutellaria Baicalensis, with previously reported antioxidant and anti- inflammatory properties in hepatic and pulmonary cells, but little is known about its effect on a cell model of cardiac hypertrophy. Here we show that OA blocks Angiotensin II (Ang II) induced hypertrophy in cardiac myocytes (H9c2), protecting them from a 50% increase in cell size. Cardiac hypertrophy was associated with 2-fold increase in mitochondrial acetylation, which was abolished by pre-treatment with OA. Furthermore, when we evaluated the opening of the mitochondrial permeability transition pore (mPTP), pre- treatment with OA protected cardiac myoblasts against Ang II induced mPTP opening. Remarkably, acetylated cyclophilin D is a positive modulator of the mPTP, and OA reduced its acetylation with a concomitant increase in sirtuin 3 activity. Our data suggests that OA acts as an activator of sirtuin 3, capable of protecting cardiac cells against hypertrophy, mitochondrial dysfunction, and cell death. Future perspectives of the present work include the evaluation of OA in an in vivo model of cardiac disease such as heart failure, which would better characterize the potential cardioprotective effects of this phytochemical.

044 The Roles of Dopamine Receptor 3 in Age- and Sex-dependent Left Ventricular Remodeling Gabriel Grilo1, Patti Shaver1, Stefan Clemens1, Lisandra de Castro Brás1, 1East Carolina University, Greenville, NC, USA

Background: With a growing aged population the prevalence of chronic and degenerative pathologies is much increased and aging is a well-established independent risk factor for cardiovascular disease. With age, the levels of the neuromodulator dopamine (DA) decline supporting a role for DA in age- related disorders. While DA roles in the pathophysiology of hypertension is well studied, much less is known on how DA signalling affect cardiac aging. We have previously shown that 2 month old (m.o.) mice with a dysfunctional DA receptor 3 (DRD3) display cardiac fibrosis comparable to those of 24 m.o. wild type (WT) mice. The goal of this study is to assess DA signalling in the left ventricle (LV) throughout the life span of WT and DRD3 knockout (D3KO) mice and compare cardiac function and LV remodelling to identify age- and sex-dependent parameters. Methods: We assessed cardiac function of male and female mice (WT and D3KO) by echocardiography at the ages of 3, 6, and 12 m.o. (n=6/sex/age/genotype). At each time point, plasma and LV were collected to assess collagen turnover, fibrosis, and expression of DA receptors. Results: Protein levels of the excitatory D1-like receptors (DRD1 and DRD5) were increased in 12 m.o. WT mice compared to 6 m.o. WT, whereas the inhibitory receptors DRD2 and DRD3 were reduced. These data show for the first time that myocardial expression of DA receptors changes

Page 50: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

50

with age towards an excitatory pathway. Echocardiography showed marked differences in the LV chamber volumes and wall thicknesses between both genotypes. Three m.o. D3KO mice presented reduced LV anterior wall (LVAW) thickness and internal diameter (LVID), both at diastole and systole, compared to WT counterparts. These differences were mostly driven by the male animals. Females showed significant reduced LVAW but not LVID. Additionally, end-diastolic and end-systolic volumes (EDV and ESV) were reduced in male D3KO compared to WT. At 6 m.o., females displayed reduced LVIDd, LVIDs, EDV, and ESV in D3KO vs WT, but not males. At 12 m.o. the majority of sex differences were blunted but D3KO continued to show reduced chamber volumes and diameters. As expected, cardiac fibrosis showed an upwards trend in the WT animals with age. This increase was exacerbated in the D3KO mice and was most marked in the male animals. Conclusion: Our data indicates a role for DA signalling in LV function in mice, particularly for the DA receptor D3, and cardiac DA roles are age- and sex- dependent.

045 In vivo reduction of mitochondrial oxidative stress abolishes spontaneous arrhythmic sudden cardiac death (SCD) in non-ischemic heart failure (HF) Swati Dey1, Deeptankar DeMazumder1, Brian O'Rourke1, 1Johns Hopkins

University, Baltimore, MD, USA

INTRODUCTION: Reactive oxygen species are central to SCD pathophysiology, but their effects depend on where and how ROS are generated. Insight into this has been limited by the lack of adequate experimental methods. Employing innovative approaches, we tested the hypothesis that mitochondrial ROS (mROS) is a principal source of oxidative- stress in HF and in vivo reduction of mROS mitigates SCD. METHODS: In a unique guinea pig model that mimics human non-ischemic HF with a high incidence of SCD, we performed a randomized 2x2 crossover-study of vehicle (v) and MitoTEMPO (M) therapies (vM/Mv). In the untreated HF-model, we used genetically encoded ratiometric redox sensors targeted to the cytosol (cROS) or mitochondria of cardiomyocytes. In isolated left ventricular (LV) myocytes, we quantified ROS dynamics during field stimulation (1 Hz, 37ᴼC) in the presence or absence of MitoTEMPO, apocynin, angiotensin II (AngII) and AngII-receptor-blockers (ARB). RESULTS: Over 4 weeks, 65% of the HF animals experienced SCD. Treatment with MitoTEMPO abolished SCD and LV dysfunction. Crossover to MitoTEMPO (vM) prevented SCD and improved LV function; the converse (Mv) worsened HF without increasing SCD for up to 5 weeks of follow up, suggesting distinct underlying mechanisms. In subcellular studies of LV myocytes, acute MitoTEMPO exposure strongly suppressed both mROS and cROS. Exogenous H2O2 increased ROS in all intracellular compartments, but mROS scavenging was impaired more than cROS, consistent with downregulation of mitochondrial antioxidant enzymes. AngII increased cROS and mROS in an ARB-inhibitable manner, while MitoTEMPO prevented the AngII induced mROS, but not the cROS response. CONCLUSIONS: Mitochondria are a principal source of cellular oxidative- stress in failing arrhythmogenic hearts, but also are the major sink of ROS. Impaired mROS scavenging cause failing hearts to become more vulnerable to demand-induced oxidative-stress and promote arrhythmic SCD. mROS scavenging may be an effective strategy for SCD therapy in HF.

046 Molecular function of Sigma-1 receptor in obesity-induced metabolic dysfunction Chowdhury S. Abdullah1, Shafiul Alam1, Richa Aishwarya2, Jonette M. Green1,

A. Wayne Orr1, Matthew D. Woolard3, Aimee E. Vozelinek3, Norman R. Harris2, Randa S. Eshaq2, Christopher G. Kevil1, Md. Shenuarin Bhuiyan1, 1Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA, 2Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA, 3Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA

Rationale: Cellular energy metabolism and integrity are maintained by mitochondria. Mitochondrial dysfunction plays a crucial role in the pathogenesis of metabolic disorders. Sigma-1 receptor (Sigmar1) is a highly expressed molecular chaperone protein in the heart but its molecular function in metabolic disorders-induced cardiac remodeling remains unknown. Objective: We aim to investigate Sigmar1’s molecular function in metabolic stress-induced cardiac remodeling. Methods and Results: We found the Sigmar1 localization in the mitochondrial and mitochondrial-associated ER membrane fraction by subcellular fractionation and biochemical experiments. To define the role of Sigmar1 in metabolic disorder, we maintained C57BL/6J male mice on standard rodent diet (control) and high fat diet (HFD) (60% calorie from fat) for 6 months. HFD-fed mice showed marked myocardial

dysfunction represented by decreased percent fraction shortening and percent ejection fraction measured by echocardiogram. Sigmar1 expression was significantly increased in HFD-fed mice heart compared to control mice. Similarly, we also found increased Sigmar1 expression in the heart of db/db mice, a genetic mouse model of obesity. Intriguingly, streptozotocin-treated hyperglycemic rodents did not exhibit any changes in Sigmar1 expression in heart compared to saline treated group. To further define the molecular function of Sigmar1 in these mice, we maintained the Sigmar1 knockout mice and control mice on HFD for the same duration. Sigmar1 knockout mice showed deteriorated cardiac function and adverse cardiac remodeling indicating a protective role of Sigmar1. Conclusions: Our findings suggested a potential protective function of Sigmar1 in metabolic-stress induced cardiac dysfunction and adverse remodeling.

047 Evidence for Reductive Stress in the Heart Failure Patients Thiagarajan Sairam1, Gobinath Shanmugam2, Madhusudhanan Narasimhan3, Meenu Subramanian1, Amit N. Patel4, Rajendran Gopalan1, Ramalingam Sankaran1, Rajasekaran Namakkal Soorappan1,2, 1PSG Institute of Medical Sciences & Research, , Coimbatore, Tamil Nadu, India, 2Department of Pathology/Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL., USA, 3Texas Tech University Health Sciences Center, Lubbock, TX, USA, 4University of Miami – Miller School of Medicine, Miami, FL, USA

Background: Oxidative stress has been linked to heart failure (HF) in humans. Antioxidant-based treatments are often ineffective. Therefore, we hypothesize that some of the HF patients might have a reductive stress (RS) condition. Investigating the RS-related mechanisms will aid in personalized optimization of redox homeostasis for better outcomes among HF patients. Methods: Blood samples were collected from age and sex matched HF patients (n=55) and healthy control (n=50) and serum was immediately preserved in –80°C for redox analysis. Malondialdehyde (MDA; lipid peroxidation) levels by HPLC, reduced glutathione (GSH) and its redox ratio (GSH/GSSG) using enzymatic-recycling assay in the serum of HF patients were measured. Further, the kinetics of key enzymatic-antioxidants (catalase, SOD, glutathione peroxidase and glutathione reductase) were analyzed using UV-Visible spectrophotometer. Results: As expected, majority of the HF patients exhibited oxidative stress with significantly increased MDA levels (lipid peroxidation index) with decreased glutathione redox status (GSH/GSSG). Surprisingly, a subset of HF patients (n=8) displayed significantly decreased MDA levels with profound increase in GSH/GSSG- redox ratio in the serum, suggesting a strong implication for reductive stress in the progression of HF. Furthermore, keen analyses of echocardiography revealed a decreased ejection fraction (EF) with significant diastolic dysfunction (MV E/A) in the HF patients exhibiting RS. Conclusion: These results suggest that RS may lead to deteriorating clinical progression for HF patients. Thus a systematic analysis of redox state is vital for personalized antioxidant treatment in HF patients.

048 Interleukin-1a Blockade Reduce Acute Myocardial Ischemic Injury In The Mouse Adolfo Mauro1, Eleonora Mezzaroma1, Juan Torrado1, Salvatore Carbone1, Benjamin Vantassel1, Antonio Abbate1, Stefano Toldo1, 1Virginia Commonwealth University, Richmond, USA

Introduction: The inflammatory response during acute myocardial infarction (AMI) amplifies myocardial tissue injury and promotes adverse remodeling and heart failure. We sought to investigate the molecular signaling for the initial response during AMI. Hypothesis: The release of Interleukin-1α (IL- 1α) by dying cells during AMI leads to pro-inflammatory signaling and activation of caspase-1 in the heart, leading to further increase in infarct size. Methods: Adult male CD1 mice underwent ligation of the left anterior descending coronary artery to induce a transient myocardial ischemia for 30 minutes, followed by 24 hours of reperfusion. The R&D systems polyclonal IL- 1α blocking antibody (15 µg/Kg) or vehicle were injected intraperitoneally at the onset of the reperfusion (N=6-8 per group). Transthoracic echocardiography was performed to measure left ventricular fractional shortening (LVFS) prior to sacrifice. We used a fluorogenic assay for caspase- 1 activity in the heart and triphenyl tetrazolium chloride (TTC-staining) to measure infarct size at 24 hours. Invitro experiments on cells viability were conducted HL-1 (mouse atrial cardiomyocyte tumor lineage). HL-1 were primed for 4 hours with exogenous IL-1α (100 ng/ml), followed by 30 min of ATP (5mM) stimulation. In simulated hypoxic conditions for 6h HL-1 cells were incubated whit IL-1α-AB (50ng/ml). Results: IL-1α blockade with a blocking antibody after ischemia significantly reduced caspase-1 activity in the heart, associated with a significant reduction in infarct size, and preservation in LVFS at 24 hours. Addition of IL-1α to the culture medium of HL-1 cells

Page 51: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

51

significantly increase cell. Invitro IL-1α-blockade significantly reduced cells death after 6 hour of simulated hypoxia. Conclusion: IL-1α release during AMI leads to amplification of the inflammatory response and exacerbation of myocardial injury due to activation of the caspase-1 during AMI. IL-1α blockade may represent a viable therapeutic strategy as an adjunct to reperfusion in patients with AMI.

049 Constitutive Activation of Nrf2 Causes Hyper-Reductive State and Heart Failure Gobinath Shanmugam1, Madhusudhanan Narasimhan2, Silvio H. Litovsky1, Jolyn Fernandes3, Kevin Whitehead4, John R. Hoidal4, Thomas W. Kensler5, Dean P. Jones3, E. Dale Abel6, Namakkal-Soorapppan Rajasekaran1,4, 1Cardiac Aging & Redox Signaling Laboratory, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA, 2Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA, 3Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory Univeristy, Atlanta, GA, USA, 4Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA, 5Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, USA, 6Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA

Background: Heart failure is a growing health issue leading to morbidity and mortality in the US. Antioxidant supplementation trials have largely failed to protect the heart from oxidative stress-related complications. Although transcriptional activation of antioxidants appears to be favourable, their chronic effects remain elusive. Hence, we tested a hypothesis that chronic activation of Nrf2, a master transcriptional regulator of antioxidants, will cause a hyper-reductive state (HR) and lead to hypertrophic cardiomyopathy. Methods: Novel cardiac-specific transgenic (TG) mice expressing constitutively active Nrf2 (α-MHC-caNrf2-TG) and their non-TG (NTg) littermates were used in this study. First, the rate of survival in TG mice was examined over a period of 80 weeks. Myocardial defence system including glutathione redox state (GSH/GSSG), mRNA (qPCR), and protein (immunoblotting) for antioxidant enzymes were quantified. Echocardiography (Vevo2100 Imager) analyses assessed the cardiac structure and function in NTg, TG-low and TG-high mice (n=6-12/gp.) at 6-8 months of age. Results: A significant increase in the rate of mortality (10 and 40% in TG-low and TG- high vs. NTG) was noted due to heart failure at 60 weeks of age. Increased myocardial glutathione and its redox ratio (GSH/GSSG) along with substantial reduction in ROS in TG-high (p<0.05) compared with TG-low or NTg mice indicated a hyper-reductive (HR) condition in the TG-high mice. Significantly increased Nrf2-promoter activity along with abundant gene and protein levels for major antioxidants (p<0.05) were noted in HR mice. Of note, echocardiography studies shown significant hypertrophic cardiomyopathy with increased ejection fraction (HCMiEF) due to a chronic HR condition. In addition to increased heart-to-body weight ratio and heart weight-to-tibia length, histological analyses confirmed cardiomyocyte hypertrophy, thickening of ventricular walls and reduced chamber volume in TG-high mice. Conclusion: Thus, chronic activation of Nrf2-antioxidants attenuates basal oxidative (ROS) signalling and promotes reductive stress, which is leading hypertrophic cardiomyopathy.

050 Mitochondrial membrane protein Sigmar1 regulates mitochondrial dynamics and function Shafiul Alam1, Chowdhury S. Abdullah1, Richa Aishwarya2, Jonette M. Green1, A. Wayne Orr1, Sumitra Miriyala3, Manikandan Panchatcharam3, Hanna Osinska4, John N. Lorenz5, Jeffrey Robbins4, Md. Shenuarin Bhuiyan1, 1Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA, 2Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA, 3Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA, 4Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Centre, Cincinnati, OH, USA, 5Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA

Rationale: Sigma 1 receptor (Sigmar1) is a molecular chaperone protein known to reside at the interface between the mitochondria and endoplasmic reticulum (ER) in different cellular systems. We recently reported that Sigmar1 is highly expressed in cardiomyocytes but its molecular function still remains unknown. Objective: We investigated the subcellular localization and molecular function of Sigmar1 in regulating mitochondrial function in the heart. Methods and Results: Subcellular fractionation and biochemical experiments confirmed Sigmar1 expression in the mitochondrial- and mitochondrial- associated ER membrane fraction. We isolated mitochondria from the mouse

and human heart cell lysates and found Sigmar1 as an integral mitochondrial outer membrane protein by biochemical experiments. To define the molecular function of Sigmar1, we used the Sigmar1 knockout mice. Sigmar1 knockout mice showed normal morphometry but exhibited significantly altered cardiac hemodynamics and ultrastructure. Ultrastructural analysis by transmission electron microscopy revealed accumulation of irregular shaped highly fused mitochondria and sarcomere disorganization. Examining the mitochondrial dynamics regulatory proteins, we found that Sigmar1 KO hearts showed decreased level of the fission protein, Drp-1 and increased expression of fusion protein, OPA1. Analysis of mitochondrial bioenergetics showed significantly reduced basal respiration and ATP turnover in the mitochondria isolated from Sigmar1 KO hearts. We also found decreased mitochondrial DNA copy number and ATP content in the Sigmar1 KO hearts. Conclusions: Our findings suggested that Sigmar1 is an integral mitochondrial outer membrane protein necessary to maintain normal mitochondrial morphology and function in the heart.

051 miR-181c Regulates Mitochondrial Calcium Influx by targeting Cytochrome C Oxidase subunit 1 Soroosh Solhjoo1, Sangeetha Kannan1,2, Deepthi Ashok1, Brian O'Rourke1, Charles Steenbergen1, Samarjit Das1, 1Johns Hopkins University, Baltimore, MD, USA, 2B.S.Abdur Rahman University, Chennai, Tamil Nadu, India

We have identified a microRNA, miR-181c, which can translocate into the mitochondria of cardiomyocytes, and regulate the mitochondrial gene cytochrome c oxidase subunit 1 (mt-COX1). Recently, we have also demonstrated that miR-181c/d-/- mice are protected against ischemia/reperfusion (I/R) injury by attenuating oxidative stress in the heart. Previous data also suggest that overexpression of miR-181c in the heart can activate Ca2+ entry into mitochondria. Here, we investigate the mechanism by which miR-181c regulates Ca2+ influx into the mitochondrial matrix. We overexpressed miR-181c in H9c2 cells cultured under 1 mM N-acetyl cysteine. After 48 hr of transfection, we have identified a significant increase of PDH activity, suggesting miR-181c regulates mitochondrial Ca2+ influx, which in turn stimulate ROS production. Using miR-181c/d-/- mice, we found both Mitochondrial Calcium Uptake 1 (MICU1) and mitochondrial respiratory complex IV (COX IV) expression are markedly higher in the heart. Immunoprecipitated with MICU1, and then immunoblot for different sub-units of COX IV confirmed a protein-protein interaction between MICU1 and COX IV. We have also found significantly less Pyruvate Dehydrogenase (PDH) activity in neonatal mouse ventricular myocytes (NMVMs) isolated from miR- 181c/d-/- mouse compared to C57BL6 (WT), suggesting significantly lower mitochondrial Ca2+-concentrations in the miR-181c/d-/- group. Utilizing a coverslip induced I/R-model, we observe that siRNAs against MICU1 (si- MICU1) during the ischemic phase significantly increase Ca2+-entry into the mitochondria of the NMVMs. Lowering MICU1 also significantly increases Ca2+-entry into the mitochondria after 30 min of ischemia in miR-181c/d-/-

NMVMs. Furthermore, 30 min ischemia followed by 30 min reperfusion in NMVM monolayers led to significantly less oscillatory instability in mitochondrial inner membrane potential (ΔΨm) in miR-181c/d-/- NMVMs compared with WT NMVMs. However, using si-MICU1 in the miR-181c/d-/-

NMVM group attenuated mitochondrial protection against I/R-injury. MICU1 is directly associated with complex IV. Thus, miR-181c can regulate mitochondrial Ca2+-entry by targeting mt-COX1 during I/R injury.

052 Dual optical mapping of the innervated Langendorff-perfused heart reveals novel insights into acute electrophysiological responses to sympathetic stimulation Lianguo Wang1, Srinivas Tapa1, Samantha Stuart1, Rachel Myles2, Kieran Brack3, Andre Ng3, Donald Bers1, Crystal Ripplinger1, 1University of California Davis, Davis, USA, 2University of Glasgow, Glasgow, UK, 3University of Leicester, Leicester, UK

Background: Sympathetic activation of the cardiac tissue has been recognized as one of the key adaptive mechanisms in the mammalian heart. However, few studies have looked at the dynamic effects of physiological sympathetic nerve activation on cardiac action potentials (AP) and intracellular Ca2+ transients (CaT). Methods and Results: The heart and posterior thoracic cavity from rabbits (NZW, N=9) and mice (C57Bl6, N=8) were dissected and perfused through the descending aorta for dual optical mapping of transmembrane potential (Vm) and intracellular Ca2+ to study the effects of direct sympathetic nerve stimulation (SNS). SNS was achieved through spinal cord stimulation at T1-T3. Our results show that acute SNS (60 sec, 5- 10Hz) increased heart rate (HR) and CaT amplitude in both species. The maximum HR (144±15 to 254±23 BPM in the rabbit and 368±33 to 506±45 BPM in the mouse) and CaT amplitude (increased 9.3±1.6% in the rabbit and 5.9±2.7% in the mouse) were reached after 20sec of SNS. The CaT

Page 52: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

52

amplitude increase was more prominent at the base than at the apex (12.3±4.6% vs. 6.3±1.5%, p<0.05 in the rabbit and 11.3±5.3% vs. 1.0±5.8%, p<0.05 in the mouse), consistent with the anatomical distribution pattern of the sympathetic nerves in the heart. While AP duration (APD) and CaT duration (CaTD) monotonically decreased during SNS in the rabbit, the mouse APD showed a biphasic response, with an initial phase of APD prolongation. The maximum APD prolongation (50.9±5.1 to 60.6±4.1ms) occurred after 20sec of SNS. In addition, rabbit hearts displayed a complete reversal of the direction of repolarization, whereas mouse hearts did not. Conclusions: These data demonstrate that the mouse heart possesses not only unique AP and intracellular CaT morphologies, but also has a significantly different response to physiological sympathetic activation. These important differences should be considered when selecting species for translational cardiac electrophysiology investigations.

053 Effect of thymoquinone on high fructose diet-induced metabolic syndrome in rats Pankaj Prabhakar1, KH Reeta1, Subir K Maulik1, Amit K Dinda1, Yogendra K Gupta1, 1All India Institute of Medical Sciences, New Delhi, India

Background: Thymoquinone (TQ), a principal active ingredient of Nigella sativa (Linn.) seed, is commonly used as a spice in Asian countries. The present study was designed to evaluate the effect of TQ on high fructose diet (HFD)-induced metabolic syndrome (MetS) in male Wistar rats. Methods: Control group was fed chow diet and HFD group was fed HFD (60%; w/w) for 42 days to induce MetS. TQ was administered in three doses (25, 50 and 100 mg/kg, p.o. once daily) along with HFD for 42 days. Fasting plasma glucose, triglycerides, total cholesterol and HDL-cholesterol were estimated at baseline and day 42. Change in blood pressure, oral glucose tolerance and insulin resistance were measured. Hepatic oxidative stress as well as histopathological changes were studied. Hepatic mRNA expressions of PPAR-α and PPAR-γ were also measured by real-time polymerase chain reaction. Results: HFD caused MetS, as manifested by significant increase in systolic blood pressure (146 ± 5 mmHg vs 110 ± 5 mmHg; p < 0.001), fasting plasma glucose (8.32 ± 0.1 mmol/L vs 5.56 ± 0.2 mmol/L; p < 0.001), total plasma cholesterol (1.91 ± 0.2 mmol/L vs. 1.13 ± 0.05 mmol/L; p < 0.01), triglycerides (1.2 ± 0.02 mmol/L vs 0.48 ± 0.04 mmol/L; p < 0.001) and a significant decrease in plasma HDL-C (0.31 ± 0.04 mmol/L vs 0.66 ± 0.03 mmol/L; p < 0.001) as compared to control. Glucose tolerance and insulin tolerance were also significantly impaired with HFD. Hepatic oxidative stress as well as micro- and macrovesicular fatty changes in hepatocytes caused by HFD were also attenuated by TQ. Moreover, TQ preserved the hepatic expression of hepatic mRNA of PPAR-α and PPAR-γ which was reduced by HFD. Conclusions: This study demonstrates protective effect of TQ against HFD-induced MetS which might have been mediated via PPAR modulation and antioxidant mechanisms.

054 Regional cardiac denervation produces supersensitivity of myocardial Ca2+ handling to β-adrenergic stimulation Srinivas Tapa1, Lianguo Wang1, Samantha Francis Stuart1, Crystal Ripplinger1, 1University of California, Davis, Davis, CA, USA

Rationale: Myocardial infarction (MI) can result in chronic loss of sympathetic nerve fibers in the infarct region (denervation). Heterogeneity of sympathetic transmission combined with ischemic damage and fibrosis are all likely contributors to post-MI arrhythmogenesis. However, the precise contribution of denervation to arrhythmogenesis – independent from ischemic damage - has not been systematically investigated. Objective: To create a novel mouse model of regional myocardial sympathetic denervation and measure resulting electrophysiological and Ca2+ handling dynamics. Methods and Results: A targeted toxin (anti-dopamine beta hydroxylase [DBH] conjugated to saporin [Advanced Targeting System, San Diego, CA]) was applied to the apical/anterior region of the ventricle during survival surgery. Untargeted anti- IGg-saporin was applied as a control. At 5 days post-surgery, action potential and Ca2+ handling parameters were measured in Langendorff-perfused hearts using optical mapping with voltage- and Ca2+-sensitive indicators at baseline and in response to isoproterenol (ISO, 1μM). Action potential durations (APD80) were similar in the denervated versus normally innervated regions both before and after ISO (baseline: 51.26 ± 3.36 vs. 48.93 ± 9.17ms, p=NS; ISO: 30.69 ± 14.89 vs. 31.88 ± 13.94ms, p=NS, denervated vs. innervated). At baseline, Ca2+ handling parameters were also similar between denervated and innervated regions. However, following ISO, spontaneous diastolic Ca2+

elevation was frequently observed in the denervated region (14.25 ± 2.23% vs. 5.41 ± 1.43%, p=0.001, denervated vs. innervated). Additionally, an approximate 4-fold reduction in sympathetic fibers in the denervated region was observed. Conclusions: Local application of anti-DBH-saporin produces regional cardiac sympathetic denervation. At 5 days post-denervation,

spontaneous Ca2+ elevation was observed in response to ISO in the denervated region, which may indicate catecholaminergic supersensitivity and an increased likelihood for triggered activity. These findings may have important implications for understanding the specific role of denervation in contributing to post-MI remodeling and arrhythmogenesis.

055 GRK2-S670A Mice reveal cardioprotection post ischemia-reperfusion Priscila Sato1, J Kurt Chuprun1, Laurel Grisanti1, Meryl Woodall1, CJ Traynham1, Anna Maria Lucchese1, Ancai Yuan1, Jessica Ibetti1, Doug Tilley1, Erhe Gao1, Walter Koch1, 1Temple University School of Medicine, Philadelphia, PA, USA

Background: β-adrenergic receptors (βARs) are critical regulators of cardiac contractility whose function are drastically impaired during heart failure (HF). Activated βARs are regulated via phosphorylation by G-protein coupled receptor kinase-2 (GRK2) and subsequent interaction with β-arrestin. Animal and human studies have shown that GRK2 is elevated in HF pathogenesis. In addition to GRK2 being involved in pathological βAR our lab has uncovered non-canonical actions of this kinase in HF development. One such novel action of GRK2 is within mitochondria where we have shown increased GRK2 localization after oxidative stress. Our lab has shown that in the heart, phosphorylation of GRK2 via Map kinase at residue Ser670 promotes binding to Hsp90 and further enhances translocation of GRK2 to the mitochondria after ischemia. Methods: To determine the ultimate role of this pathway in the post-ischemic heart we have developed and generated novel GRK2-S670A knock-in mice where all endogenous GRK2 cannot be regulated by phosphorylation at this residue. Baseline characterization of these mice show a minimal phenotype, however we have found significant differences after ischemia-reperfusion (I/R) injury. Results: Compared to control mice (WT) expressing wild-type GRK2, GRK2-S670A mice have significantly less infarction size and improved cardiac function post-24h I/R. GRK2-S670A showed improved ejection fraction post-IR (53.56%+/- 2.08 S670A vs 42.38%+/- 1.6 WT) and smaller infarcted areas (13.67%+/- 1.976 S670A vs 20.78%+/- 3.579 WT) when compared to control mice. Further, we found that mitochondrial GRK2 protein levels were lower in the GRK2-S670A mice at the area at risk post I/R when compared to WT mice (0.8103 +/- 0.1458 S670A vs 1.610 +/- 0.1983 WT). Conclusion: Overall, our data suggest that phosphorylation at Ser670 of GRK2 promotes translocation to the mitochondria leading to detrimental effects including cell death. This mechanism suggests a novel way to develop pharmacological interventions to aid in the treatment of HF.

056 Model establishment for cardiovascular evaluation using the novel Stellar TSE’s Type PPBTA-XL Telemetry Transmitter in the Vervet (St. Kitts green monkey) Shervin Liddie1, Aryamitra Banerjee1, David Moddrelle1, Xavier Morton1, Matthew Lawrence1, 1RxGen Inc, New Haven, CT, USA

The use of the St. Kitts green monkey (Chlorocebus Sabaeus) in safety pharmacology is not as well documented as other primates, but is gaining popularity as the importance of a more genetically homogeneous primate species for use in biomedical research is being understood. In this test system we have characterized Stellar TSE telemetry devices for acquiring left ventricular pressure (LVP), systemic blood pressure (BP), epicardial electrocardiography (ECG) and core body temperature. In the current study 4 normotensive monkeys were implanted. The systemic pressure catheter was advanced from the internal iliac to the mid aorta. The transmitter body was anchored to the abdominal wall, and the bio-potential leads and LVP catheter were tunneled to the left thorax. LVP was acquired and the bio-potential leads placed for epicardial ECG. Systemic BP, LVP, HR and ECG parameters were determined. Fifteen seconds of data were collected every 30 min for a 48 hr period. The mean for each test parameter were calculated for each 48h acquisition session and subsequently averaged over the in-life duration of the study. The mean systemic systolic, diastolic and pulse pressures as well as heart rate (from LVP) have been stable and consistent with expectation. The mean LV systolic, LV diastolic, mean LVP, pulse pressure as well as the heart rate (from ECG) have been slightly higher in 3 of the 4 monkeys at later time points compared to baseline, possibly due to the use of a shorter catheter that may be squeezed by the ventricular wall, for which reason a longer catheter model has been designed for subsequent studies. To date (6 months), the data generated with the TSE system have been comparable to other telemetry devices and has further supported the green monkey as an acceptable and reliable primate alternative for safety pharmacology or combination cardiovascular/toxicology studies.

Page 53: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

53

057 Dose-dependent effect of Hyperglycemia during Cardiac Development Amelia Cephas1,2, Madhumita Basu2, Vidu Garg1,2, 1The Ohio State University, Columbus, OH, USA, 2Nationwide Children's Hospital, Columbus, OH, USA

Congenital heart disease (CHD) constitutes a major proportion of birth defects among infants. It has a complex etiology, involving both genetic and environmental determinants. Pre-existing maternal diabetes mellitus (DM) is one environmental risk factor associated with increased risk of CHDs. Previous studies have reported a direct correlation between maternal glucose level and the onset of specific cardiac malformations. Hyperglycemia (HG) is the primary teratogen in DM mediated complications and results in increased levels of reactive oxygen species (ROS) and reduced nitric oxide (NO) bioavailability. However, the exact molecular mechanism underlying the influence of altered redox homeostasis on the development of CHD is unclear. In this study, we hypothesized that in response to various concentrations of glycemic stress in utero, there is dose-dependent change in ROS regulating enzymes. We utilized a Streptozotocin induced type1 DM mouse model as well as atrioventricular cushion mesenchymal cells (AVM), an embryonic cardiac cell line for our study. AVM cells were cultured for 6, 24 and 48-hours in normal glucose (NG, 5.5mM) and three different concentrations of D- glucose (HG; 10mM, 20mM and 30mM). Our preliminary data indicated that mitochondrial ROS generation constituted more than 70% of total ROS across HG concentrations compared to NG after 24hr exposure. ROS regulating genes did not show a linear trend in expression across different time points: Sod1 (p=0.0004) and Sod2 (p=0.0108) gene expression was found to be significantly increased after 24hr and 48hr exposure with HG (30mM)while Nox1 (p=0.0004) showed an opposite trend after 24hr exposure (30mM)). In addition, E13.5 embryos from chemically induced maternal type 1 model revealed a spectrum of CHD’s, including septal and outflow tract defects, and neural tube malformations with increased glucose levels. Further histologic and molecular characterization in vivo will provide a better understanding of the mechanism of how maternal hyperglycemic dose influences cardiac development.

058 Radiofrequency Renal Denervation Decreases Renal Fibrosis in Spontaneously Hypertensive Rats (SHR) Juan Gao1, Ian Denys1, Liang Xiao2, David Polhemus1, Frank Smart1, Traci Goodchild1, David Lefer1, David Harrison2, Daniel Kapusta1, 1LSUHSC, New Orleans, LA, USA, 2Vanderbilt University, Nashville, TN, USA

We have shown that radiofrequency ablation (RF-ABL) of the renal arteries decreases sympathetic nerve activity in hypertensive SHR. Findings over the past decade support a positive correlation between heightened sympathetic nerve activity and increased inflammation. The goal of this study was to determine whether RF-ABL decreases renal inflammation and renal fibrosis in hypertensive SHR. Methods: Nineteen-week old SHR received bilateral RF- ABL or Sham-ABL (Biosense Webster Stockert 70 generator and RF-probe). After 4 weeks, kidneys were collected and processed for flow cytometric determination of infiltrating tissue numbers of B cells and T cells (n=8/group), for measurement of tissue cytokine levels (n=10/group), or for Masson’s trichrome staining of fibrotic kidney tissue (n=3/group). Results: In hypertensive SHR, RF-ABL significantly (p<0.05) decreased kidney B cells, CD4+ T cells and CD8+ T cells as compared to levels in the Sham-ABL group. At 4 weeks following RF-ABL, kidney tissue levels of IL-17, INF-γ, MIP-3a, and TNF-α were significantly decreased as compared to levels from Sham- ABL SHR. Masson’s trichrome staining showed a decrease in renal fibrosis of 80% in cortex and 50% in medulla of SHR that underwent RF-ABL as compared to fibrotic expression in Sham-ABL rats. RF-ABL also reduced the ratio of tissue growth factor β (TGF-β) by 30% as compared with SHR treated with Sham-ABL. Conclusions: These findings demonstrate that RF-ABL significantly attenuates renal fibrosis in hypertensive rats and that this beneficial effect may, at least in part, be mediated by the action of RF-ABL to decrease renal inflammation.

059 Renal Sympathetic Denervation Reverses Diastolic Dysfunction in a Rodent Model of Heart Failure with Preserved Ejection Fraction (HFpEF) Hiroshi Koiwaya1, David Polhemus1, Rishi Trivedi1, David Lefer1, 1Cardiovascular Center of Excellence, Louisiana State University Health Science Center, New Orleans, LA, USA

Background: Heart failure with preserved ejection fraction (HFpEF) accounts for 50% of heart failure and has generally poor prognosis. However, there are no approved therapies available for reducing mortality or hospitalization rates in these patients. Renal sympathetic denervation (RDN) is under investigation for the treatment of resistant hypertension and primarily acts primarily by

modulating afferent and efferent renal sympathetic nerves activity. We have previously reported the cardioprotective effects of RDN in myocardial ischemia-reperfusion injury. The aim of this study is to investigate the therapeutic potential of RF-RDN in a rodent model of HFpEF. Methods: 4- week-old male Sprague-Dawley rats were subjected to supracoronary aortic banding (SAB) for 14 weeks followed by administration of L-NAME (2.6mg/kg/24h) for 8 weeks to develop HFpEF. At 22-weeks after SAB, bilateral radiofrequency (RF)-RDN was performed in combination with the application of phenol on the renal arteries. Echocardiography was performed to assess systolic and diastolic function at 4 week intervals throughout the experimental protocol. Results: Twenty-two weeks following SAB and L- NAME, echocardiography revealed left ventricular (LV) hypertrophy with preserved EF and diastolic dysfunction [attenuated E/A ratio and prolonged deceleration time (DcT)]. Following RF-RDN, E/A significantly increased (1.10 ± 0.05 vs. 1.63 ± 0.06, p < 0.01) and DcT significantly shortened (38.7 ± 0.9 ms vs. 31.8 ± 0.8 ms, p < 0.01) compared to those of prior to RF-RDN. There were no significant changes in LV ejection fraction throughout the study. Conclusion: RF-RDN reversed diastolic dysfunction in a rodent model of HFpEF without affecting LV systolic function. Our preliminary results suggest that RF-RDN may have therapeutic potential in the setting of HFpEF.

060 High-Throughput Screen Identifies Novel Small Molecule Stress Regulator That Confers Cardioprotection During Ischemia-Reperfusion Injury Erik Blackwood1, Lars Plate2, Ryan Paxman2, Kyle Malter1, Luke Wiseman2, Jeff Kelly2, 1San Diego State University Heart Institute and Department of Biology, San Diego, CA, USA, 2Departments of Chemistry, Molecular and Experimental Medicine, Chemical Physiology - The Scripps Research Institute, La Jolla, CA, USA

Rationale: During ischemia-reperfusion (IR) injury undue amounts of reactive oxygen species are generated and potentiate myocyte death and cardiac dysfunction. Recently, our lab published a new role for the adaptive cardiac sensor, ATF6, as a key contributor to promoting an adaptive response to oxidative stress. These results highlight the need for the development of small molecules that preferentially activate ATF6 to ameliorate myocardial damage. Through the use of a cell-based high throughput-screen we have identified a small molecular activator of ATF6 and seek to test its efficacy in the heart. Objective/Methods: To address this question, we performed dose response curves of the activator, Compound 147, in cultured neonatal rat ventricular myocytes to examine specific activity of ATF6. We then performed viability studies with chemical stressors and simulated IR. We replicated these activity and IR viability experiments in adult mouse ventricular myocytes and in an ex vivo Langendorff model of the isolated perfused heart. Results: Treatment of cultured ventricular myocytes with Compound 147 specifically and acutely activated ATF6 and primed cells to mount an adaptive response when treated with pharmacological cytotoxic stressors. Treatment of both neonatal and adult ventricular myocytes with Compound 147 increased survivability when subjected to simulated IR injury in vitro. The specificity and activation potential of Compound 147 was confirmed in vivo, by injecting mice intravenously. Furthermore, hearts from mice receiving Compound 147 injections displayed better recovery in response to IR ex vivo. Conclusions: ATF6 is an adaptive cardiac sensor responsible for promoting cardioprotection through the induction of protective targets (e.g. catalase) in response to oxidative stress. As such, it was pertinent to find a small molecule activator of this pathway and examine its efficacy in a clinically relevant scenario of ischemia-reperfusion injury. Compound 147 specifically activates ATF6 and confers cardioprotection during IR in vitro and ex vivo.

061 Multi-omics approach to identify disease signatures in cardiac remodeling Edward Lau1,2, Maggie P.Y. Lam1, Peipei Ping1, 1University of California, Los Angeles, Los Angeles, CA, USA, 2Stanford University, Palo Alto, CA, USA

Background: Alterations in transcript expression and protein expression are often observed to correlate poorly in biological models. There is ongoing debate on whether this reflects inter-platform technical variability or genuine biological regulation. How transcript-protein non-correlation impacts the discovery of disease signatures and mechanisms in practice is not adequately explored. Method: We analysed data from three types of large-scale omics experiments, namely (i) transcript abundance, (ii) protein abundance, and (iii) protein turnover rate, all of which acquired from the normal and remodeling hearts of six genetically diverse strains of laboratory mice. Data from 1,901 genes quantified over 36 measurements (6 strains x 2 conditions x 3 omics types) were computationally integrated to discover disease signatures. Results: We observed poor pairwise correlations between changes in transcript abundance, protein abundance, and protein turnover rates

Page 54: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

54

(Spearman’s correlation coefficients rho: 0.09-0.11). We reason that if transcript-protein non-correlation was largely due to technical variability, then greater overlaps might be expected among high-confidence disease signatures that are reproducibly identified across all mouse replicate experiments. Instead, we found that the three analyzed types of omics data each nominated a distinct set of disease signatures with modest overlaps (12- 18%), and each data type indicated perturbations in separate cardiac pathways. The integration of transcript abundance, protein abundance, and protein turnover data led to 75% gain in reproducibly identified disease gene candidates, including 70 out of 273 disease signatures that were nominated only when protein turnover data were included (e.g., USP47, HADHA, PPIF). In total, 36 nonredundant Reactome pathways were implicated in cardiac remodeling, representing a 57% gain over using only transcript and protein abundance data. Moreover, up to 38% of examined genes (e.g., CLU, MYH6, APOA1) were found to exhibit contradirectional changes in transcript abundance, protein abundance, and protein turnover ratios in the remodelling heart, suggesting the nonoverlap of nominated signatures was not due to arbitrarily chosen cut-offs in data filtering. Conclusion: Transcriptomics and proteomics inquiries nominate distinct disease signature candidates and are enriched in different cardiac pathways. Our results suggest that there may well be value in performing integrated “multi-omics” analyses to provide more comprehensive insights into the pathogenesis of various cardiovascular diseases.

062 Readmission rates after Acute Decompensated Heart Failure Waqas Siddiqui1, Andrew Kohut1, Syed Hasni1, Jesse Goldman1, Benjamin Silverman1, Ellie Kelepouris1, Howard Eisen1, Sandeep Aggarwal1, 1Drexel University College of Medicine, Philadelphia, PA, USA, 2Hahnemann University Hospital, Philadelphia, PA, USA

Introduction: Acute Decompensated Heart Failure (ADHF) is a major health concern worldwide. Ultrafiltration (UF) versus Conventional Diuretics (CD) in ADHF treatment has been studied in several trials with variable outcomes. Objective: To determine if UF is superior to CD in reducing readmissions after ADHF. Methods: MEDLINE was searched using PUBMED for potential studies. Total studies found=590, Total trials=34, Randomized control trials (RCT) = 9 were included (n=820, UF=403, CD =417). RevMan Version 5.3 Copenhagen was used for statistical analysis. Sensitivity analysis was done for heterogeneity. Results: Baseline characteristics were similar in both groups. Mean age was 66 years and 74% were males. Mean EF was 32.9%. Total of 188 patients were readmitted secondary to ADHF with 77 in UF group vs 111 in CD group; RR was 0.71 (95% CI, 0.49-1.02, p=0.07, I2=47%) . There was significantly reduced number of readmissions at 90 days 43 vs 67 in favor of UF; RR was 0.65 (95% CI, 0.47-0.90, P=0.01, I2= 0%). Fluid removal and weight change were significantly higher in UF compared to CD. Change in creatinine pre and post intervention and the number of acute kidney injury were similar in both groups. There was also no difference in the length of hospital stay. Hypotension was more common in UF group compared to CD group (24 vs 13, OR = 2.06, 95%CI = 0.98-4.32, P=0.06, I2=0%) but it was statistically insignificant. Major Adverse Cardiovascular Events (MACE) and mortality was also similar in both groups. Sensitivity analysis was done for heterogeneity and is reported using I2. Conclusions: In this meta-analysis, UF was associated with significantly reduced heart failure readmissions at 90 days and there was trend towards reduced cumulative hospital readmissions. There was no difference in renal outcomes, fluid removal, weight loss, MACE and length of hospital stay in either intervention group.

063 Pacemaker syndrome; an often overlooked diagnosis in patients with pacemakers Awais Arif1, Rizwan Khan1, Nicole Tran1, 1University Of Oklahoma, Oklahoma, USA

The use of cardiac pacemakers has expanded rapidly in recent times. The complicated technology of cardiac pacemakers often discourages clinicians other than electrophysiology specialists from approaching and evaluating for a pacemaker malfunction. As a result, these malfunctions may be missed or confused with other medical etiologies. We present a case of pacemaker syndrome that was initially thought to be an acute exacerbation of heart failure. Case: 88-year-old female with newly diagnosed heart failure with preserved ejection fraction (HF-pEF) and sick sinus syndrome status post pacemaker placement 8 years prior (DC-PPM Medtronic), who presented with acute decompensation of her underlying heart failure. History, exam, labs and chest x-ray findings were classic for heart failure exacerbation so she was treated with IV diuretics. No other triggers for her acute decompensation of heart failure could be identified including dietary indiscretion, medication noncompliance, cardiac ischemia, substance abuse or possible drug

interactions. EKG showed that a right ventricular paced rhythm had replaced atrial pacing. In light of these EKG changes device interrogation of her pacemaker was performed which showed that the pacemaker battery was at end of life (EOL) and as a result, the pacing mode had automatically switched to VVI (right ventricle being paced irrespective of native atrial contraction, resulting in AV dyssynchrony) to conserve energy. Her symptoms were attributed to pacemaker syndrome. She underwent generator change of her pacemaker and following that her symptoms markedly improved and remained asymptomatic at 3 month follow up. Conclusion: Pacemaker syndrome is a common problem faced by physicians who take care of patients with cardiac pacemakers. Patients often present with clinical signs and symptoms of other common cardiac conditions. Clinicians should always have a high index of suspicion for a commonly missed diagnosis of pacemaker syndrome.

064 Tuberculous aortitis, an unusual presentation of tuberculosis Rizwan Khan1, Awais Arif1, Nicole Tran1, 1University Of Oklahoma, Oklahoma, USA

51-year-old female who recently emigrated from Mexico with past history of coronary artery disease (CAD) status post coronary artery bypass grafting (CABG) two years prior who presented with chest pain. She reported diffuse anterior chest pain that radiated to the back. She also reported a 4 kg weight loss, night sweats, chills and fevers over the same time period. In the emergency department her EKG was normal, cardiac enzymes were negative and preliminary CTA report was concerning for possible aortitis vs aortic hematoma. Her ESR and CRP were elevated but autoimmune work-up including ANCA and ANA was negative, raising suspicion for an underlying infectious etiology. The patient had a positive PPD skin test with 17 mm induration, a positive quantiferon test and chest X-ray showed numerous lung nodules bilaterally, consistent with a diagnosis of tuberculosis. Transesophageal echocardiogram showed regional thickening of ascending aorta with the major differential being aortitis vs intramural hematoma vs chronic aortic dissection. Her records from her CABG done in Mexico were obtained and described a porcelain aorta, which further supported a diagnosis of TB aortitis. Cardiothoracic surgery was consulted in light of these results, and they believed that the patient’s aortic thickening was caused by diffuse aortitis. Thus she was diagnosed with aortitis secondary to her underlying tuberculosis. Most cases of aortitis are noninfectious in nature. However, although rare, infectious causes should always be considered due to associated high morbidity and mortality as well as potential reversibility. Tuberculosis as a cause of aortitis is very rare even in developing countries. Tuberculous aortitis should always be considered among patients with aortitis or aortic hematomas, especially if they are among high risk population for tuberculosis or have a history of untreated tuberculosis.

065 Recurrent episodes of loss of consciousness; autoimmune autonomic ganglionopathy with features of postural orthostatic tachycardia syndrome treated with plasmapheresis: a prospective case study Rizwan Khan1, Murtaza Mazhar1, Christian Kaufman1, 1University Of Oklahoma, Oklahoma City, Oklahoma, USA

Objective/Purpose: To report on response to plasmapheresis in a patient with recurrent episodes of loss of consciousness attributed to Seronegative Autoimmune Autonomic Ganglionopathy with intermittent clinical features of Postural orthostatic tachycardia syndrome in light of paucity of RCTs and enough data/recommendations regarding their management and treatment. Design: Case Report. Setting: University based inpatient hospital and clinic setting. Patient: 35-year-old patient with history of lifelong LOC episodes attributed to multiple different etiologies in the past with no clear diagnosis. Eventually diagnosed with autoimmune autonomic ganglionopathy but she was negative for serum antiganglionic antibodies. She also intermittently displayed clinical features of Postural orthostatic tachycardia syndrome. Intervention: Plasma exchange sessions. Outcome Measures: Autonomic assessment, including orthostatic hypotension, pulse, blood pressure, subjective symptoms, number of LOC spells at home and number of LOC spells requiring ER visits. Results: On follow up visits in the following 3 months, patient had just one mild LOC spell and her pulse and blood pressure were continuously stable, her symptoms were subjectively improved. At 4 month follow up she had one spell requiring ER visit with drop in blood pressure to 60/30 necessitating rescue by IV fluids and another spell at home. Her symptoms were subjectively starting to worsen. Conclusions: Plasmapheresis therapy resulted in remarkable but transient improvement in symptoms & loss of consciousness episodes of this patient with sero-negative AAG. Presence of intermittent symptoms of POTS further confounds the already complex clinical picture of this patient, but with response to PLEX it confirms possible autoimmune etiology of both clinical conditions. With the paucity of RCTs for these clinical conditions, further data is needed before

Page 55: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

55

PLEX can be recommended for routine therapy. This also opens up door for discussion and possible use of drugs such as rituximab for long-term symptom relief.

066 Glucose Promotes Cell Growth by Suppressing Branched-chain Amino Acid Degradation Dan Shao1, Outi Villet1, Zhen Zhang1, Sung Won Choi1, Jie Yan2, Haiwei Gu1, Danijel Djukovic1, Danos Christodoulou2, Julia Ritterhoff1, Stephen C Kolwicz Jr1, Daniel Raftery1, Rong Tian1, 1University of Washington, Seattle, WA, USA, 2Brigham and Women’s Hospital, Boston, MA, USA

Cell growth is dependent on glucose but the mechanistic role of glucose metabolism in the transduction of growth signaling is not understood. The present study reports that elevation of intracellular glucose during a growth response is required for the activation of mTOR signaling pathway without affecting the signal input through the PI3K or MAPK pathway. This mechanism is independent of ATP provision by glucose but requires intracellular accumulation of branched chain amino acids (BCAAs). Using transcriptome and metabolomics analysis we identified that high glucose inhibited the expression of enzymes in the BCAA degradation pathway through downregulation of KLF15, a transcriptional regulator of development and metabolism. Interruption of the glucose-KLF15-BCAA degradation axis prevented cardiac hypertrophy in response to pressure overload in wildtype mice but not in mutant mice deficient of BCAA degradation gene. Thus, by demonstrating a novel glucose-BCAA cycle through KLF15 the study reveals a cascade of obligatory metabolic responses previously unrecognized for cell growth. Moreover, identification of KLF15 as a glucose responsive gene will stimulate future studies of how cellular metabolism impacts the biological response through transcriptional mechanisms.

067 The Sphingosine-1-Phosphate Receptor Modulator, FTY720, Reverses Diastolic Dysfunction and Hypertrophy in Hypertrophic Cardiomyopathy David M. Ryba1, Chad M. Warren1, Chehade N. Karam1, Robert T. Davis,

down-regulation of NOX2 and other maladaptive remodeling signals.

068 Effect of moderate exercise training and continuous normobaric hypoxia on postinfarction heart failure in rats Jaroslav Hrdlicka1, Jan Neckar1, Frantisek Papousek1, Jana Vasinova1, Petra Alanova1, Frantisek Kolar1, 1Institute of Physiology CAS, Prague, Czech Republic

Adaptation to continuous normobaric hypoxia (CNH) and exercise training (ET) are known to protect the heart against acute ischemia/reperfusion (I/R) injury. Much less is known about potential therapeutic effect of these interventions on myocardial infarction (MI). The aim of this study was to find out whether CNH (12% O2) or ET (treadmill; 60 min/day for 5 days/wk) can attenuate the progression of postinfarction heart failure. MI was induced by coronary artery occlusion in 2-month-old male rats. Animals were assigned to six groups: i) sham-operated, ii) MI sedentary normoxic controls, iii) CNH for 3 wks before MI, iv) CNH for 3 wks since day 7 after MI, v) ET (30 m/min) for 2 wks before MI, and vi) ET (15 m/min) for 3 wks since day 7 after MI. Echocardiographic examination of the left ventricle (LV) was performed before and 7, 14 and 28 days after MI. Invasive LV pressure measurement was then performed and hearts were collected for histology. MI resulted in LV scar circumference of 38%, gradual increase in systolic and diastolic LV diameter and markedly decreased fractional shortening compared to sham group. CNH before MI reduced mortality from 55% in normoxic controls to 26%. Postinfarction exposure to CNH attenuated LV dilatation. ET before or after MI had no effect on analyzed parameters. Our data suggest that preventive adaptation to CNH reduces acute mortality during I/R insult. Therapeutic exposure to CNH has certain potential to attenuate the progression of unfavorable changes in ventricular geometry induced by post-ischemic heart failure.

069 Cardioprotective adaptation to chronic hypoxia stimulates the ROS- dependent/cytosolic phospholipase A a pathway in rat heart

1 1 1 2 2 3rd , Shamim A. K. Chowdhury , Manuel G. Alvarez , David F. Wieczorek , R. Marketa Hlavackova1,2, Petra Micova2, Klara Hahnova2, Barbora Elsnicova2,

1 1 1 John Solaro , Beata M. Wolska , Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA, 2Department of Molecular Genetics, Biochemistry, & Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA, 3Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, USA

Background. Hypertrophic cardiomyopathy (HCM) patients present with progressively worsening hypertrophy, diastolic dysfunction and fibrosis, ending in heart failure with preserved ejection fraction. At the molecular level, a common pathology found is an increase in myofilament-calcium (Ca) sensitivity. Previously, we have shown that oxidative modification of myofilament proteins, as a result of increased oxidative stress in HCM, may be a direct mediator of this increase. FTY720 is an immunomodulating drug, clinically used in the treatment of multiple sclerosis, which we have previously identified to have anti-fibrotic and anti-hypertrophic effects in a model of pressure-overload-induced hypertrophy. We sought to understand whether agonism of the sphingosine-1-phosphate receptor by FTY720 would be of therapeutic benefit in HCM, and its effects on oxidative modification. Methods. We treated an HCM mouse model linked to a mutation in tropomyosin (Tm-E180G) and non-transgenic (NTG) littermates with FTY720 or vehicle for six weeks. We assessed cardiac function and morphology using echocardiography and pressure-volume (PV) relations and the myofilament- Ca-response using detergent-treated (skinned) fibers. We also investigated if signaling pathways were altered. Results. We found that FTY720-treated Tm- E180G mice had a significant reduction in left atrial size, E/A and E/Em ratios compared to significantly increased baseline measurements as assessed by echocardiography. PV relations revealed significant improvements in the end- diastolic pressure volume relationship, preload recruitable stroke work, and relaxation constant (Tau). FTY720-treated NTG mice displayed a significant decrease in ejection fraction and rate of maximum pressure development in early systole compared to vehicle-treated NTG mice assessed by PV relations. Skinned fiber bundlesrevealed a significant decrease in myofilament- Ca-responsiveness in FTY720-treated Tm-E180G mice. We attributed these functional improvements to a down-regulation of S- glutathionylation of cardiac myosin binding protein-C in FTY720-treated Tm- E180G mice. The increases in oxidative modification in vehicle treated Tm- E180G mice were due to an up-regulation of NADPH oxidase 2 enzyme expression (NOX2). We attributed the and fibrosis and hypertrophy to increased periostin, TGF-beta expression, and GATA4 phosphorylation respectively. These maladaptive changes were reversed by FTY720 treatment. Conclusions. In Tm-E180G mice, FTY720 improves diastolic function and morphology by reversing oxidative modification of myofilament

proteins, resulting in a decrease in myofilament- Ca-responsiveness, via

Page 56: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

56

Anna Chytilova1, Kristyna Holzerova1, Jiri Novotny2, Jitka Zurmanova2, Jan Neckar1, Olga Novakova1, Frantisek Kolar1, 1Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic, 2Department of Physiology, Faculty of Science, Charles University in Prague, Prague, Czech Republic

Adaptation to chronic intermittent hypoxia (CIH) is associated with reactive oxygen species (ROS) generation implicated in the cardiac ischemia-resistant phenotype. Phospholipases A2 (PLA2s) belong to ROS-dependent enzymes and are the key enzymes that take part in the repairing and remodeling of the cell membranes. The aim was to study the effect of CIH on PLA2s isoform expression and β-adrenoceptors (β-AR) signaling in the rat heart. Chronic administration of antioxidant tempol was used to verify the ROS involvement in CIH effect on PLA2s expression. Selected proteins were analysed in the left ventricular (LV) myocardium of adult male Wistar rats adapted to CIH (7,000 m, 8h/day, 5 weeks). CIH increased the total cPLA2α protein in cytosol and membranes (by 191% and 38%, respectively) and p-cPLA2α (by 23%) in membranes. On the other hand, both iPLA2 and sPLA2IIA were down- regulated. Tempol treatment prevented only CIH-induced cPLA2α up- regulation and its phosphorylation on Ser505.The total number of β-AR did not change. Nevertheless, the proportion of β2-AR significantly increased from 30% in normoxic LV to 40% in CIH. Concomitantly, the protein abundance of Gi1,2 and Gi3 increased by 39% and 35%, respectively, by CIH, whereas there was no significant change in the expression of the stimulatory Gsα protein; adenylyl cyclase 5/6 was down-regulated. Furthermore, active/phosphorylated forms of upstream cPLA2

activating signaling molecules (PKCα, ERK1/2 and p38) were increased by CIH. The protein level of cyclooxygenase 2 (COX-2) and the concentration of prostaglandin E2 increased by 36% and 84%, respectively. Our results show that CIH diversely affects myocardial PLA2s and suggest that ROS are responsible for the activation of cPLA2α under these conditions. Moreover, CIH enhances β2-AR-Gi signaling which activates the cPLA2α-COX-2 pathway via ERK/p38. Further experiments should assess the potential role of this pathway in the induction of protected phenotype of CIH hearts.

070 Renal Sympathetic Denervation Improves Left Ventricular Function and Vascular Reactivity in Heart Failure David Polhemus1, Rishi Trivedi1, Zhen Li1, Hiroshi Koiwaya1, Traci Goodchild1, Juan Gao1, Daniel Kapusta1, David Lefer1, 1LSU Health Sciences Center, New Orleans, LA, USA

Background: Renal sympathetic denervation (RDN) is in clinical trials for the treatment of resistant hypertension and acts via interruption of sympathetic

Page 57: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

57

pathways that modulate cardiovascular function. We utilized radiofrequency (RF)-RDN in a rodent model of ischemic heart failure (HF) to investigate the mechanisms by which RF-RDN exerts cardioprotective and vasculoprotective actions in the setting of HF. Methods: 20-week-old male Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHR) were subjected to 45 minutes of transient LAD coronary artery ligation followed by reperfusion for 12 weeks. 4-weeks following reperfusion animals were subjected to either bilateral RF- RDN or Sham-RDN. 12 weeks after reperfusion, thoracic aorta were harvested and vascular reactivity was assessed for endothelium-dependent (acetylcholine) an independent (sodium nitroprusside) vasodilation. Pulse pressure (LV stroke volume/ carotid pulse pressure) was calculated at the 12- week endpoint. LV function (i.e. ejection fraction) was assessed using 2-D echocardiography. Results: RF-RDN significantly improved cardiovascular function as reflected in improved LV ejection fraction at 12 weeks following reperfusion in SHR (46.8 vs. 30.4, p<0.01) and WKY (50.4 vs 35.4, p<0.05). RF-RDN treatment significantly improved endothelium mediated vasorelaxation in both hypertensive and normotensive animals as reflected in ACh EC50 (SHR: 98.8 vs. 349.6 nM, p<0.0001; WKY: 10.03 vs. 44.97, p<0.01). Similar improvements were seen in smooth muscle mediated vasorelaxation to SNP (SHR: 29.4 vs. 145.3 nM, p<0.0001; WKY: 11.68 vs. 39.68 nM, p<0.01). Vascular compliance was significantly improved in SHRs treated with RF-RDN compared to sham (6.6 vs. 3.3 µL/mmHg, p<0.01). Conclusion: RF-RDN therapy at 4-weeks following acute MI significantly improved LV function in the setting of HF independent of blood pressure reductions. RF-RDN improved vascular reactivity suggesting that the cardiovascular benefits afforded by RF-RDN may be partially due to improved vascular function and compliance. Our results reveal a novel mechanism related to the beneficial effects of RF-RDN in HF.

071 Cardiosphere-Derived Cells Combined with Renal Sympathetic Denervation Improves Ventricular Function and Remodeling Following Ischemic Injury David Polhemus1, Rishi Trivedi1, Traci Goodchild1, Geoffrey De Couto2, Eduardo Marban2, David Lefer1, 1LSU Health Sciences Center, New Orleans, LA, USA, 2Cedars Sinai Heart Institute, Los Angeles, CA, USA

Background: Cellular postconditioning with cardiosphere-derived cells (CDCs) in acute myocardial infarction reduces infarct size and preserves viable myocardium. We have previously reported that delayed renal sympathetic denervation (RDN) improves post-infarction remodeling and function. We hypothesize that following ischemia reperfusion injury, CDC postconditioning with delayed RDN therapy will lead to superior cardiac function and remodeling. Methods: 20-week-old male Wistar-Kyoto rats were subjected to 45 minutes of LAD coronary artery ligation followed by reperfusion for 14 weeks. 20 minutes into reperfusion, rats were treated with CDCs (0.5x106 intra-cardiac with aortic cross clamp) or PBS. 4 weeks into reperfusion, rats received either bilateral RDN or Sham-RDN. LV function and remodeling were assessed biweekly using 2-D echocardiography. Results: CDC + RDN therapy significantly improved LV ejection fraction (EF) compared to PBS + Sham-RDN (58.5 ± 2.5 vs 38.7 ± 2.1 %, p<0.0001) at the 14-week endpoint. Interestingly, CDC + RDN combination therapy was superior to both CDCs alone and RDN alone (Figure 1). LV end-diastolic diameter (LVEDD) was less in the CDC + RDN group (7.4 ± 0.2mm) compared to PBS + Sham-RDN treated rats (8.5 ± 0.2 mm, p<0.001) and CDC + Sham-RDN treated rats (8.3 ± 0.2mm, p<0.01) at the 14-week endpoint. Septal wall thickness at end-systole (IVSs) was greater in the CDC + RDN treated rats (3.0 ± 0.1 mm) compared to PBS + Sham-RDN treated rats (2.3 ± 0.1 mm, p<0.001) and CDC + RDN treated rats (2.4 ± 0.1 mm, p<0.01) at 14 weeks. Conclusion: The use of RDN with CDC following ischemic injury improves ventricular function and remodeling and preserves septal thickness. Importantly, this combination therapy has superior effects than either treatment alone. Our results reveal that the protective effects of CDC cellular postconditioning may be enhanced if used in combination with RDN in treating post infarction injury.

072 A Novel Hydrogen Sulfide Donor, JK1, Protects the Heart Against Pressure Overload Induced Heart Failure A Novel Hydrogen Sulfide Donor, JK1, Protects the Heart Against Pressure Overload Induced Heart Failure Zhen Li1, Chelsea Organ1, David Polhemus1, Rishi Trivedi1, Jianming Kang2, Ming Xian2, David Lefer1, 1LSU Health Sciences Center, New Orleans, USA, 2Washington State University, Pullman, USA

Background and Hypothesis: Hydrogen sulfide (H2S) protects against acute myocardial ischemia/reperfusion injury and heart failure by ameliorating oxidative stress, improving mitochondrial function, and attenuating apoptosis. One major limitation in the development of H2S-based therapy is a lack of

evidence supporting its cardioprotective effects in a clinically relevant scenario that mimics heart failure in humans. A second limitation is that current H2S donating agents have very poor pharmacokinetic profile. In the present study we tested the efficacy of a novel H2S donor, JK1, with controlled H2S release and increased half-life compared to traditional H2S-releasing agents. We evaluated JK1 in a clinically relevant murine model of transverse aortic constriction (TAC) induced heart failure (HF). We hypothesized that delayed administration of JK1 would protect the myocardium against TAC-induced heart failure. Methods: Male C57/BL6J mice at 10 weeks (wks) of age were subjected to TAC (27 g needle) for 16 weeks. Optimal dosing of JK1 was determined from preliminary studies of in vivo myocardial ischemia and reperfusion in which 100 µg/kg was shown to significantly reduce myocardial infarct size. JK1 (100 µg/kg/d, n=16) or control compound (100 µg/kg/d, n=15) were administered (i.p.) starting at 3 wks post TAC. Echocardiography was performed at baseline and following TAC to assess cardiac function. Results: Delayed treatment with JK1 preserved left ventricular (LV) ejection fraction (LVEF 48.48% vs. 31.21%, p < 0.0001), reduced LV dilation (LVEDD/LVESD 4.18/3.20 mm vs. 4.73/4.03 mm, p < 0.001), and attenuated eccentric hypertrophy (IVSd 0.90 mm vs. 0.78 mm, p < 0.01. LVPWd 0.85 mm vs. 0.73 mm, p < 0.001) at 16 wks post TAC as compared to control compound. Conclusion: These results demonstrate that delayed administration of JK1 significantly preserves LV function and attenuates adverse remodeling in HF. And the beneficial effects of JK1 sustained through the 16-week HF study. Studies are currently underway to define the molecular mechanisms involved in JK1 induced cardioprotection.

073 A Novel Hydrogen Sulfide Prodrug, SG-1002, Augments Angiogenesis and Coronary Vascular Tone in a Swine Model of Critical Limb Ischemia Amanda Rushing1, Erminia Donnarumma1, Amy Scarborough1, Sarah Boisvert1, Rishi Trivedi1, David Polhemus1, Zhen Li1, Kevin Au2, Sam Victoria2, Jeffrey Schumacher3, David Lefer1, Traci Goodchild1, 1Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA, 2Department of Vascular Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA, 3Department of Animal Care, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA

Introduction: Critical limb ischemia (CLI) is a peripheral arterial disease (PAD) caused by atherosclerosis that results in obstructions in the arteries resulting in limited blood supply to organs other than the heart. CLI is further defined as reduced blood flow to the lower limb(s) resulting in pain, even at rest, with or without associated ulcers or gangrene. Patients with CLI have a 6-month risk (25-40%) of major amputation and an annual mortality of 20% (11). Surgical and catheter-based revascularization are the preferred approaches for CLI though many patients with CLI are poor candidates for revascularization. Additionally, even after successful revascularization, stenosis or failure of the graft often reoccur resulting in a need for the development of new treatments for patients with CLI who are not candidates for revascularization. Objective: We sought to determine whether a novel H2S prodrug, SG-1002, promotes peripheral revascularization and coronary artery vascular function in a clinically relevant swine model of CLI. Approach and Results: Current studies have shown hydrogen sulfide (H2S) has various cytoprotective and cardioprotective properties. This study evaluated the therapeutic potential of the H2S donor, SG-1002, for critical limb ischemia in a swine model. 7 days after miniswine CLI induction, animals (n=17) received placebo or H2S donor, SG-1002 (800 mg PO BID), for 35 days. We observed higher circulating levels of H2S (5.0 ± 1.2 uM verses 1.8 ± 0.50 uM; P<0.05), sulfane sulfur (10.6 ± 2.3 uM verses 2.6 ± 0.8 uM; P<0.05), and nitrite (0.5 ± 0.05 uM verses 0.3 ± 0.03 uM; P<0.005) in pigs treated with SG-1002 at Day 35. DSA revealed an increase (P<0.05) in ischemic limb vessel number in the SG-1002 treated pigs (27.6 ± 1.6) compared to the placebo group (22.2 ± 1.8). SG-1002 treatment improved bradykinin relaxation response of the LAD (EC50: 6.49 ± 1.2 nM verses 2.52 ± 1.2 nM, P<0.001) and the LCX (EC50: 2.58 ± 1.2 nM verses 7.36 ± 1.2 nM, P<0.0001). SG-1002 treatment improved SUB P relaxation responses of the LAD (EC50: 0.08 ± 0.02 nM verses 0.22 ± 0.01 nM, P<0.0001) and the LCX (EC50: 0.07 ± 0.01 nM verses 0.14 ± 0.01 nM, P<0.001). SG-1002 treatment also improved SNP relaxation responses of the LAD (0.42 ± 0.01 uM verses 0.54 ± 0.01 µM, P<0.05) and the LCX (EC50: 0.38 ± 0.01 uM verses 0.81 ± 0.01 µM, P<0.0001) compared to placebo. Conclusions: Results of this study suggest that daily administration of the novel H2S prodrug, SG-1002, leads to an elevation in circulating metabolites for H2S and NO signaling. By investigating the effects of SG-1002 on coronary artery vascular function and angiogenesis in a clinically relevant swine model of CLI, we show that oral administration of a novel long-acting H2S prodrug improves coronary artery vascular reactivity and vessel growth via an NO- dependent mechanism.

Page 58: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

58

074 A Novel Histone Deacetylase (HDAC) Inhibitor Attenuates Cardiac Dysfunction in the Setting of Pressure Overload Heart Failure Chelsea Organ1, 2, Zhen Li1, 2, Craig Zibilich1, Traci Goodchild1,2, Shubing Wang3, Kersten M. Small4, Jeffrey Madwed4, Jian Liu4, Joseph Kozlowski4, David J. Lefer1, 2, 1Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA; 2Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, USA; 3Merck Research Labs, Rahway, NJ, USA; 4Merck Research Laboratories, Kenilworth, NJ, USA

Background: Histone deacetylases (HDACs) are a family of epigenetic regulator enzymes responsible for chromatin conformation and reversible protein acetylation. HDAC inhibitors have previously been shown to be cardioprotective in acute myocardial infarction and heart failure. HDAC inhibition is thought to attenuate cardiac remodeling and improve cardiac function by enhancing acetylation of sarcomeric proteins, suppressing autophagy and apoptosis, bolstering protective genes and inhibiting pro- inflammatory genes. Objective: We investigated the efficacy of a novel, orally active HDAC class I inhibitor, MRL-003, in a murine model of pressure overload induced heart failure. Methods and Results: All studies were performed in a fully blinded manner, data analysis was performed off site by independent statisticians, and data was only unblinded following completion of statistical analysis of all parameters. Mice (C57 BL6J) at age 8-10 weeks were subjected to transverse aortic constriction (TAC) to induce pressure overload heart failure. Cardiac function was monitored sequentially using echocardiography and terminal hemodynamic measurements were taken. At 3 weeks post-TAC 3 doses (10, 30, and 100 mg/kg/day) of MRL-003 were provided to the mice in a blinded manner in chow ad libitum for 13 weeks. MRL-003 (100 mg/kg/d) significantly improved left ventricular (LV) ejection fraction and LV chamber dimensions compared to TAC-vehicle. LV end- diastolic pressure (LVEDP) was significantly increased in the vehicle treated group compared to sham and there was no significant difference in LVEDP in mice treated with MRL-003 (100 mg/kg/d) compared to sham. Conclusion: HDAC inhibition with MRL-003 significantly improves cardiac function and remodeling in the setting of pressure overload heart failure. Further studies are warranted to assess the molecular changes associated with these functional improvements.

075 Hypercholesterolemic LDLr Knockout Swine as a Clinically Relevant Model of Hypertension Amanda Rushing1, Amy Scarborough1, James Stephen Jenkins2, John Reilly2, Seena Khosravi1, Rishi Trivedi1, David Polhemus1, Traci Goodchild1, David Lefer1, 1Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA, 2Ochsner Interventional Cardiology, Ochsner Medical Center, New Orleans, LA 70121, USA

Introduction: Coronary artery atherosclerosis is the single largest killer of men and women in the United States. Mutations in the LDL receptor (LDLr) gene lead to hypercholesterolemia and are associated with elevated plasma levels of total and LDL cholesterol. Renal denervation (RDN) has recently been investigated in clinical trials as a possible therapeutic for hypertensive patients. Although the clinical trials showed no significant difference in the blood pressures in control and RDN patients, researchers have shown reduction in myocardial infarction injury in a hypertensive rat model. Although RDN has been shown to have no change in blood pressure, we sought to investigate the change RDN inflicts on the sympathetic nervous system in the setting of hypercholesterolemic swine. Objective: We sought to develop a clinically relevant large animal model of cardiovascular dysfunction and to replicate experimental design of clinical renal denervation (RDN) in swine to test human catheters and devices. We also sought to investigate the efficacy of renal denervation on the sympathetic nervous system in regards to blood pressure, nor-epinephrine levels, Tyrosine Hydroxylase staining, BNP, ANP, and kidney neprilysin activity. Approach and Results: LDLr -/- swine (n=8) were implanted with DSI radiotelemetry transmitters for continuous arterial blood pressure measurement followed by renal denervation (n=4) or a sham procedure (n=4) after one month of high fat high salt diet (HFHS). Tyrosine Hydroxylase staining showed successful renal denervation via a significant difference in stain intensity between the control and RDN groups in both the proximal (p=0.02) and distal (p<0.001) renal arteries. There was no significant difference in plasma BNP, plasma ANP, or kidney NEP activity between the control and RDN animals. LDLr -/- swine on control diet were hypertensive (SYS 156±5, DIA 104±3 mmHg) with limited increase in blood pressure on the HFHS diet. RDN treated animals showed a slight increase in blood pressure (SYS 159±5, DIA 117±16 mmHg) compared to control animals. Conclusions: Our study confirmed early clinical trials findings that blood pressure is not reduced after ablation of the renal arteries by radio frequency. Our study showed that LDLr -/- swine are a relevant large animal model of hypertension

without HFHS diet. Through significant changes in Tyrosine Hydroxylase, we confirmed successful ablation of renal arteries in the RDN treated swine. Further investigation is need to understand the molecular changes that renal denervation evokes as well as its efficacy as a therapeutic treatment for myocardial infarction and hypercholesterolemia.

076 Genetic inhibition of the UPR gene Chac1 preserves cardiac function in a murine model of pressure overload induced heart failure. Zhen Li1,2, Lisa O Nguyen1, Chelsea L Organ1,2, David J Lefer1,2, Imran N Mungrue1, 1Lsu Health- Dept Pharmacology, New Orleans, USA, 2LSU Health- Cardiovascular Center of Excellence, New Orleans, USA

Introduction: The Unfolded Protein Response (UPR) is induced accompanying endoplasmic reticulum (ER) stress in cardiac myocytes, and has been implicated in the development of heart failure. Chac1 is a UPR induced gene, which directs enzymatic cleavage of intracellular glutathione and promotes cell death. Ablation of the Chac1 gene (Chac1-/-) in mice is lethal, whereas Chac1 heterozygous (Chac1+/−) mice were produced and are viable. Purpose: To investigate the effects of Chac1 haplo-insufficiency on cardiac structure and function in mice, in the setting of heart failure after aortic banding. Methods and Results: Chac1+/− and control (WT) mice (n ≥ 12) were subjected to aortic banding to induce pressure overload heart failure. After banding, the extent of cardiac dysfunction was evaluated using 2-D echocardiography, including ventricular hypertrophy and reduced cardiac output. At baseline, we found Chac1+/− mice had cardiac hypertrophy and increased ejection fraction (70 ±1 % vs 61 ±1 %, p < 0.01) compared to WT. Querying early time points 6-9 weeks after aortic banding, a parallel hypertrophic response occurred in Chac1+/− vs controls. Notably, by 15 weeks after aortic banding Chac1+/− mice had preserved cardiac ejection fraction (58±4 vs 36±2%, p < 0.01) compared to WT that had progressed into heart failure. Chac1+/− mice also displayed attenuated chamber dilation (LVEDD: 3.9 ±0.1 vs 4.5 ±0.1 mm p <0.01), lower normalized cardiac mass (9.6 ±0.3 vs 10.9 ±0.5, p<0.05), and reduced pulmonary edema (99 ±6 vs 120 ±3 mg fluid). Examining cardiac ventricle mRNA expression 15 weeks after TAC, Chac1+/−

mice showed decreased Nppb (BNP), MLC2v, and Pin1 expressions versus WT. Conclusions: A benefit for Chac1 haplo-sufficiency in mice accompanying heart failure is revealed, evidenced by preserved cardiac structure and function. Additionally, Chac1+/− mice have increased cardiac mass and enhanced function at baseline, mimicking exercise-induced cardiac hypertrophy. Chac1 inhibition may represent a novel therapeutic strategy for heart failure, phenocopying the cardiac benefits of exercise.

077 Ischemic vs. Non-Ischemic Dilated Cardiomyopathy: a Comparative Study in Stem Cell Therapy Efficacy Bryon A. Tompkins1, Angela C. Rieger1, Victoria Florea1, Makoto Natsumeda1, Evan D. Nigh1, Ana Marie Landin1, Gianna M. Rodriguez1, Konstantinos E. Hatzistergos1, Ivonne H. Schulman1, Joshua M. Hare1, 1Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA

Background: Ischemic (ICM) versus dilated cardiomyopathy (DCM) have different pathogenesis and outcomes with standard medical therapy. Mesenchymal stem cell (MSC) therapy has produced improvements in both diseases. However, whether the efficacy of MSCs between ICM and DCM subjects is comparable, remains undetermined. Hypothesis: Subjects with DCM respond better than those with ICM. Methods: We analysed the effects of transendocardial MSC injections in patients with ICM vs. DCM from three single-institution, randomized, double-blinded, clinical trials: TAC-HFT, POSEIDON and POSEIDON-DCM. Cardiac structure, function and subject quality of life data were compared between ICM and DCM groups at baseline and one year follow-up. Results: Ejection fraction improved in DCM 7% (2.9, 11.0, p=0.002) vs. ICM 1.5% (-0.2, 3.2, p=0.08) subjects with a significant difference between groups (p=0.005). Similarly, stroke-volume increased in DCM 10.59mL (0.2, 21, p=0.046) vs. ICM -0.16mL (-4.6, 4.3, p=0.94) subjects with a significant difference between groups (p=0.02). End-diastolic volume improved only in ICM subjects, -10.68mL (-21, -0.1, p=0.047), and end- systolic volume improved only in DCM subjects, -17.8mL (-54.5, 17.0, p=0.049). Sphericity index only decreased in ICM, -0.04 (-0.06, -0.02, p=0.0002), subjects. End-diastolic mass increased in ICM, 3.57g (-3.1, 28, p=0.004), vs. DCM subjects, -4.1g (-15, 7, p=0.45), with a significant difference between groups (p=0.007). Six-minute walk test improved in DCM, 34m (18, 53, p=0.009), and ICM, 30m (-16, 89, p=0.0007), subjects with a difference between groups (p=0.03) favoring ICM subjects. NYHA class improved in both DCM (p=0.005) and ICM (p=0.02) subjects (between group p=0.20). Similarly, the Minnesota Heart Failure Questionnaire improved in DCM, -19 (-31, -7.1, p=0.003), and ICM, -9.3 (-16, -2.7, p=0.007), subjects (between group difference p=0.12). Conclusion:

Page 59: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

59

MSC therapy exerts distinct effects in ICM vs DCM. It improves functionality in DCM, cardiac remodelling in ICM, and enhances quality of life parameters in both disease processes.

078 Regulation of cardiac excitation-contraction coupling by fibroblasts in health and disease Brian Wang1, Cesare Terracciano1, Kenneth Macleod1, 1Imperial College London, London, UK

Introduction: Cardiomyocyte structure and function are influenced directly via cell-cell communication, the ubiquitous extra-cellular matrix (ECM), and paracrine interactions. These interactions modulate cardiomyocytes in health and the changes that occur in disease. Clear mechanisms in these interactions are still lacking, particularly how fibroblasts may affect myocyte excitation-contraction coupling in a human model. Methods: Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were co-cultured with human cardiac fibroblasts, isolated from heart failure patients, at a ratio of 2:1 fibroblasts:hiPSC-CMs for 24 hours in three different conditions: (1) hiPSC-CMs in fibroblast-conditioned medium, (2) co-culture with fibroblasts in tissue culture inserts, and (3) fibroblasts in direct contact with hiPSC-CMs. These conditions differentiate the various aspects of cardiomyocyte-fibroblast interaction. Optical mapping experiments were conducted to visualise changes in excitation-contraction coupling. Results: Human fibroblasts- conditioned medium altered excitation-contraction coupling by decreasing calcium transient amplitude and prolonging time to 80% decay vs baseline (p<0.001, -30±2.6% and +6±2.6%). Direct contact with fibroblasts further decreased calcium transient amplitude (p<0.001 vs. baseline, -46±4.3%) and further prolonged time to 80% decay (p<0.001 vs. baseline, +47±8.7%). Soluble Arginine-Glycine-Aspartic acid (RGD) motif-containing peptides present in the attachment site of many ECM proteins reduce the duration of transients and cause hiPSC-CM clustering and detachment from the glass surface substrate. These effects are not attenuated in the presence of anti-β1 or anti-β3-integrin receptor antibodies, but are also likely to prevent integrin receptor activation. Discussion:Human fibroblasts isolated from the ventricles of cardiomyopathy patients modulate calcium transients in hiPSC-CMs. The effects are dependent on whether the interaction is paracrine or direct, and unidirectional or bidirectional. Although it is unclear which interactions play the greatest role in modulating cardiomyocytes, it is accepted that multiple pathways and factors are involved. Further work is needed to identify mechanisms and understand their subsequent consequences in health and in disease.

079 DWORF overexpression prevents heart failure in an experimental mouse model of dilated cardiomyopathy Cat Makarewich1,2, Svetlana Bezprozvannaya1,2, Rhonda Bassel-Duby1,2, Eric Olson1,2, 1Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA, 23Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, USA

Background: Defective intracellular Ca2+ homeostasis is a hallmark feature of heart failure. Increasing the expression level or activity of the sarcoplasmic reticulum Ca2+-ATPase (SERCA) pump can normalize Ca2+-handling and rescue several genetic and experimental models of heart failure. Results: Our lab has recently identified and characterized a novel transmembrane protein named DWORF (DWarf Open Reading Frame) that binds to SERCA and enhances its activity by displacing phospholamban (PLN), a potent SERCA inhibitor. Transgenic mice with cardiac-specific DWORF over- expression (αMHC-DWORF) have a cellular phenotype that mimics that of PLN KO mice, exhibiting an increase in peak Ca2+-transient amplitude, faster cytosolic Ca2+ decay rates, higher SR Ca2+ load and enhanced cardiomyocyte contractility. Using a well-characterized mouse model of dilated cardiomyopathy (DCM) due to deletion of the muscle-specific LIM protein (MLP KO), other laboratories have previously shown that genetic ablation of PLN leads to a restoration of SERCA activity and prevention of DCM in the hearts of MLP KO mice. Invoking a similar scientific rationale here, we generated αMHC-DWORF Tg/MLP KO mice and observed that DWORF overexpression in the heart of MLP KO mice resulted in a complete rescue of in vivo cardiac function and prevented the pathological remodeling, development of fibrosis and ultrastructural defects that are all characteristic features of MLP KO hearts. At the cellular level, cardiomyocytes isolated from αMHC-DWORF Tg/MLP KO displayed enhanced Ca2+-cycling and contractile properties that contrasted sharply with MLP KO cardiomyocytes, which exhibited depressed contractility. Conclusion: Cardiac-specific over- expression of DWORF prevents the development of the DCM phenotype characteristic of MLP KO mice and provides an attractive candidate for a novel heart failure therapeutic.

080 CaMKII oxidation causes increased atrial fibrillation in diabetic mice Olurotimi Mesubi1, Adam Rokita2, Neha Abrol1, Yuejin Wu1, Biyi Chen2, Qinchuan Wang1, Jonathan Granger1, Elizabeth Luczak1, Lars Maier4, Xander Wehrens5, Joel Pomenrantz3, Long-Sheng Song2, Gerald Hart3, Mark Anderson1, 1Division of Cardiology, Dept of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA, 2Division of Cardiovascular Medicine and Cardiovascular Research Center, Carver College of Medicine, Iowa City, IA, USA, 3Department of Biological Chemisty, The Johns Hopkins University School of Medicine, Baltimore, MD, USA, 4Division of Cardiology and Pneumology, German Heart Center, University Hospital Goettingen, Goettingen, Germany, USA, 5Department of Molecular Physiology & Biophysics and Medicine (Cardiology), Baylor College of Medicine, Houston, TX, USA

Background: Diabetes mellitus (DM) is an independent risk factor for atrial fibrillation (AF), the most frequent clinical arrhythmia, but the mechanism(s) underlying this clinical association are unknown. Increased reactive oxygen species (ROS) and O-GlcNAcylation (OGN) is a hallmark of DM. Hyperactivated CaMKII promotes AF, and CaMKII may become constitutively active and proarrhythmic by oxidation (ox-CaMKII) or OGN (OGN-CaMKII), post translational modifications to adjacent amino acids in the regulatory domain. We hypothesize that CaMKII promotes diabetes-induced AF through ox-CaMKII and OGN-CaMKII. Methods and Results: We generated knock-in mouse models, based on the predominant CaMKII isoform in myocardium (CaMKIIδ), to selectively eliminate ox-CaMKII and OGN-CaMKII. We used the pancreatic β-cell toxin, streptozocin (STZ) or a combination of high fat diet and STZ, to generate validated mouse models of T1D and T2D, and an established right atrial pacing model of paroxysmal AF. T1D and T2D mice showed increased AF [70% (14/20), p < 0.05; and 61% (11/18), p < 0.05] compared to non-diabetic controls [25% (5/20)], suggesting diabetes was a risk factor for AF in mice. We focused on T1D because it is a simpler model system, compared to T2D. We found that enhanced AF risk was prevented in mice with T1D by myocardial CaMKII inhibition due to transgenic expression of a CaMKII inhibitory peptide, AC3-I, suggesting that CaMKII is required for AF in diabetes. Atria from T1D mice showed increased ROS, OGN, and ox- CaMKII. Mice selectively resistant to ox-CaMKII due to knock-in replacement of regulatory domain methionines with valines (MMVV) with T1D were resistant to AF. In contrast, knock-in mice with putative OGN resistant CaMKII (S280A) and wild type (WT) littermate mice with T1D showed similar increases in AF, indicating that OGN-CaMKII did not contribute to AF. WT T1D atrial myocytes, but not MMVV T1D atrial myocytes, showed increased RyR2 Ca2+ leak, delayed afterdepolarizations (DADs) and triggered action potentials, indicating ox-CaMKII coupled ROS to a fundamental proarrhythmic cellular pathway. Addition of an OGN antagonist prevented increased AF, RyR2 Ca2+ leak, DADs and triggered action potentials. Knock-in mice lacking a CaMKII phosphorylation site on RyR2 (S2814A) with T1D were resistant to AF, compared to WT T1D controls, establishing the contribution of CaMKII and RyR2 to AF in this model, and suggesting that OGN is proarrhythmic by a CaMKII independent pathway involving RyR2. Conclusions: These studies establish CaMKII as a critical ROS sensor and proarrhythmic signal in AF, and suggest that ox-CaMKII and OGN converge on RyR2 to promote AF in diabetes. These provide new insights for understanding important clinical phenotypes that currently lack clear mechanisms and adequate therapies.

081 LCZ696, the First-in-Class Angiotensin Receptor Neprilysin Inhibitor, Improves Vascular Reactivity in the Setting of Heart Failure Rishi K. Trivedi1, Zhen Li1, David J. Polhemus1, Daniel Yoo1, Hiroshi Koiwaya1, Traci T. Goodchild1, David J. Lefer1, 1LSU Health Sciences Center - New Orleans, New Orleans, LA, USA

Background: LCZ696 combines an angiotensin receptor blocker (ARB) with a neprilysin inhibitor (NEPi). This inhibits the renin-angiotensin system while simultaneously augmenting the natriuretic peptide system. LCZ696 has been shown to improve myocardial function in the setting of heart failure (HF), however, its effects on vascular function have not been defined. Methods: Male spontaneously hypertensive rats (SHRs) at 18-20 weeks of age were subjected to 45 minutes of transient LAD coronary artery ligation followed by reperfusion for 8 weeks to induce HF. Starting at 4 weeks post-reperfusion animals received daily oral therapy of either vehicle (VEH) or 68 mg/kg LCZ696 (LCZ). After 8 weeks of therapy, thoracic aorta was collected and evaluated for vascular reactivity in a tissue chamber apparatus connected to force transducers. Vasoreactivity responses were assessed for acetylcholine (ACh, 0.1nM–10mM), sodium nitroprusside (SNP, 0.1nM–10 mM), and N(omega)-nitro-L-arginine methyl ester (L-NAME, 30mM). Results: LCZ treatment significantly improved vascular reactivity for both ACh and SNP. There was a significant improvement in maximal relaxation in response to ACh as well as ACh EC50 suggesting augmented endothelium mediated vasorelaxation. Similar changes were seen in SNP maximal relaxation and

Page 60: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

60

SNP EC50 suggesting improvements in vascular smooth muscle function. Lastly, there was an increased contraction in response to administration of L- NAME following maximal relaxation by ACh suggesting the potential role of nitric oxide in the improvements seen in vascular reactivity. Conclusion: LCZ696 significantly improved LV function during heart failure in the setting of hypertension with concomitant improvements in vascular reactivity. These improvements in vascular reactivity are likely a result of increased activity of endothelial nitric oxide synthase (eNOS) and increased bioavailability of nitric oxide (NO). Studies are currently underway to evaluate eNOS function and NO levels following LCZ administration in heart failure.

082 LCZ696 Reduces Myocardial Fibrosis in Hypertensive Rats in the Setting of Heart Failure Rishi K. Trivedi1, Zhen Li1, David J. Polhemus1, Daniel Yoo1, Hiroshi Koiwaya1, Traci T. Goodchild1, David J. Lefer1, 1LSU Health Sciences Center, New Orleans, LA, USA

Background: LCZ696 combines an angiotensin receptor blocker (ARB) with a neprilysin inhibitor (NEPi). LCZ696 has been shown to improve myocardial performance in the setting of heart failure with reduced ejection fraction (HFrEF) in patients (PARADIGM-HF), however, the mechanisms by which this occurs are poorly understood. Methods: Male spontaneously hypertensive rats (SHRs) at 18-20 weeks of age were subjected to 45 minutes of transient LAD coronary artery ligation followed by reperfusion for 8 weeks to induce HFrEF. At 4 weeks post-reperfusion animals received daily oral therapy consisting of vehicle (VEH), 31 mg/kg Valsartan (VAL) or 68 mg/kg LCZ696 (LCZ). Cardiac structure and function were determined via 2-D echocardiography (Vevo 2100) and measurements of left ventricle (LV) pressures were performed at 12 weeks post-MI. After 8 weeks of therapy, myocardial tissue in the mid-ventricular region was collected and processed for fibrosis histology using Picrosirius Red and Masson’s Trichrome. Results: LCZ treatment improved LV ejection fraction, LV end-diastolic pressure, and LV diastolic relaxation (tau) compared to both VEH and VAL. Interestingly, treatment with either LCZ or VAL resulted in significant reductions of peri- infarct zone fibrosis but there was no difference between the two treatment groups (see Figure below). However, myocardial interstitial fibrosis was similar (data not shown) in all groups suggesting that the benefit of treatment in this group was targeted toward the infarcted tissue and not to the overall left ventricle. Conclusion: LCZ69 significantly improved LV function during heart failure in the setting of hypertension with concomitant reductions in border zone fibrosis although overall left ventricular fibrosis was consistent in all groups. The reductions in collagen deposition seen in the LCZ group were similar to those seen by valsartan group suggesting that the additional cardiovascular benefits of LCZ696 are not due to changes in fibrosis.

083 Reversible expression of cardiac MyBP-C using an inducible tet-off system Jasmine Giles1, Adam Miller1, Daniel Fitzsimons1, Richard Moss1, 1University of Wisconsin, Madison, WI, USA

Loss of cMyBP-C has been linked to left ventricular dilation, cardiac hypertrophy and impaired ventricular function in both cMyBP-C constitutive and conditional knockout mice. At present, it remains unclear whether the structural and functional phenotypes associated with the MYBPC3 null mouse are reversible. To test this idea, we generated a cardiac-specific transgenic mouse model using a Tet-Off inducible system to permit the controlled expression of wild-type (WT) cardiac myosin binding protein-C (cMyBP-C) on the MYBPC3 null background. Functional cMyBP-C Tet-off mice were generated by crossing tetracycline transactivator mice with responder mice carrying the WT cMyBP-C transgene. Prior to dietary DOX administration, cMyBP-C expression and left ventricular function in cMyBP-C Tet-off mice were not statistically different from WT mice. Introduction of dietary doxycycline (DOX) for four weeks resulted in a partial knock-down of cMyBP- C expression and commensurate impairment of systolic and diastolic function to levels approaching that observed in MYBPC3 null mice. Subsequent withdrawal of DOX from the diet resulted in the re-expression of cMyBP-C to levels comparable to that observed in WT mice, along with the near complete recovery of in vivo cardiac function. These results indicate that the cardiac phenotypes associated with the MYBPC3 null mouse are reversible, at least in part, and furthermore, validate the use of the Tet-Off inducible system as a means to study the mechanisms underlying hypertrophic cardiomyopathy.

084 The hypertrophic cardiomyopathy-causing W792R and T1075 mutations in cardiac myosin binding protein-C generate cardiac dysfunction in mice

Thomas Lynch IV1, Jasmine Giles1, Elizabeth Iverson1, Daniel Fitzsimons1, Richard Moss1, 1University of Wisconsin, Madison, WI, USA

Background: Mutations in the MYBPC3 gene, which encodes the contractile regulatory protein cardiac myosin binding protein-C (cMyBP-C), account for approximately 40% of known cases of hypertrophic cardiomyopathy (HCM). Therefore, elucidating the pathogenicity of HCM-causing cMyBP-C mutations is critical for understanding how they contribute to the development of HCM. Objective: Our study aimed to determine the functional effects of two distinct HCM-causing mutations in MYBPC3-encoded cMyBP-C, a missense mutation in the C6 domain (W792R) and a C-terminal truncation of the C9/C10 domain (T1075 fs/5). Methods and Results: Cardiac specific transgenic mouse models were developed using a Tet-Off inducible system allowing for the controlled expression of W792R and T1075 cMyBP-C on the MYBPC3 null background. Functional cMyBP-C Tet-off (FT) mice were generated by crossing tetracycline transactivator mice with responder mice carrying the W792R and T1075 transgenes, which were compared to FT-WT controls. Short-axis M-mode echocardiography identified depressed percent ejection fraction and fractional shortening and increased left ventricular chamber size in hearts from FT-W792R and FT-T1075 mice compared to FT-WT controls. SDS-PAGE using cardiac myofibrillar fractions demonstrated a reduction in the expression of total W792R full-length and T1075 truncated cMyBP-C transgenic proteins compared to WT transgenic cMyBP-C. Immunofluorescence analysis of cardiac tissue sections revealed that the WT and W792R transgenic proteins localized in the classic cMyBP-C doublet pattern within cardiac sarcomeres. Intriguingly, the T1075 transgenic protein localized at the Z-lines within cardiac sarcomeres, suggesting a contrast in the localization patterns of cMyBP-C missense-mutated and truncated proteins. Conclusions: These results demonstrate that the W792R and T1075 cMyBP- C mutations generate cardiac contractile dysfunction in mice, although the molecular mechanisms of dysfunction differ between the two mutations, in that W792R is a missense mutation with normal localization of cMyBP-C and T1075 is a truncation mutation with reduced expression and altered sites of binding of cMyBP-C.

085 Phosphorylation of cMyBP-C accelerates the rate of force relaxation in murine skinned myocardium Jitandrakumar Patel1, Daniel Fitzsimons1, Richard Moss1, 1University of Wisconsin, Madison, WI, USA

Previous studies from our laboratory have demonstrated that ablation of cMyBP-C (i.e., null myocardium) or PKA treatment of wild-type (WT) myocardium similarly accelerate cross-bridge cycling kinetics, as well as the rate of force development in living myocardium. However, these interventions differ in their effects on the rate of relaxation in living myocardium, i.e., relaxation is slowed in null myocardium and accelerated in WT myocardium treated with a β1-adrenergic agonist. Studies were done on skinned myocardium to determine the direct effects on myocardial relaxation kinetics due to ablation or PKA phosphorylation of cMyBP-C. Relaxation from steady- state Ca2+-activated force was initiated following flash photolysis of the photolabile Ca2+ chelator diazo-2. Relaxation transients were resolved into two phases: an initial linear phase followed by a double-exponential phase with rate constants k1 (i.e., reflecting the kinetics of ADP release) and k2 (i.e., reflecting the slowing of relaxation kinetics due to cooperative re-binding of cross-bridges during relaxation). Under control conditions, k1 was significantly faster while k2 was significantly slower in cMyBP-C null myocardium compared to values observed in WT myocardium at all levels of submaximal activation. In WT myocardium, PKA treatment accelerated k1 and slowed k2

to values similar to that exhibited in cMyBP-C null myocardium under control conditions. PKA treatment had no effect on k1 and k2 values in cMyBP-C null myocardium. These results indicate that myocardial relaxation is accelerated due to ablation or phosphorylation of cMyBP-C. Although enhanced cooperative cross-bridge binding due to cMyBP-C phosphorylation would be predicted to slow relaxation, the phosphorylation-dependent increase in the rate of ADP release appears to dominate in accelerating the overall rate of relaxation. Furthermore, the previous observation that the rate of relaxation is slowed in living null myocardium is presumably due to prolongation of the Ca2+

transient that is also observed in these preparations.

086 Ablation of cMyBP-C eliminates the activation-dependence of unloaded shortening velocity at low levels of Ca2+ activation Daniel Fitzsimons1, Jitandrakumar Patel1, Richard Moss1, 1University of Wisconsin, Madison, WI, USA

At low levels of Ca2+ activation, the time-course of unloaded shortening velocity (Vo) in striated muscle is biphasic, i.e., shortening is comprised of an initial high-velocity phase and a subsequent low-velocity phase. The

Page 61: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

60

respective velocities of both the fast and slow phases are known to scale with the level of activation, culminating in a single high-velocity phase at saturating Ca2+ (i.e., Vmax). The molecular mechanism underlying the activation- dependence of Vo most likely involves a shortening-induced cooperative inactivation of the thin filament that results in reduced numbers of strongly- bound cross-bridges, giving rise to the slow phase of unloaded shortening. Cardiac myosin binding protein-C (cMyBP-C) is believed to modulate the cooperative activation of the thin filament by binding to myosin and/or actin and thereby regulating the probability of cross-bridge binding to actin. To test the idea that cMyBP-C contributes, at least in part, to the activation- dependence of unloaded shortening velocity, we measured Vo in skinned trabeculae isolated from wild-type (WT), cTnI-Ala2 (i.e., Ser23,24Ala) and cTnI-Ala2 x cMyBP-C-/- mice. During maximal activation, Vmax was monophasic and not significantly different among the three groups. However, at low levels of activation, biphasic shortening was observed in both the WT and cTnI-Ala2 myocardium, but only the high-velocity phase was observed in the cTnI-Ala2 x cMyBP-C-/- myocardium. Treatment of WT and cTnI-Ala2 trabeculae with PKA accelerated both the low- and high-velocity phases of shortening during submaximal activation, but did not eliminate the low-velocity phase. Thus, phosphorylation of cMyBP-C but not cTnI was required to evoke these effects. The results can be explained in terms of a model in which cMyBP-C regulates the cooperative binding of myosin cross-bridges to actin with the degree of cooperative propagation dependent on the phosphorylation state of cMyBP-C.

087 A missense mutation within the C6-domain of cMyBP-C results in cardiac enlargement and depressed ventricular function Jitandrakumar Patel1, Jasmine Giles1, Adam Miller1, Daniel Fitzsimons1, Richard Moss1, 1University of Wisconsin, Madison, WI, USA

Cardiac myosin-binding protein-C (cMyBP-C) plays a key role in determining the force and kinetics of myocardial contraction, primarily by binding to either myosin subfragment-2 or actin or both. By modulating the probability of myosin cross-bridge binding to actin, cMyBP-C normally acts to govern the kinetics of force development and relaxation. To determine the effects on cardiac morphology and function due to a novel cMyBP-C W792R missense mutation associated with clinical hypertrophic cardiomyopathy in humans. We generated cMyBP-C-W792R heterozygous and homozygous knock-in mice to investigate the structural and functional phenotypes associated with a missense mutation within the C6-domain of cMyBP-C. Compared to wild-type, homozygous W792R knock-in hearts exhibited significant ventricular and atrial dilation. While the W792R mutant protein was expressed at equivalent levels as in wild-type mice, the extent of phosphorylation of the mutant cMyBP-C protein was markedly reduced. Measurements of steady-state force and cross-bridge cycling kinetics showed that homozygous cMyBP-C W792R skinned myocardium exhibited a significant increase in the Ca2+-sensitivity of force and accelerated cross-bridge cycling kinetics (ktr) at maximal levels of Ca2+-activation, compared to WT myocardium. Cardiac function, as assessed by transthoracic echocardiography, revealed depressed indices of systolic and diastolic ventricular function in the homozygous W792R KI mice. These results demonstrate that a mutation within the mid-region of cMyBP-C can disrupt the dynamic regulation between myosin and actin that is normally governed by cMyBP-C, thereby leading to aberrant cardiac enlargement and commensurate diminution of in vivo ventricular function.

088 The mAKAP complex orchestrates the dephosphorylation of MEF2D in muscle cells to stimulate its activity Shania Aponte París1, Michael Kapiloff1, Kimberly Dodge-Kafka1, 1University of Connecticut, Farmington, CT, USA

Myocyte Enhancer Factor 2D (MEF2D) is a transcription factor required for the development of pathological remodeling as well as the induction of cardiac hypertrophy by pressure overload. However, little is known about how this transcription factor is regulated. Previous work showed that the scaffolding protein mAKAP (muscle A-Kinase Anchoring Protein) orchestrated the calcineurin (CaN)-mediated stimulation of MEF2D in skeletal muscle cells. CaN and mAKAP immunoprecipitate with MEF2D in skeletal muscle cells and primary neonatal cardiac myocytes (RNV); MEF2D and CaN association was dependent on mAKAP expression. Furthermore, expression of anchoring disrupting peptides, which displaced CaN binding to mAKAP, blocked the induction of MEF2D gene transcription in skeletal myoblasts as well as the induction of cardiac hypertrophy in RNV, demonstrating the importance of the mAKAP complex for stimulation of MEF2D activity. The goal of this study was to elucidate the molecular mechanism that underlies the regulation of MEF2D activity by mAKAP-bound CaN. Stimulation of C2C12 cells with differentiation media induces the binding of active CaN to mAKAP, resulting in the dephosphorylation of MEF2D at Serine 444. Our current focus is on

determining if Serine 444 dephosphorylation results in the desumoylation of MEF2D, allowing for increased gene transcription. Overall, our data suggests that the mAKAP complex focuses the actions of CaN onto MEF2D, to promote regulation of its post-transcriptional modifications, which we propose will allow for the increased transcriptional activity seen in both stimulated skeletal muscle myoblasts and stressed cardiac myocytes. Understanding the regulation and activity of the transcription factor MEF2D and its associated proteins is a crucial step towards the ability to block the development of cardiac hypertrophy.

089 Integrated omics analysis of isoform switching under cardiac hypertrophy Maggie Lam1, Peipei Ping1, Yi Xing1, 1UCLA, Los Angeles, USA

Background: Alternative isoform switching is a critical molecular phenotype implicated in multiple heart diseases. Notable examples of differential alternative isoform expressions include sarco/endoplasmic reticulum Ca2+- ATPase (SERCA2a/2b), cardiac sodium channel SCN5A, and titin. Recent advances in RNA sequencing technologies have enabled the profiling of alternative isoforms at the transcriptomics level. However, the physiological consequence of alternative isoform expression under diseases remains largely unknown. In this study, we utilize a multi-omics approach combining both transcriptomics and proteomics technologies to elucidate protein isoform switching under cardiac hypertrophy. Methods: Using a publicly available GEO RNA-seq dataset on C57BL/6 mouse heart, we derived the expression of alternative isoforms through an in-house proteotranscriptomics pipeline (ProteoSeq). An in-house proteomics dataset on an isoproterenol-stimulated cardiac hypertrophy model was searched against the custom protein sequence database generated through our pipeline to identify alternatively spliced protein isoforms. Results: Using our integrated omics approach, we identified 816 distinct splice junction peptides in the mammalian heart. These correspond to ~50 proteins expressing 2 or more splice isoforms. Our analysis has provided, for the first time, protein-level evidence of many of the alternative splicing events under cardiac hypertrophy. Quantification efforts have also revealed differential expression and turnover of alternative isoforms under disease conditions (e.g., pyruvate kinase, alpha-enolase). Conclusion: We obtained protein-level evidence of isoform switches under cardiac hypertrophy. Our integrated omics approach has the potential to further expand knowledge of alternative splicing events in heart diseases beyond cardiac hypertrophy.

090 MANF, a structurally unique redox-sensitive chaperone, restores ER- protein folding in the ischemic heart. Adrian Arrieta1, Erik Blackwood1, Winston T. Stauffer1, Michelle Santo Domingo1, Amber N. Pentoney1, Donna J. Thuerauf1, Shirin Doroudgar2,3, Christopher C. Glembotski1, 1San Diego State University, San Diego, CA, USA, 2Department of Cardiology, Angiology, and Pneumology, University Hospital Heidelberg, Innere Medizin III, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany, 3DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany

Rationale: In cardiomyocytes, most secreted and membrane proteins are synthesized and folded in the sarcoplasmic/endoplasmic reticulum (SR/ER). We previously showed that during ischemia, decreased oxygen creates a reducing environment in the SR/ER, preventing protein disulfide isomerases (PDIs) from forming disulfide bonds in nascent proteins, causing ER stress– the toxic accumulation of unfolded proteins which contributes to cardiomyocyte death. Upon ER stress, the transcription factor, ATF6 induces chaperones that restore SR/ER protein folding. We found that ATF6 induces mesencephalic astrocyte-derived neurotrophic factor (MANF), a recently identified protein of unknown function. MANF is structurally unique, so its function cannot be inferred from other proteins. Since MANF ATF6-inducible, ER-localized, and contains a conserved redox-sensitive motif found in PDIs, we hypothesized that MANF is a redox-sensitive chaperone that optimizes cardiomyocyte viability during ischemia. Methods: The redox status of MANF during reductive ER stress, and ability to bind misfolded proteins during ischemia were assessed in neonatal rat ventricular myocytes (NRVM). The ability of recombinant MANF to suppress misfolded protein aggregation was examined in an in vitro chaperone assay. Finally, the effects of MANF loss-of- function in the ischemic heart, in vivo, were determined by generating a transgenic mouse that expresses a cardiomyocyte-specific MANF-targeted microRNA. Results: In NRVM subjected to ER stress MANF was as sensitive to changes in ER redox status as the sentinel PDI, PDIA1 and formed disulfide-linked complexes with misfolded proteins during ischemia-mediated ER stress. Under reducing conditions, recombinant MANF suppressed aggregation of model misfolded proteins, in vitro. MANF knockdown in the heart, in vivo, increased damage from myocardial infarction, and an AAV9-

Page 62: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

61

based gene therapy approach rescued the effects of MANF deficiency, in vivo. Conclusions: MANF is a redox-sensitive SR/ER-resident chaperone that is a critical contributor to SR/ER protein folding during the adaptive ER stress response and decreases tissue damage in the ischemic heart.

091 Loss of caveolin-1 alters cardiac mitochondrial function and increases susceptibility to stress Jan M Schilling1,2, Mehul Dhanani1,2, Kristofer J Haushalter1,2, Sarah A Howell1,2, Ravina Verma1,2, Ingrid R Niesman1, Alice E Zemljic-Harpf1,2, Hemal H Patel1,2, 1University of California, San Diego, CA, USA, 2VA San Diego Healthcare System, San Diego, CA, USA

Introduction: Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in the United States. Caveolae, membrane micro- domains enriched in cholesterol, glycosphingolipids, sphingomyelin and the protein caveolin, are critical to ischemic tolerance. We have previously described a specific role for caveolin-3 (Cav-3) in cardiac protection via regulation of mitochondrial function; however, little is known about the role of caveolin-1 (Cav-1) in cardiac mitochondrial bioenergetics. We hypothesized that Cav-1 is crucial in maintaining healthy cardiac mitochondria. Material and Methods: We performed a series of experiments to assess the role of Cav-1 upon mitochondrial functionality, comparing heart tissues and isolated mitochondria from wildtype (WT) and global Cav-1 knock-out (KO) mice. Percoll-purified WT mitochondria were immunogold labeled and imaged by TEM to localize Cav-1 protein to specific mitochondrial membranes. Furthermore, oxygen consumption was evaluated in isolated papillary muscle fibers by high-resolution respirometry using Oroboros O2k oxygraph. Superoxide free radical production was investigated by electron paramagnetic resonance imaging (EPR) using the spin probe DEPMPO in isolated mitochondria. Results: Immunogold labeling showed Cav-1 localization to outer and inner mitochondrial membranes. In Cav-1 KO mice relative oxygen flux was slightly increased in the presence of complex I substrates and ADP. Furthermore, in Cav-1 KO mice, cytochrome c response (a marker for outer mitochondrial damage) was significantly correlated to total complex I & II respiration (malate, pyruvate, glutamate, ADP, and succinate) with increased respiration at higher cytochrome c responses, indicating potential outer mitochondrial membrane damage. ROS was decreased in Cav-1 KO mice during state IV respiration with complex I & II substrates measured by EPR suggesting overall decreased mitochondrial function. Conclusion: Cav-1 may be a critical regulator of cardiac mitochondrial function. Cav-1 may therefore be a viable therapeutic target to alter cardiac mitochondrial bioenergetics.

092 Top-down proteomics for assessing the maturation of stem cell-derived cardiomyocytes Wenxuan Cai1, Jianhua Zhang1, William J. de Lange1, Zachery Gregorich1, J. Carter Ralphe1, Timothy Kamp1, Ying Ge1, 1University of Wisconsin-Madison, Madison, WI, USA

Background: Recent advancements in human pluripotent stem cell (hPSC) technology and cardiomyocyte (CM) differentiation have empowered research in cardiac development and diseases. However, the relative immaturity of stem cell-derived CMs is well documented. Since the expression of sarcomeric protein isoforms, as well as their post-translational modifications (PTMs), change as CMs mature, we sought to develop a method utilizing powerful top-down high-resolution mass spectrometry (MS) to assess the relative maturity of CMs focusing on the expression of sarcomeric protein isoforms and their PTMs. Methods: hPSCs were differentiated to CMs using a small molecule directed protocol and cultured in either 2D monolayer or 3D engineered cardiac tissue (ECT). Sarcomeric proteins were extracted from the CMs and analyzed using a top-down proteomics platform coupling reverse phase chromatography to high-resolution MS. Results: We identified and quantified the major sarcomeric protein isoforms and their PTMs in CMs from 2D monolayer or 3D ECT culture. Slow skeletal troponin I (ssTnI) was highly expressed while cardiac troponin I (cTnI) was under detection limit in 2D-CMs at both day 30 and 60, indicating the relative immaturity of 2D-CMs by day 60. However, the relative expression of the ventricular regulatory light chain versus the atrial isoform in CMs increased by about 7-fold at day 60 compared to the 2D-CMs at day 30, confirming that CMs exhibit maturation with longer 2D culture as anticipated. Tropomyosin phosphorylation decreased in CMs with longer 2D culture, which may serve as a novel marker for assessing CM maturation. CMs cultured in 3D ECT for 42 days after 30 days in 2D culture exhibited a 2-fold increase in cTnI expression relative to ssTnI, compared to day-72 CMs maintained in 2D culture, suggesting that 3D culture can promote CM maturation. These results demonstrate the power of top-down proteomics to assess the maturation of hPSC-derived CMs.

093 Regulation of ryanodine receptor mediated perinuclear calcium by the mAKAP complex Moriah Gildart1, Michael Kapiloff2, Kimberly Dodge-Kafka1, 1University of Connecticut Health Center, Farmington, Connecticut, USA, 2University of Miami Miller School of Medicine, Miami, Florida, USA

The cAMP pathway is the major stress pathway responsible for activating proteins that cause calcium release leading to cardiac hypertrophy. A key player in these signaling events is a nuclear envelope associated scaffold protein known as muscle A-Kinase Anchoring Protein (mAKAP), which binds protein kinase A (PKA) and the calcium channel the ryanodine receptor (RyR2) and localizes them to the nuclear envelope. The mAKAP signalosome coordinates calcium signaling at the perinuclear space, and this pool of calcium leads to activation of hypertrophic transcription factors. Although a multitude of work has implicated mAKAP in hypertrophy development, the source of calcium modulated by mAKAP is unknown. Our central hypothesis is that stimulation of the cAMP pathway leading to activation of mAKAP-bound PKA will phosphorylate mAKAP-bound RyR2 leading to release of perinuclear calcium. We utilized a FRET based sensor localized to the mAKAP complex to measure perinuclear calcium levels in neonatal rat cardiomyocytes in real time. The cardiomyocytes were treated with a stimulatory and inhibitory concentration of ryanodine, in order to specifically affect RyR2. Nanomolar concentrations of ryanodine caused calcium increase at the nuclear envelope, and micromolar concentrations caused no effect. To test that PKA activation also causes calcium release, we treated the cells with forskolin, a drug that activates adenylyl cyclase leading to PKA activity. Results show that forskolin does cause calcium increase at the nuclear envelope. Finally, to show the relationship of PKA activation to calcium release by the ryanodine receptor, we treated the cells with forskolin followed by micromolar concentrations of ryanodine, which eliminated calcium release. This supports that RyR2 releases calcium modulated by mAKAP and activation of PKA is needed to stimulate this release. Since targeting mAKAP is not feasible therapeutically, this finding is significant in the search for new targets for therapies to prevent development of cardiac hypertrophy.

094 Cardiac muscle function across the natural history of a genetic minipig model of hypertrophic cardiomyopathy Marcus Henze1, Robert Anderson1, Fiona Wong1, Robert Weiss2, Abhay Divekar2, David Meyerholz2, Ferhaan Ahmad2, Eric Green0, 1MyoKardia, Inc., South San Francisco, CA, USA, 2University of Iowa, Iowa City, IA, USA

Hypertrophic cardiomyopathy (HCM) is a heritable disease of the myocardium characterized by hypertrophy and ventricular stiffening, resulting in reduced ventricular filling and exercise capacity. With a prevalence of 1:500, HCM is a common cause of disabling cardiac symptoms and sudden cardiac death (SCD). The first discovered and most studied mutation linked to HCM in humans is the MYH7-R403Q mutation. To enable detailed characterization of this mutation at the cellular, molecular, and organismal levels, we have developed a transgenic minipig expressing the R403Q mutation. These animals rapidly develop an in vivo phenotype consistent with HCM; specifically altered systolic/diastolic function, hypertrophy, myocyte disarray, fibrosis, and elevated circulating TnI and BNP. At 3-months of age (juvenile), R403Q pigs show early signs of HCM with increased ejection fraction (EF), impaired measures of diastole, interstitial fibrosis, and myocyte disarray. By 9- months (late adolescence), the phenotype progresses to normal EF with furthering diastolic impairment, fibrosis, and disarray, followed by increased mortality from SCD. Here we investigated the in vitro properties of cardiac muscle function in pigs expressing the MYH7-R403Q mutation and the effects of small molecule modulators of myosin in these tissues. With age, wild-type (Wt) muscle fibers showed progressive increased maximal tension and Ca2+

cooperativity with decreased Ca2+ sensitivity. R403Q pigs follow a similar tension/pCa profile to Wt with ageing, however, with a consistent profile of increased Ca2+ sensitivity accompanied by decreased Ca2+ cooperativity and maximal tension. Mutant pig fibers showed an increased rate of tension redevelopment compared to age-matched Wt fibers, indicating an increased rate of crossbridge cycling. Together, these data are the first to show mechanistic evidence for hypercontractile muscle function across the natural history of a large-animal model of HCM. Detailed mechanistic knowledge of cardiac muscle function in this model will be an invaluable tool in the development of therapies for HCM.

095 The muscle-specific ubiquitin ligase Atrogin-1 (MAFbx) inhibits age- associated cardiac fibrosis by enhancing MMP-9 levels in vivo Traci Parry1, Roberto Mota1, Monte Willis1, 1Univ of North Carolina, Univ of North Carolina, USA

Page 63: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

62

Background: The muscle-specific Atrogin-1 associates with Skp1, Cul1, and Roc1 to assemble an SCF(Atrogin-1) complex with ubiquitin ligase activity. In the context of pathological cardiac hypertrophy, Atrogin-1 attenuates agonist- induced calcineurin activity, repressing NFAFc4 transactivation and translocation. In physiological cardiac hypertrophy, Atrogin-1 inhibits Akt- dependent cardiac hypertrophy by ubiquitin-dependent coactivation of Forkhead proteins. In the present study, we identified the role of Atrogin-1 in age-associated function and remodeling. Methods: Transgenic cardiomyocyte-specific (alphaMHC-) Atrogin-1 mice, previously characterized by our group, were followed by conscious echocardiography for 18 months. Mice were harvested and analyzed by histology, Western blot analysis, and RT-qPCR for alterations in age-associated remodeling. Results: No significant differences in cardiac function were identified in Atrogin-1 Tg+ mice compared to sibling wildtype controls at four months of age. However, at 12 months of age, Atrogin-1 Tg+ hearts were significantly larger, with anterior wall thickness 1.40+/-0.6 mm (diastole) and 1.38+/-0.03 mm (systole) compared to wildtype (1.10+/-0.02 mm diastole, 1.15+/-0.04 mm systole). Histological analysis of trichrome-stained wildtype hearts at 18 months of age revealed ~4.5+/-0.5% collagen deposition; in contrast, Atrogin-1 Tg+ hearts had significantly less collagen deposition (~1.5%+/-0.25%). By RT-qPCR, Atrogin-1 Tg+ had increased MMP-9 mRNA, suggesting a role for Atrogin-1 in regulating MMP-9 transcriptionally. Conclusions: Chronic increased Atrogin- 1 in cardiomyocytes alters age-associated function, which reducing fibrosis detected histologically, which may be related to alterations in MMP-9. These findings indicate a novel role of muscle-specific Atrogin-1 in preventing fibrosis, identifying for the first time a muscle-specific anti-fibrotic target in the aging heart.

096 Curation and Phenotyping of Cardiovascular Case Reports Achieved by ICD Based Index System and MeSH Supported Query Platform Yijiang Zhou1,2, David Liem1,2, Quan Cao1,2, Jessica Lee1,2, Wei Wang1,3, Alex Bui1,4, Karol Watson1,2, Jiawei Han5, Peipei Ping1,2, 1The NIH BD2K Center of Excellence at UCLA, Los Angeles, California, USA, 2Departments of Physiology, Medicine/Cardiology, Bioinformatics, University of California at Los Angeles, Los Angeles, California, USA, 3Departments of Computer Science, University of California at Los Angeles, Los Angeles, California, USA, 4Departments of Radiology, University of California at Los Angeles, Los Angeles, California, USA, 5NIH BD2K KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA

Background: Accurate phenotyping of patients and efficient extraction of detailed clinical information from large amounts of accumulated case reports in open resources have been challenging, in part, due to inadequate Medical Subjects Headings (MeSH) term and insufficient indexing systems. To overcome this challenge, we have created a novel indexing system integrating MeSH terms together with the current edition of the International Classification of Diseases (ICD-10). Methods: We have manually selected case reports using a combination of four MeSH terms: "myocardial Infarction", "coronary angiography", "echocardiography" and "shock, cardiogenic". In parallel, we extracted clinical features using ICD-10-based indexing, which combines the codes from ICD-10 and from International Classification of Health Interventions (ICHI). An evaluation metrics (100 total) consisting of key clinical information was assembled; including 1) symptoms; 2) demographics; 3) lifestyle, 4) family history; 5) medical history; 6) diagnostic tests; 7) pathology; 8) treatment interventions; 9) drug therapies; 10) diagnosis; and 11) outcomes. Subsequently, we compared the ICD based index with that of MeSH supported query applying this metrics (11 components) using Wilcoxon Signed-Rank Test. Results: A total of 46 case reports were identified using the aforementioned indexing strategy. Ten case reports on “ST-segment elevation acute myocardial infarction” were analyzed. Our results show that ICD-10-based indexing scored higher than MeSH-supported indexing in overall setting (51.8 vs 38.3, p<0.05). Furthermore, ICD-10 based indexing also exhibits better accuracy in components of “symptoms and signs” (p<0.05), “past medical history” (p<0.05), “treatment interventions” (p<0.05) and “diagnosis” (p<0.05). However, MeSH indexing scored higher in “demographics” (p<0.05), “pathology and pathophysiology” (p<0.05) and “outcomes” (p<0.05). Conclusion: ICD-10-based indexing scored higher than MeSH for extracting key clinical information; the two systems emphasize on distinct clinical features. Integration of ICD-10 and MeSH terms would offer better precision and comprehensiveness to accurately phenotype and curate case reports.

097 Construction a Standardized Metadata Template to Extract Relevant Biomedical Insights from Clinical Case Reports

Yijiang Zhou1, David A. Liem1, Quan Cao1, Jessica Lee1, Wei Wang2, Alex Bui3, Karol Watson4, Jiawei Han5, Peipei Ping6, 1The NIH BD2K Center of Excellence at UCLA, Departments of Physiology, Los Angeles, California, USA, 21The NIH BD2K Center of Excellence at UCLA, Department of Computer Science, University of California at Los Angeles, CA 90095, USA, Los Angeles, California, USA, 31The NIH BD2K Center of Excellence at UCLA, Departments of Radiology, University of California at Los Angeles, CA 90095, USA, Los Angeles, California, USA, 41The NIH BD2K Center of Excellence at UCLA, Departments of Medicine/Cardiology, University of California at Los Angeles, CA 90095, USA, Los Angeles, California, USA, 5NIH BD2K KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA, Urbana-Champaign, Illinois, USA, 61The NIH BD2K Center of Excellence at UCLA, Departments of Physiology, Medicine/Cardiology, Bioinformatics, Computer Science, and Radiology, University of California at Los Angeles, CA 90095, USA, Los Angeles, California, USA

Background: Clinical case reports (CR) have a time-honored tradition in serving as an important means of sharing clinical experiences on patients presenting with atypical disease phenotypes or receiving new therapies. However, the huge amount of accumulated case reports are isolated, unstructured, and heterogeneous data, posing a great challenge to clinicians and researchers in mining relevant information through existing indexing tools. In this investigation, we analyzed the current publication trends of case reports, their disease specific citation impact, and summarized the merits and limitations of CR genre with respect to their utilities to future knowledge production in cardiovascular medicine. Importantly, the results of our analysis highlight the importance of a standardized metadata template and metrics to guide and evaluate case report contents and metadata; as wel as to better index, annotate, and curate online case reports to support innovations and discoveries in the biomedical community. Methods: To construct a metadata template and metrics we categorized three major metadata components: (i) case report identification; e.g., title, author, PMID/DOI numbers; (ii) medical content; e.g., disease diagnosis, signs and symptoms, diagnostic procedures, and therapies; and (iii) other; e.g., references, funding source, award numbers, and disclosures. We analyzed the CR metadata retrieved from MeSH term & Pubmed indexing using these metrics. Results and Conclusion: Among the 100 manually screened CRs, 65% of them are missing key metadata components with the majority missing in the “medical content” category (51% from key “medical content” components). Taken together, our study on the evalution and quantification of the exhisting metadata for CRs underscores the importance of creating a standardized metadata template, as well as to develop an automated metadata enrichment system to populate the metadata for all existing CRs (~1.8M) in open source. This task would be essential to achieve an enhanced digital curation of this largely untapped clinical resource.

098 Identification of cardiomyocytes' characteristics responsible for dynamical changes in calcium profile in response to mechano-chemo transduction Zana Coulibaly1, Rafael Shimkunas1, Bence Hegyi1, Zhong Jian1, Ye Chen- Izu1, Leighton Izu1, 1University of California, Davis, Davis, California, USA

Background: We embedded cardiomyocytes in a cross-linked hydrogel to study the effect of increased afterload on in the heart. We observed that mechanically-loaded cardiomyocytes undergo noticeable changes in their Ca2+ dynamics that can compensate for the increase in load. We have shown that these changes are mediated by the upregulation of nitric oxide (NO) signaling, which in turns affects Ca2+ handling. Because many different Ca2+

pathways could be affected we used an agnostic approach based on mathematical modeling to tease out changes in cell characteristics responsible for the difference in calcium and contraction profiles between load-free and after-load environments. Mathematical Method: We use a mathematical model to simulate the extracellular environment, and use the simulation results coupled with the experimental observations to identify parameters that change between load-free and after-load conditions. The mathematical model details a closed feedback loop between the calcium system and the extracellular environment to give rise to the self-regulation we observed. We ran extensive simulations where parameters associated with the influx and efflux of calcium are modulated such that they are either up- regulated or down-regulated by NO. In silico results are filtered out to qualitatively match cell-in-gel in vivo results. The filtering process is based on measures that capture multiple properties of calcium profiles. Conclusion: Our agnostic approach of identification hints that upregulation of NO modulates multiple parameters of the Ca2+ handling pathways simultaneously. Of the modulated parameters, the L-type current amplitude has to be consistently increased. Coupled with the increase in L-type current, cell’s parameters associated with release of calcium from the SR and uptakes of calcium by the SR have to be modulated in opposite directions.

Page 64: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

63

100 Aortic acceleration as noninvasive index of left ventricular contractility Anilkumar Reddy1,2, Celia Pena Heredia1, Thuy Pham1, George Taffet1, 1Baylor College of Medicine, Houston, Texas, USA, 2Indus Instruments, Webster, Texas, USA

Background: Left ventricular (LV) contractility is assessed with the invasively measured LV pressure derivative (+dP/dtmax), which is a terminal procedure in mice and other similar sized animals. Noninvasive methods to assess LV contractility have been reported in patients, sheep, dogs, and rats. We undertook this study to validate the use of acceleration of aortic flow velocity in lieu of +dP/dtmax to assess LV contractility in mice. Methods: We measured aortic blood flow velocity noninvasively simultaneous with invasive measurement of LV pressure in the anesthetized mouse. Mean, Ym (=Vp/Tp) and peak, Yp (=dV/dtmax) aortic accelerations were calculated and compared to +dP/dtmax (X) calculated from LV pressure. Measurements were made at baseline and post-dobutamine administration (1µg/g BW) in 4 wild type male mice (C57BL6/J). Results: Peak and mean aortic accelerations were compared to +dP/dtmax at several heart rates (range: 387 to 701 bpm, n=18 data points). Linear regression of both, peak aortic acceleration (Yp) versus +dP/dtmax (X) and mean aortic acceleration (Ym) versus +dP/dtmax (X), showed a high correlation - (Yp = 2.8X + 1617; p<0.0001; r2 = 0.896) and (Ym = 1.4X + 1319; p<0.0001; r2 = 0.903), respectively. Conclusion: The high correlation between Yp versus +dP/dtmax is in agreement with that reported in dogs, while that between Ym versus +dP/dtmax is similar to that reported in sheep. Mean acceleration requires measurement of time (Tp) to peak velocity (Vp) which can be difficult at times due to the ambiguity in the location of the peak velocity. Peak aortic acceleration is much easier to measure using the first derivative and is also highly correlated with +dP/dtmax. Preliminary result of this study suggest that both peak and mean aortic acceleration may be used as a noninvasive index of LV contractility in mice. We expect to evaluate these relationships further under various loading conditions.

101 Phrase Mining and Machine Learning in Textual Data to Uncover Distinct Protein Patterns in Cardiovascular Disease David A. Liem1, Vincent Kyi1, Yu Shi2, Fangbo Tao2, Jiawei Han2, Peipei Ping1, 1NIH BD2K Center of Excellence at UCLA, Departments of Physiology, Medicine and Bioinformatics, UCLA School of Medicine, Los Angeles, CA 90095, USA, Los Angeles, California, USA, 2NIH BD2K KnowEng Center, Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA, Urbana-Champaign, Illinois, USA

Background: Decades of successful biomedical research have led to the rapid accumulation of large-volume text data (unstructured data). Defining relationships and structuring connections of these fragmented data from the many published can uncover valuable information hidden in this treasure trove. However, the quantity of text data is overwhelming and manual extraction is a forbitten task. This situation demands the development of automated tools to effectively and accurately processing the giant volume of available textual data (e.g., ~27M on PubMed, ~160M on Google Scholar). Objectives: To address this challenge, we developed a text-mining and machine learning algorithm to dissect textual data on CVD and identify protein patterns in datasets to uncover valuable information. Methods: We applied a novel phrase mining workflow, Context-aware Semantic Online Analytical Processing (CaseOLAP), to recognize patterns from six CVD datasets based on their MeSH-terms: cerebrovascular accidents (CVA), cardiomyopathies (CM), ischemic heart diseases (IHD), arrhythmias (ARR), valvular heart disease (VHD) and Congenital Heart Disease (CHD). We analyzed the patterns of 8,325 cardiac proteins in 1.1 million publications (1995-2016). Results: Over 8,325 proteins only a subset exhibited high CaseOLAP scores indicating high relevance in CVD, mainly displayed in IHD, CM and CVA. We identified six high scoring protein clusters unique to one CVD group. A principle component analysis indicated that IHD, CVA and CM showed distinct protein scoring patterns while CHD, VHD and ARR were clustered. We identified 10 protein clusters shared between two or more CVD groups with biological functions in inflammation, contractility, blood coagulation, hemodynamic regulation, cytoskeletal organization and neurotransmission. Inflammatory proteins appeared to be relevant in all CVDs, while proteins in neurotransmission and memory processing were relevant in CVA, ARR, VHD, and CHD. Conclusions: Using CaseOLAP on textual data across six CVDs we gained novel insights into patterns and relationships of their proteins. This text-mining algorithm offers promising biomedical applications to facilitate patient studies in clinical trials, case reports and electronic health records.

102 Temporal Dynamics of Plasma Metabolites in ISO-induced Cardiac Remodeling in Mice

Quan Cao1,2, Howard Choi1,2, Ding Wang1,2, David Liem1,2, Chelsea Ju1,3, Jennifer Polson1,2, Wei Wang1,3, Peipei Ping1,2, 1NIH BD2K Center of Excellence at University of California, Los Angeles, Los Angeles, California, USA, 2Departments of Physiology, Medicine and Bioinformatics, UCLA, Los Angeles, California, USA, 3Departments of Computer Science, UCLA, Los Angeles, California, USA

Background: Accumulating evidence suggests that maladaptive cardiac remodeling, a common stage of many heart diseases, is associated with disrupting levels of multiple metabolites both in the heart and in the whole body. Isoproterenol (ISO) is a neurohormonal factor associated with pathologic remodeling of the heart. Hence, we utilized an ISO-induced cardiac hypertrophy mouse model, in which cardiac function was compromised by continuous β-adrenergic overstimulation, to capture the plasma metabolomics fingerprint during maladaptive cardiac remodeling. Methods: We conducted ISO-treatment on six genetic mouse strains displaying variable susceptibilities towards ISO-induced hypertrophy over fourteen days. Employing plasma metabolomics profiling by Multiple-Reaction-Monitoring (MRM)-based MS quantitative platform, we obtained a temporal dataset containing 6 mouse strains, 6 time points, and 610 metabolites from 20 biological classes for statistical analysis. A paired t-test (adjusted p<0.001; ±1.5-fold as the threshold) was utilized to identify metabolites sensitive to the progression of cardiac remodeling. A Linear Mixed Model (LMM) was employed to correlate temporal metabolomics dynamics with HW/BW ratios. Results: We have established a criterion to evaluate candidate metabolites. A metabolite must fulfill the criteria for both the paired t-test and LMM to be considered as a biomarker candidate. In this study, 4 free fatty acid metabolites were identified, including Myristic acid, Palmitoleic acid, Oleic acid and α-Linolenic acid. Six mouse strains were divided into resilient group and susceptive group based on their temporal changes in HW/BW ratio. In mouse strains sharing similar susceptibility to ISO treatment, the 4 metabolites displayed similar temporal patterns in plasma levels during maladaptive remodeling. Conclusion: The temporal profiles of the four identified plasma metabolites demonstrated their potentials as molecular signatures to differentiate ISO- resilient mouse strains from ISO-susceptible ones. Our results suggest that this novel workflow is capable of identifying significant metabolic changes that are associated with the cardiac phenotypes and outcomes.

103 The spatial distribution of the Na+/Ca2+ exchanger in cardiac mitochondria enhances the efficiency of mitochondrial Ca2+ signal generation Sergio De La Fuente1, Celia Fernandez-Sanz1, Jonathan Lambert2, John Elrod2, Shey-Shing Sheu1, Gyorgy Csordas1, 1Thomas Jefferson University, Philadelphia, PA, USA, 2Temple University, Lewis Katz School of Medicine, Philadelphia, PA, USA

Control of myocardial energetics by mitochondrial matrix Ca2+ signals includes local Ca2+ delivery from SR-resident ryanodine receptors (RyR2) to the mitochondrial Ca2+ uniporter (mtCU). Ca2+ is extruded primarily by the robust Na+-Ca2+ exchanger (NCLX). We have recently reported that the relatively scarce mtCU is strategically positioned in cardiac muscle at the mitochondria- junctional SR (jSR) association to support the local Ca2+ signals. We also established a fractionation-based model system to compare mitochondrial segments rich in jSR association (mitochondria in the jSR fraction) with those that are not (purified mitochondria). Comparing the abundances of mitochondrial proteins between the mitochondrial and jSR fractions of mouse and rat heart showed that while mtCU pore constituents MCU and EMRE were enriched in the jSR fraction, the NCLX was strongly enriched in the mitochondrial fraction. We hypothesized, that the zone of mtCU hot spots at the dyad junctions was unfavorable for NCLX and that it would be localized at areas where mitochondria associate with the network SR. Consistently, NCLX abundance in the jSR fraction was lower than that in the microsomal or ‘MAM’ fractions. Mitochondrial 45Ca2+ uptake assays using isolated mitochondria or jSR fraction showed a massive enhancement of the net uptake upon pharmacological NCLX inhibition or omission of Na+ in the mitochondrial but not the jSR fraction. Direct measurement of the fractional Ca2+ release from the two membrane fractions also revealed more efficient NCLX-mediated Ca2+ extrusion in the mitochondrial fraction than in the jSR. Thus, in the cardiac muscle the relatively modest mitochondrial Ca2+ uptake and robust Ca2+ extrusion seem to be strategically but reciprocally distributed with respective ‘hot’ and ‘cold’ spots at the jSR interface. This arrangement likely supports the locally taken-up Ca2+ to give rise to matrix Ca2+ signals (to control metabolism) before being extruded towards the SERCA pumps of the network SR.

Page 65: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

64

104 Autologous bone marrow stem cell therapy in patients with ST-elevation myocardial infarction: a systematic review and meta-analysis Maya M. Jeyaraman1,10, Rasheda Rabbani1,10, Leslie Copstein1, Wasan Sulaiman1, Farnaz Farshidfar1, Hessam Kashani1, Sheikh M.Z. Qadar1, Qingdong Guan2,3, Becky Skidmore4, Elissavet Kardami5, John Ducas6, Samer Mansour7,8, Ryan Zarychanski1,9, Ahmed M. Abou-Setta1,10, 1George & Fay Yee Center for Healthcare Innovation, Winnipeg, Manitoba, Canada, 2Manitoba Center for Advanced Cell and Tissue Therapy, Winnipeg, Manitoba, Canada, 3Cellular Therapy Laboratory, CancerCare Manitoba, Winnipeg, Manitoba, Canada, 4Information Specialist Consultant, Ottawa, Ontario, Canada, 5Department of Human Anatomy and Cell Sciences, University of Manitoba, Winnipeg, Manitoba, Canada, 6Section of Cardiology, Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada, 7Centre Hospitalier de l’Université de Montreal, Montreal, Quebec, Canada, 8Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada, 9Department of Haematology and Medical Oncology, Cancer Care Manitoba, Winnipeg, Manitoba, Canada, 10Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada

Background: ST-elevation myocardial infarction (STEMI) is a major cause of morbidity and mortality worldwide despite treatment with primary percutaneous coronary intervention (PPCI). Randomized controlled trials (RCTs) investigating BMSCs to improve outcomes after PPCI, have reported conflicting results. Previous systematic reviews have been inundated with marked heterogeneity leading to misleading results, thus raising serious concerns. The primary objective of this systematic review was to identify, critically appraise and meta-analyze the best-available evidence on the efficacy and safety of intracoronary administration of autologous BMSC therapy in STEMI patients after PPCI. Methods: A systematic search of MEDLINE, PubMed, EMBASE, CENTRAL, Global Health, CINAHL and conference proceedings was conducted in February 2017. Two reviewers independently and in duplicate identified RCTs, extracted trial level data and assessed risk of bias. Primary outcome was all-cause mortality. Secondary and safety outcomes included cardiac death, incidence of heart failure, arrhythmias, repeat myocardial infarction or target vessel revascularizations; or improved health-related quality of life, LVEF, or infarct size. Summary relative risks in meta-analysis were obtained using random-effects model. Strength of evidence was assessed. Results: A comprehensive database search identified 42 RCTs (3,365 STEMI patients). BMSC therapy was not associated with a decrease in mortality (Absolute risk reduction (ARR) -0.8%; 40 trials; 3289 participants; I2 0%). Subgroup analysis indicated a potential industry bias. BMSC therapy did not significantly decrease the incidence of other important clinical outcomes such as cardiac death, incidence of heart failure, arrhythmias, repeat myocardial infarction or target vessel revascularizations; or improve health-related quality of life, LVEF, or infarct size. No major adverse effects were evident in either group. Conclusions: Based on the current evidence from 42 RCTs published in the past 15 years, we conclude that intracoronary administration of autologous BMSCs after PPCI has no significant beneficial effect in STEMI patients.

105 Fractionation of embryonic cardiac progenitor cells and evaluation of their differentiation potential Tiam Feridooni1, Kishore Pasumarthi1, 1Dalhousie University, Halifax, Nova

Scotia, Canada

Mid-gestation mouse ventricles (E11.5) contain a larger number of Nkx2.5+

cardiac progenitor cells (CPCs). The proliferation rates are consistently higher in CPCs compared to myocyte population of developing ventricles. Recent studies suggested that CPCs are an ideal donor cell type for replacing damaged tissue in diseased hearts. Thus, the ability to isolate and expand CPCs from embryos or stem cell cultures could be useful for cell fate studies and regenerative therapies. Since embryonic CPCs possess fewer mitochondria compared to cardiomyocytes, we reasoned that CPCs can be fractionated using a fluorescent mitochondrial membrane potential dye (TMRM) and these cells may retain cardiomyogenic potential even in the absence of cardiomyocytes. FACS sorting of TMRM stained embryonic ventricular cells indicated that over 99% of cells in TMRM-high fraction stained positive for sarcomeric myosin (MF20) and all of them expressed Nkx2.5. Although majority of cells present in TMRM-low fraction expressed Nkx2.5, very few cells (~1%) stained positive for MF20. Further culturing of TMRM-low cells over a period of 72 hrs showed a progressive increase in MF20 positive cells. Additional analyses revealed that MF20 negative cells in TMRM-low fraction do not express markers for fibroblasts (DDR2, vimentin), endothelial cells (vWF, CD31) or smooth muscle cells (SM myosin). Treatment of TMRM- low cells with known cardiogenic factors DMSO and dynorphin B as well as a nonselective beta-AR agonist isoproterenol (ISO) significantly increased the percentage of MF20+ cells compared to untreated cultures. Notably, the

morphology of a small fraction of MF20+ cells in ISO treated TMRM-low cultures resembled that of cultured Purkinje cells. Collectively, these studies suggest that embryonic CPCs can be separated as a TMRM-low fraction and their differentiation potential can be enhanced by exogenous addition of known cardiomyogenic factors. Future studies will focus on beta-AR responses and calcium handling properties of embryonic CPC and CM populations.

Page 66: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

65

Social Events & Dinner Reception

Information

On Tuesday, May 30th at noon, we’re hosting the ECI Luncheon at Drago’s Seafood Restaurant, conveniently located in- side the first floor of the Hilton Riverside Hotel. Famously known for their charbroiled oysters, Drago’s is the perfect setting for a “speed dating” lunch with Senior Investigators. Live, learn, and bon appetit!

On Wednesday, May 31st at 7:30pm, we’re hosting the ECI Social at the Napoleon House located in the French Quarter on 500 Chartres Street. Feel free to take a streetcar to this event. There is a Riverfront Streetcar Depot located near the front entrance of the Hilton Riverside Hotel. For only a $1.25 you can experience New Orleans’ famous streetcar and enjoy the Mississippi River views. Please depart at the Toulouse Street stop to arrive to Napoleon House, which is a block and a half

from this stop. Don’t forget to try Napoleon House’s famous Pimm’s Cup beverage and enjoy socializing with Pokemon Go!

On Thursday, June 1st at 6:30pm, a Dinner Reception and Award Banquet will be held at the Mercedes - Benz Super- dome’s Champion Square. Following this event will be a Social Celebration in the adjacent room, Club XLIV. Buses will be avail- able for transportation at the front entrance of the Hilton Riverside Hotel and will be departing promptly at 6pm. Bus transportation will also be available at the end of the event and will depart at 11:30pm. Let the good times roll and celebrate safely.

Page 67: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

66

2017 ISHR-NAS SPONSORS

PLATINUM SPONSORS

COBRE

Page 68: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

67

2017 ISHR-NAS SPONSORS

PLATINUM SPONSORS

GOLD SPONSORS

Page 69: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

68

2017 ISHR-NAS SPONSORS

IRIDIUM SPONSORS

Page 70: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

69

2017 ISHR-NAS SPONSORS IRIDIUM SPONSORS

Page 71: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

70

2017 ISHR-NAS SPONSORS

SILVER SPONSORS

Page 72: 36 th Annual...Srinivas Tapa, University of California, Davis 9. ABCG2-EXPRESSING CELLS FUSE WITH EXISTING CARDIOMYOCYTES, Amritha Yellamilli, University of Minnesota, Minneapolis

71

Concierge

Guest Room

Elevators

Riverfront Streetcar Depot

Grand Ballroom

Front Entrance

Grand Salon

World Trade Center Parking Garage

1st Floor Check-In Counters

Escalator Gift Shop

Pelican

Poy

dra

s St

reet

Compass

Bridge

Windward Escalator

Leeward

Chart

Quarter D

eck

Esc

alat

or

Reg

istr

atio

n

Hilton Riverside Hotel Floorplan

Steering

Mississippi River

River/ Port/ Starboard

Crescent City

Marketplace

Kabacoff

Riverside Complex Conference Area

First Level