110 results on '"Thomas P Cappola"'
Search Results
2. Clinical Phenogroups in Heart Failure With Preserved Ejection Fraction
- Author
-
Julio A. Chirinos, Zhaoqing Wang, Stuart B. Prenner, David A. Gordon, Payman Zamani, Zhuyin Li, Sarah J. Schrauben, Jordana B. Cohen, Dietmar A. Seiffert, Priyanka Bhattacharya, Thomas P. Cappola, Michael Basso, Lei Zhao, Mary Ellen Cvijic, Melissa Yarde, Bruce D. Car, Kenneth B. Margulies, and Diana A. Chirinos
- Subjects
Cardiac function curve ,medicine.medical_specialty ,business.industry ,Atrial fibrillation ,030204 cardiovascular system & hematology ,medicine.disease ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,Heart failure ,Internal medicine ,medicine ,Spironolactone ,Clinical endpoint ,Left atrial enlargement ,Cardiology ,Arterial stiffness ,030212 general & internal medicine ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,business - Abstract
Objectives This study sought to assess if clinical phenogroups differ in comprehensive biomarker profiles, cardiac and arterial structure/function, and responses to spironolactone therapy. Background Previous studies identified distinct subgroups (phenogroups) of patients with heart failure with preserved ejection fraction (HFpEF). Methods Among TOPCAT (Treatment of Preserved Cardiac Function Heart Failure with an Aldosterone Antagonist Trial) participants, we performed latent-class analysis to identify HFpEF phenogroups based on standard clinical features and assessed differences in multiple biomarkers measured from frozen plasma; cardiac and arterial structure/function measured with echocardiography and arterial tonometry; prognosis; and response to spironolactone. Results Three HFpEF phenogroups were identified. Phenogroup 1 (n = 1,214) exhibited younger age, higher prevalence of smoking, preserved functional class, and the least evidence of left ventricular (LV) hypertrophy and arterial stiffness. Phenogroup 2 (n = 1,329) was older, with normotrophic concentric LV remodeling, atrial fibrillation, left atrial enlargement, large-artery stiffening, and biomarkers of innate immunity and vascular calcification. Phenogroup 3 (n = 899) demonstrated more functional impairment, obesity, diabetes, chronic kidney disease, concentric LV hypertrophy, high renin, and biomarkers of tumor necrosis factor-alpha–mediated inflammation, liver fibrosis, and tissue remodeling. Compared with phenogroup 1, phenogroup 3 exhibited the highest risk of the primary endpoint of cardiovascular death, heart failure hospitalization, or aborted cardiac arrest (hazard ratio [HR]: 3.44; 95% confidence interval [CI]: 2.79 to 4.24); phenogroups 2 and 3 demonstrated similar all-cause mortality (phenotype 2 HR: 2.36; 95% CI: 1.89 to 2.95; phenotype 3 HR: 2.26, 95% CI: 1.77 to 2.87). Spironolactone randomized therapy was associated with a more pronounced reduction in the risk of the primary endpoint in phenogroup 3 (HR: 0.75; 95% CI: 0.59 to 0.95; p for interaction = 0.016). Results were similar after excluding participants from Eastern Europe. Conclusions We identified important differences in circulating biomarkers, cardiac/arterial characteristics, prognosis, and response to spironolactone across clinical HFpEF phenogroups. These findings suggest distinct underlying mechanisms across clinically identifiable phenogroups of HFpEF that may benefit from different targeted interventions.
- Published
- 2020
3. Multiple Plasma Biomarkers for Risk Stratification in Patients With Heart Failure and Preserved Ejection Fraction
- Author
-
Michael Basso, David A. Gordon, Julio A. Chirinos, Thomas P. Cappola, Zhaoqing Wang, Mary Ellen Cvijic, Dietmar A. Seiffert, Payman Zamani, Priyanka Bhattacharya, Melissa Yarde, Bruce D. Car, Stuart B. Prenner, Kenneth B. Margulies, Jason H. Moore, Zhuyin Li, Thomas E. Spires, Alena Orlenko, Anupam Kumar, and Lei Zhao
- Subjects
Oncology ,medicine.medical_specialty ,Ejection fraction ,business.industry ,medicine.drug_class ,Hazard ratio ,030204 cardiovascular system & hematology ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Osteoprotegerin ,Fibrosis ,Internal medicine ,Heart failure ,Natriuretic peptide ,Medicine ,Biomarker (medicine) ,030212 general & internal medicine ,Cardiology and Cardiovascular Medicine ,business ,Heart failure with preserved ejection fraction - Abstract
Background Better risk stratification strategies are needed to enhance clinical care and trial design in heart failure with preserved ejection fraction (HFpEF). Objectives The purpose of this study was to assess the value of a targeted plasma multi-marker approach to enhance our phenotypic characterization and risk prediction in HFpEF. Methods In this study, the authors measured 49 plasma biomarkers from TOPCAT (Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist) trial participants (n = 379) using a Multiplex assay. The relationship between biomarkers and the risk of all-cause death or heart failure-related hospital admission (DHFA) was assessed. A tree-based pipeline optimizer platform was used to generate a multimarker predictive model for DHFA. We validated the model in an independent cohort of HFpEF patients enrolled in the PHFS (Penn Heart Failure Study) (n = 156). Results Two large, tightly related dominant biomarker clusters were found, which included biomarkers of fibrosis/tissue remodeling, inflammation, renal injury/dysfunction, and liver fibrosis. Other clusters were composed of neurohormonal regulators of mineral metabolism, intermediary metabolism, and biomarkers of myocardial injury. Multiple biomarkers predicted incident DHFA, including 2 biomarkers related to mineral metabolism/calcification (fibroblast growth factor-23 and OPG [osteoprotegerin]), 3 inflammatory biomarkers (tumor necrosis factor-alpha, sTNFRI [soluble tumor necrosis factor-receptor I], and interleukin-6), YKL-40 (related to liver injury and inflammation), 2 biomarkers related to intermediary metabolism and adipocyte biology (fatty acid binding protein-4 and growth differentiation factor-15), angiopoietin-2 (related to angiogenesis), matrix metalloproteinase-7 (related to extracellular matrix turnover), ST-2, and N-terminal pro–B-type natriuretic peptide. A machine-learning–derived model using a combination of biomarkers was strongly predictive of the risk of DHFA (standardized hazard ratio: 2.85; 95% confidence interval: 2.03 to 4.02; p Conclusions Various novel circulating biomarkers in key pathophysiological domains are predictive of outcomes in HFpEF, and a multimarker approach coupled with machine-learning represents a promising strategy for enhancing risk stratification in HFpEF.
- Published
- 2020
4. Circulating Neprilysin in Patients With Heart Failure and Preserved Ejection Fraction
- Author
-
John C. Burnett, Seethalakshmi R. Iyer, Naveen L. Pereira, Christopher G. Scott, Melissa A. Lyle, G. Michael Felker, Margaret M. Redfield, Adrian F. Hernandez, Yogesh N.V. Reddy, and Thomas P. Cappola
- Subjects
Male ,medicine.medical_specialty ,Heart Ventricles ,Diastole ,Tetrazoles ,030204 cardiovascular system & hematology ,Asymptomatic ,Ventricular Function, Left ,Article ,Sacubitril ,Angiotensin Receptor Antagonists ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Aged ,Heart Failure ,Ejection fraction ,business.industry ,Aminobutyrates ,Biphenyl Compounds ,Stroke Volume ,Middle Aged ,medicine.disease ,Drug Combinations ,Valsartan ,Echocardiography ,Case-Control Studies ,Heart failure ,Cardiology ,Biomarker (medicine) ,Female ,Neprilysin ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Heart failure with preserved ejection fraction ,Biomarkers ,medicine.drug - Abstract
In heart failure with reduced ejection fraction (HFrEF), elevated soluble neprilysin (sNEP) levels are associated with an increased risk of cardiovascular death, and its inhibition with sacubitril/valsartan has improved survival.This study sought to determine the relevance of sNEP as a biomarker in heart failure with preserved ejection fraction (HFpEF) and to compare circulating sNEP levels in patients with HFpEF with normal controls.A case-control study was performed in 242 symptomatic patients with HFpEF previously enrolled in the Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in Heart Failure with Preserved Ejection Fraction (RELAX) and Nitrates's Effect on Activity Tolerance in Heart Failure With Preserved Ejection (NEAT-HFpEF) clinical trials and 891 asymptomatic subjects without HF or diastolic dysfunction (confirmed by NT-proBNP levels 200 pg/ml and echocardiography) who were enrolled in the Prevalence of Asymptomatic Left Ventricular Dysfunction study. sNEP was measured using a sandwich enzyme-linked immunosorbent assay (ELISA) in all subjects.Overall, sNEP levels were lower in HFpEF compared with controls (3.5 ng/ml; confidence interval [CI]: 2.5 to 4.8 vs. 8.5 ng/ml; CI: 7.2 to 10.0; p 0.001). After adjusting for age, gender, body mass index (BMI), and smoking history, mean sNEP levels were also lower in HFpEF compared with controls (4.0 ng/ml [CI: 2.7 to 5.4] vs. 8.2 ng/ml [CI: 6.8 to 9.7]; p = 0.002). The cohorts were propensity matched based on age, BMI, diabetes, hypertension, smoking history, and renal function, and sNEP levels remained lower in HFpEF compared with controls (median 2.4 ng/ml [interquartile range: 0.6 to 27.7] vs. 4.9 ng/ml [interquartile range: 1.2 to 42.2]; p = 0.02).Patients with HFpEF on average have significantly lower circulating sNEP levels compared with controls. These findings challenge our current understanding of the complex biology of circulating sNEP in HFpEF.
- Published
- 2020
5. Abstract 100: Integrated Cardiac Metabolism In End-Stage Human Heart Failure
- Author
-
Ben Prosser, Danielle Murashige, Jeff Brandimarto, Zolt Arany, Kenneth B. Margulies, Thomas P. Cappola, Michael Morley, Emily Flam, Ken Bedi, Yifan Yang, Cholsoon Jang, and Josh Rabinowitz
- Subjects
medicine.medical_specialty ,Physiology ,business.industry ,Internal medicine ,medicine ,Cardiology ,Cardiac metabolism ,Human heart ,Stage (cooking) ,Cardiology and Cardiovascular Medicine ,business - Abstract
Heart failure affects millions of people worldwide with mortality near 50% within five years. This disease is characterized by widespread cardiac and systemic metabolic changes, but a comprehensive evaluation of metabolism in failing human hearts is lacking. Here, we provide a comprehensive depiction of cardiac and systemic metabolic changes in 89 explanted failing and non-failing human hearts through integration of plasma and cardiac tissue metabolomics, genome-wide RNAseq, and proteomic data. The data confirm a profound bioenergetic defect in end-stage human heart failure and demonstrate extensive changes in metabolic homeostasis. The data indicate a substantial defect in fatty acid (FA) use in failing hearts, in particular unsaturated FAs. Reduction of FAs and acyl-carnitines in failing tissue in contrast to concomitant elevations in plasma suggest a defect in import of FAs into the cell, rather than a defect in FA oxidation. Intermediates of glycolysis, the pentose phosphate pathway, and glycogen synthesis are all similarly reduced, as is expression of GLUT1, indicating diminished glucose uptake. However, there was no significant change in tissue pyruvate content, suggesting an increase in lactate utilization. The data suggest increased flux of pyruvate into mitochondria, likely promoting pyruvate oxidation but not pyruvate carboxylation. Blunted anabolic pyruvate flux, in turn, likely leads to insufficient TCA cycle intermediates. Ketone levels were increased in both failing tissue and plasma, as previously reported. The phospholipid content of failing human hearts is greatly increased in both failing tissue and plasma. Nucleotide synthesis pathways also appear to be reprogrammed, with a notable decrease in adenosine metabolism, specifically. Together, these data indicate widespread change in the local cardiac and greater systemic metabolic landscape in severe human heart failure.
- Published
- 2021
6. The genomics of heart failure: design and rationale of the HERMES consortium
- Author
-
Svati H. Shah, Olle Melander, Neneh Sallah, Quinn S. Wells, Jerome I. Rotter, Faye Zhao, Charlotte Andersson, Guðmundur Thorgeirsson, Ghazaleh Fatemifar, Alex S. F. Doney, Michael E. Dunn, David E. Lanfear, Ian Ford, Eric Boersma, Sonia Shah, Christopher Newton-Cheh, Douglas L. Mann, Niek Verweij, Carolina Roselli, Laura M. Yerges-Armstrong, Jian Yang, Christian Torp-Pedersen, Veikko Salomaa, Mary L. Biggs, Alaa Shalaby, Christoph Nowak, Stefan Gross, Patrick T. Ellinor, Mari Liis Tammesoo, Diane T. Smelser, Peter M. Visscher, Hans L. Hillege, Ruth C. Lovering, Honghuang Lin, Colin N. A. Palmer, Louis Philippe Lemieux Perreault, Jeffrey Brandimarto, Uwe Völker, Perttu Salo, Andrea Koekemoer, Rebecca Gutmann, Åsa K. Hedman, Nilesh J. Samani, Heming Xing, Faiez Zannad, Jaison Jacob, Harry Hemingway, Michael R. Brown, Franco Giulianini, Anubha Mahajan, Xing Chen, Alexander Niessner, Peter Almgren, Daniel I. Swerdlow, Gunnar Engström, Lars Lind, Tõnu Esko, Tomasz Czuba, Anna Helgadottir, Harvey D. White, David J. Stott, Johan Ärnlöv, Lars Køber, Chim C. Lang, Krishna G. Aragam, Kent D. Taylor, Anders Mälarstig, Frederick K. Kamanu, Kenneth B. Margulies, Michelle L. O'Donoghue, Andrew D. Morris, Sahar Ghasemi, J. Wouter Jukema, Jessica van Setten, Abbas Dehghan, Guillaume Paré, Luca A. Lotta, Giorgio E. M. Melloni, Albert Henry, Bruce M. Psaty, Paul M. Ridker, David J. Carey, Marie-Pierre Dubé, John S. Gottdiener, Xiaosong Wang, Per H. Svensson, Xu Chen, Patrik K. E. Magnusson, Claudia Langenberg, Alexander Teumer, Vilmantas Giedraitis, Simon de Denus, Michael W. Nagle, Marcus Dörr, Thomas P. Cappola, André G. Uitterlinden, Michael Morley, Eliana Portilla-Fernandez, J. Gustav Smith, Abirami Veluchamy, Peter Weeke, Ify R. Mordi, Unnur Thorsteinsdottir, Naveed Sattar, Folkert W. Asselbergs, Daniel I. Chasman, Daníel F. Guðbjartsson, Jonathan H. Chung, Marcus E. Kleber, Raul Weiss, Christopher P. Nelson, Spiros Denaxas, Bing Yu, Simon P. R. Romaine, Nicholas A Marston, Anjali T. Owens, Cecilia M. Lindgren, John J.V. McMurray, Joshua D. Backman, Michael V. Holmes, Stella Trompet, Hilma Holm, Kerri L. Wiggins, Jian'an Luan, Stephan B. Felix, Yifan Yang, Jemma B. Wilk, Maryam Kavousi, Markus Perola, Christian T. Ruff, Jean-Claude Tardif, G Sveinbjörnsson, Samuel C. Dudley, Nicholas J. Wareham, Teemu J. Niiranen, Andrew P. Morris, Danny Tuckwell, Maris Teder-Laving, R. Thomas Lumbers, James P. Cook, Géraldine Asselin, William A. Chutkow, Winfried März, Steven A. Lubitz, John G.F. Cleland, Bill Kraus, Ramachandran S. Vasan, Christopher M. Haggerty, Olympe Chazara, Chris Finan, Heather L. Bloom, Hans-Peter Brunner-La Rocca, Francoise Fougerousse, Kenneth Rice, Craig L. Hyde, Graciela E. Delgado, Mark Chaffin, Marc S. Sabatine, Alanna C. Morrison, Kay-Tee Khaw, Kari Stefansson, Felix Vaura, Barry London, Isabella Kardys, Aroon D. Hingorani, Hongsheng Gui, Steen Stender, René Fouodjio, Mohsen Ghanbari, Pim van der Harst, Nicholas L. Smith, Karoline Kuchenbaecker, Adriaan A. Voors, Benoit Tyl, University College of London [London] (UCL), University College London Hospitals (UCLH), Boston University School of Medicine (BUSM), Boston University [Boston] (BU), Lund University [Lund], Pfizer, Karolinska Institutet [Stockholm], Broad Institute of MIT and Harvard (BROAD INSTITUTE), Harvard Medical School [Boston] (HMS)-Massachusetts Institute of Technology (MIT)-Massachusetts General Hospital [Boston], University of Groningen [Groningen], University of Oxford [Oxford], Dalarna University, Massachusetts General Hospital [Boston], Montreal Heart Institute - Institut de Cardiologie de Montréal, Regeneron Genetics Center, 777 Old Saw Mill River Road, Tarrytown, University of Washington [Seattle], Emory University [Atlanta, GA], Erasmus University Medical Center [Rotterdam] (Erasmus MC), University of Pennsylvania [Philadelphia], The University of Texas Health Science Center at Houston (UTHealth), Maastricht University Medical Centre (MUMC), Maastricht University [Maastricht], Department of Molecular and Functional Genomics, Geisinger, Danville, PA, Brigham & Women’s Hospital [Boston] (BWH), Harvard Medical School [Boston] (HMS), AstraZeneca, Novartis Institutes for BioMedical Research (NIBR), University of Glasgow, University of Liverpool, Université de Montréal (UdeM), Imperial College London, University of Heidelberg, Medical Faculty, University of Dundee, Universität Greifswald - University of Greifswald, University of Minnesota Medical School, University of Minnesota System, German Center for Cardiovascular Research (DZHK), Berlin Institute of Health (BIH), Institut de Recherches Internationales Servier [Suresnes] (IRIS), Uppsala University, University of Maryland School of Medicine, University of Maryland System, University of Iceland [Reykjavik], deCODE genetics [Reykjavik], Henry Ford Hospital, Carver College of Medicine, University of Iowa, Geisinger Autism & Developmental Medicine Institute [Danville, PA, USA] (ADMI), ICIN - Netherlands Heart Institute, Leiden University Medical Center (LUMC), Netherlands Heart Institute, Partenaires INRAE, University of Cambridge [UK] (CAM), Rigshospitalet [Copenhagen], Copenhagen University Hospital, University of Leicester, Duke University [Durham], University of Iowa [Iowa City], Genentech, Inc., Genentech, Inc. [San Francisco], Washington University School of Medicine in St. Louis, Washington University in Saint Louis (WUSTL), Skane University Hospital [Malmo], University of Edinburgh, Medizinische Universität Wien = Medical University of Vienna, University of Turku, National Institute for Health and Welfare [Helsinki], McMaster University [Hamilton, Ontario], Kaiser Permanente Washington Health Research Institute [Seattle] (KPWHRI), Harbor UCLA Medical Center [Torrance, Ca.], University Medical Center [Utrecht], University of Pittsburgh Medical Center [Pittsburgh, PA, États-Unis] (UPMC), University of Tartu, Aalborg University [Denmark] (AAU), Leiden University, University of Queensland [Brisbane], Ohio State University [Columbus] (OSU), Auckland City Hospital, GlaxoSmithKline, Glaxo Smith Kline, University of Texas Health Science Center, Cardiovascular and Renal Clinical Trialists [Vandoeuvre-les-Nancy] (INI-CRCT), Institut Lorrain du Coeur et des Vaisseaux Louis Mathieu [Nancy], French-Clinical Research Infrastructure Network - F-CRIN [Paris] (Cardiovascular & Renal Clinical Trialists - CRCT ), Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Université de Lorraine (UL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Duke University Medical Center, Regeneron Pharmaceuticals [Tarrytown], Vanderbilt University [Nashville], European Project, Langenberg, Claudia [0000-0002-5017-7344], Luan, Jian'an [0000-0003-3137-6337], Wareham, Nicholas [0000-0003-1422-2993], Apollo - University of Cambridge Repository, Cardiovascular Centre (CVC), Life Course Epidemiology (LCE), Groningen Kidney Center (GKC), Cardiology, University of Oxford, University of Pennsylvania, Universiteit Leiden, Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Cardiologie, MUMC+: MA Med Staf Spec Cardiologie (9), RS: Carim - H02 Cardiomyopathy, Epidemiology, and Internal Medicine
- Subjects
Male ,Study Designs ,Cardiomyopathy ,Disease ,030204 cardiovascular system & hematology ,0302 clinical medicine ,AFRICAN ANCESTRY ,Epidemiology ,80 and over ,WIDE ASSOCIATION ,EPIDEMIOLOGY ,Cardiac and Cardiovascular Systems ,AGING RESEARCH ,RISK ,Aged, 80 and over ,0303 health sciences ,Kardiologi ,Genomics ,Middle Aged ,Prognosis ,3. Good health ,Female ,Cardiology and Cardiovascular Medicine ,medicine.medical_specialty ,Heart failure ,CLASSIFICATION ,Heart Failure/genetics ,03 medical and health sciences ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Internal medicine ,Genetic model ,medicine ,Genetics ,Diseases of the circulatory (Cardiovascular) system ,Humans ,Allele frequency ,Genotyping ,METAANALYSIS ,030304 developmental biology ,Aged ,Association studies ,Study Design ,business.industry ,Odds ratio ,ADULTS ,COHORTS ,medicine.disease ,RC666-701 ,REPLICATION ,business ,Biomarkers ,Genome-Wide Association Study - Abstract
Aims The HERMES (HEart failure Molecular Epidemiology for Therapeutic targets) consortium aims to identify the genomic and molecular basis of heart failure.Methods and results The consortium currently includes 51 studies from 11 countries, including 68 157 heart failure cases and 949 888 controls, with data on heart failure events and prognosis. All studies collected biological samples and performed genome-wide genotyping of common genetic variants. The enrolment of subjects into participating studies ranged from 1948 to the present day, and the median follow-up following heart failure diagnosis ranged from 2 to 116 months. Forty-nine of 51 individual studies enrolled participants of both sexes; in these studies, participants with heart failure were predominantly male (34-90%). The mean age at diagnosis or ascertainment across all studies ranged from 54 to 84 years. Based on the aggregate sample, we estimated 80% power to genetic variant associations with risk of heart failure with an odds ratio of >1.10 for common variants (allele frequency > 0.05) and >1.20 for low-frequency variants (allele frequency 0.01-0.05) at P < 5 x 10(-8) under an additive genetic model.Conclusions HERMES is a global collaboration aiming to (i) identify the genetic determinants of heart failure; (ii) generate insights into the causal pathways leading to heart failure and enable genetic approaches to target prioritization; and (iii) develop genomic tools for disease stratification and risk prediction.
- Published
- 2021
7. Genome-wide association analysis in dilated cardiomyopathy reveals two new players in systolic heart failure on chromosomes 3p25.1 and 22q11.23
- Author
-
Magdalena Harakalova, Benjamin Meder, Philippe Charron, Manuel Gómez-Bueno, Jorg J. A. Calis, François Cambien, David-Alexandre Trégouët, Maurizia Grasso, Steven McGinn, Uwe Völker, Thomas Meitinger, Stefan Weiss, L. Duboscq-Bidot, Richard Dorent, Vera Regitz-Zagrosek, Folkert W. Asselbergs, Hélène Blanché, Olivier Dubourg, Patrick Lacolley, Pierre Boutouyrie, Delphine Bacq-Daian, Vincent Fontaine, Volker Ruppert, Marine Germain, K Lehnert, Jean-Noël Trochu, Stuart A. Cook, Angélique Curjol, Brendan J. Keating, Ibticem Raji, Anne Boland, J. Erdmann, Michael Morley, Jean-François Aupetit, Paloma Remior, Luigi Tavazzi, Gérard Roizès, Michal Mokry, Konstantin Strauch, Richard Isnard, Jean-Philippe Empana, Robert Olaso, Kenneth B. Marguiles, Zofia T. Bilińska, Stephan B. Felix, Marcus Dörr, Thomas P. Cappola, Stefan Blankenberg, Jan Haas, Céline Besse, Jean-François Deleuze, Christine E. Seidman, Christian Hengstenberg, Jessica van Setten, Hakon Hakonarson, Sanjay K Prasad, Daiane Hemerich, Pascal de Groote, Thomas Wichter, Alain van Mil, Michel Komajda, Renee Maas, Carole Proust, Declan P. O'Regan, Xavier Jouven, Ganapathi Varma Saripella, Georgios Kararigas, Eloisa Arbustini, Jin Li, Klaus Stark, Laurent Fauchier, Flavie Ader, Melanie Waldenberger, Martina Müller-Nurasyid, Eric Villard, Sophie Garnier, Cardiology, Imperial College Healthcare NHS Trust- BRC Funding, British Heart Foundation, Unité de Recherche sur les Maladies Cardiovasculaires, du Métabolisme et de la Nutrition = Institute of cardiometabolism and nutrition (ICAN), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU), Service de Cardiologie [CHU Pitié-Salpêtrière], CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), CHU Tenon [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-CHU Pitié-Salpêtrière [AP-HP], Centre hospitalier Saint-Joseph [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Bordeaux population health (BPH), Université de Bordeaux (UB)-Institut de Santé Publique, d'Épidémiologie et de Développement (ISPED)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de Génétique Cytogénétique et Embryologie [CHU Pitié-Salpêtrière], Assistance publique-Hôpitaux de Paris, Fondation Leducq, Société Française de Cardiologie, Deutsche Forschungsgemeinschaft, Helmholtz Zentrum München, Université de Bordeaux, Medical Research Council, ANR-10-LABX-0013,GENMED,Medical Genomics(2010), Unité de Recherche sur les Maladies Cardiovasculaires, du Métabolisme et de la Nutrition = Research Unit on Cardiovascular and Metabolic Diseases (ICAN), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Institut de Cardiométabolisme et Nutrition = Institute of Cardiometabolism and Nutrition [CHU Pitié Salpêtrière] (IHU ICAN), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), University Medical Center [Utrecht], Universität Greifswald - University of Greifswald, German Center for Cardiovascular Research (DZHK), Berlin Institute of Health (BIH), Charité - UniversitätsMedizin = Charité - University Hospital [Berlin], University of Pennsylvania, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), National Heart Centre Singapore (NHCS), Children’s Hospital of Philadelphia (CHOP ), University of Regensburg, Royal Brompton Hospital, Imperial College London, Istituti Clinici Scientifici Maugeri [Pavia] (IRCCS Pavia - ICS Maugeri), Helmholtz Zentrum München = German Research Center for Environmental Health, Ludwig-Maximilians-Universität München (LMU), Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM), University Medical Center of the Johannes Gutenberg-University Mainz, Perelman School of Medicine, Harvard Medical School [Boston] (HMS), University of Iceland [Reykjavik], Heidelberg University, Stanford University Medical School, Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Défaillance Cardiovasculaire Aiguë et Chronique (DCAC), Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), Universität zu Lübeck = University of Lübeck [Lübeck], Universitätklinikum Gießen und Marburg GmbH, Maria Cecilia Hospital [Cotignola], Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Institut de génétique humaine (IGH), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Hôpital cardiologique, Université de Lille, Droit et Santé-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), CHU Trousseau [Tours], Centre Hospitalier Régional Universitaire de Tours (CHRU Tours), Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Centre hospitalier Saint Joseph - Saint Luc [Lyon], National Institute of Cardiology [Varsovie, Pologne], University of Medicine Greifswald, Centre de Référence Maladies Cardiaques Héréditaires, Centre National de Recherche en Génomique Humaine (CNRGH), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Laboratory of Excellence GENMED [Paris] (Medical Genomics), Hospital Universitario Puerta de Hierro-Majadahonda [Madrid, Spain], Swedish University of Agricultural Sciences (SLU), Centre d'Etude du Polymorphisme Humain (CEPH), Institut Universitaire d'Hématologie (IUH), Université Paris Diderot - Paris 7 (UPD7)-Université Paris Diderot - Paris 7 (UPD7)-Fondation Jean Dausset-Université Paris Cité (UPCité), Faculté de Pharmacie de Paris - Université Paris Descartes (UPD5 Pharmacie), Université Paris Descartes - Paris 5 (UPD5), Groupe Hospitalier Paris Saint Joseph, Fondation Jean Dausset - Centre d’Etudes du Polymorphisme Humain [Paris] (CEPH), and University College of London [London] (UCL)
- Subjects
Cardiac & Cardiovascular Systems ,Cardiomyopathy, Dilated/genetics ,[SDV]Life Sciences [q-bio] ,Signal Transducing/genetics ,Dilated cardiomyopathy ,Genome-wide association study ,Adaptor Proteins, Signal Transducing/genetics ,030204 cardiovascular system & hematology ,TAURINE ,0302 clinical medicine ,GWAS ,Medicine ,POSITION STATEMENT ,1102 Cardiorespiratory Medicine and Haematology ,Genetics ,0303 health sciences ,education.field_of_study ,Genetic Predisposition to Disease/genetics ,Adaptor Proteins ,4C-sequencing ,Polymorphism, Single Nucleotide/genetics ,Genetic risk score ,Cardiology and Cardiovascular Medicine ,Life Sciences & Biomedicine ,Single Nucleotide/genetics ,Cardiomyopathy, Dilated ,Cardiomyopathy ,Population ,Locus (genetics) ,Single-nucleotide polymorphism ,Polymorphism, Single Nucleotide ,Chromosomes ,03 medical and health sciences ,Systolic/genetics ,Heart Failure, Systolic/genetics ,SNP ,Animals ,Humans ,Genetic Predisposition to Disease ,Allele ,Polymorphism ,education ,Imputation ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Heart Failure ,Science & Technology ,business.industry ,WORKING GROUP ,1103 Clinical Sciences ,medicine.disease ,Genetic architecture ,Cardiovascular System & Hematology ,Dilated/genetics ,Cardiovascular System & Cardiology ,[SDV.SPEE]Life Sciences [q-bio]/Santé publique et épidémiologie ,business ,Apoptosis Regulatory Proteins ,Heart Failure, Systolic ,Genome-Wide Association Study - Abstract
Aims Our objective was to better understand the genetic bases of dilated cardiomyopathy (DCM), a leading cause of systolic heart failure. Methods and results We conducted the largest genome-wide association study performed so far in DCM, with 2719 cases and 4440 controls in the discovery population. We identified and replicated two new DCM-associated loci on chromosome 3p25.1 [lead single-nucleotide polymorphism (SNP) rs62232870, P = 8.7 × 10−11 and 7.7 × 10−4 in the discovery and replication steps, respectively] and chromosome 22q11.23 (lead SNP rs7284877, P = 3.3 × 10−8 and 1.4 × 10−3 in the discovery and replication steps, respectively), while confirming two previously identified DCM loci on chromosomes 10 and 1, BAG3 and HSPB7. A genetic risk score constructed from the number of risk alleles at these four DCM loci revealed a 3-fold increased risk of DCM for individuals with 8 risk alleles compared to individuals with 5 risk alleles (median of the referral population). In silico annotation and functional 4C-sequencing analyses on iPSC-derived cardiomyocytes identify SLC6A6 as the most likely DCM gene at the 3p25.1 locus. This gene encodes a taurine transporter whose involvement in myocardial dysfunction and DCM is supported by numerous observations in humans and animals. At the 22q11.23 locus, in silico and data mining annotations, and to a lesser extent functional analysis, strongly suggest SMARCB1 as the candidate culprit gene. Conclusion This study provides a better understanding of the genetic architecture of DCM and sheds light on novel biological pathways underlying heart failure.
- Published
- 2021
8. Genetic and Phenotypic Landscape of Peripartum Cardiomyopathy
- Author
-
Denise Hilfiker-Kleiner, Zolt Arany, Quentin McAfee, Kenneth B. Margulies, Rami Alharethi, Eileen Hsich, Lisa D. Levine, Sorel Goland, Christine E. Seidman, Peter Damm, Jonathan G. Seidman, Sarosh Rana, Daniel Jacoby, Thomas P. Cappola, Chizuko Kamiya, Julie B. Damp, Anne S Ersbøll, Jeff Brandimarto, Steven R. DePalma, Rahul R. Goli, Richard Sheppard, Imac, Uri Elkayam, Ipac Investigators, Valerie Riis, John P. Boehmer, Finn Gustafsson, George A. Macones, Dennis M. McNamara, Jeffrey D. Alexis, Alireza Haghighi, Daniel P. Judge, and Jian Li
- Subjects
Adult ,0303 health sciences ,medicine.medical_specialty ,Peripartum cardiomyopathy ,Obstetrics ,business.industry ,030204 cardiovascular system & hematology ,medicine.disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Phenotype ,Pregnancy ,Physiology (medical) ,medicine ,Peripartum Period ,Humans ,Female ,Cardiology and Cardiovascular Medicine ,business ,Cardiomyopathies ,030304 developmental biology ,Retrospective Studies - Abstract
Background: Peripartum cardiomyopathy (PPCM) occurs in ≈1:2000 deliveries in the United States and worldwide. The genetic underpinnings of PPCM remain poorly defined. Approximately 10% of women with PPCM harbor truncating variants in TTN (TTNtvs). Whether mutations in other genes can predispose to PPCM is not known. It is also not known if the presence of TTNtvs predicts clinical presentation or outcomes. Nor is it known if the prevalence of TTNtvs differs in women with PPCM and preeclampsia, the strongest risk factor for PPCM. Methods: Women with PPCM were retrospectively identified from several US and international academic centers, and clinical information and DNA samples were acquired. Next-generation sequencing was performed on 67 genes, including TTN , and evaluated for burden of truncating and missense variants. The impact of TTNtvs on the severity of clinical presentation, and on clinical outcomes, was evaluated. Results: Four hundred sixty-nine women met inclusion criteria. Of the women with PPCM, 10.4% bore TTNtvs (odds ratio=9.4 compared with 1.2% in the reference population; Bonferroni-corrected P [ P *]=1.2×10 –46 ). We additionally identified overrepresentation of truncating variants in FLNC (odds ratio=24.8, P *=7.0×10 –8 ), DSP (odds ratio=14.9, P *=1.0×10 –8 ), and BAG3 (odds ratio=53.1, P *=0.02), genes not previously associated with PPCM. This profile is highly similar to that found in nonischemic dilated cardiomyopathy. Women with TTNtvs had lower left ventricular ejection fraction on presentation than did women without TTNtvs (23.5% versus 29%, P =2.5×10 –4 ), but did not differ significantly in timing of presentation after delivery, in prevalence of preeclampsia, or in rates of clinical recovery. Conclusions: This study provides the first extensive genetic and phenotypic landscape of PPCM and demonstrates that predisposition to heart failure is an important risk factor for PPCM. The work reveals a degree of genetic similarity between PPCM and dilated cardiomyopathy, suggesting that gene-specific therapeutic approaches being developed for dilated cardiomyopathy may also apply to PPCM, and that approaches to genetic testing in PPCM should mirror those taken in dilated cardiomyopathy. Last, the clarification of genotype/phenotype associations has important implications for genetic counseling.
- Published
- 2021
9. Genomic Context Differs Between Human Dilated Cardiomyopathy and Hypertrophic Cardiomyopathy
- Author
-
Lorenzo L. Pesce, Samuel Kearns, Jane E. Wilcox, Wenyu Pan, Lisa Dellefave-Castillo, Avery C. Robinson, Thomas P. Cappola, Gerald W. Dorn, Megan J. Puckelwartz, Sharlene M. Day, Euan A. Ashley, Anthony Gacita, Zachary J. Schoppen, Matthew T. Wheeler, Gene Kim, Elizabeth M. McNally, Allen S. Anderson, and Tess D. Pottinger
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,medicine.medical_specialty ,Genotype ,Cardiomyopathy ,Heart Ventricles ,Context (language use) ,030204 cardiovascular system & hematology ,Polymorphism, Single Nucleotide ,Ventricular Function, Left ,Variable Expression ,03 medical and health sciences ,0302 clinical medicine ,modifier genes ,Internal medicine ,variable expressivity ,medicine ,Genetics ,Humans ,cardiovascular diseases ,Allele frequency ,030304 developmental biology ,Original Research ,Heart Failure ,0303 health sciences ,Ejection fraction ,business.industry ,Hypertrophic cardiomyopathy ,Dilated cardiomyopathy ,Stroke Volume ,Genomics ,Hypertrophy ,Cardiomyopathy, Hypertrophic ,Middle Aged ,medicine.disease ,hypertrophic cardiomyopathy ,Penetrance ,dilated cardiomyopathy ,Echocardiography ,variant burden ,Cardiology ,cardiovascular system ,Female ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background Inherited cardiomyopathies display variable penetrance and expression, and a component of phenotypic variation is genetically determined. To evaluate the genetic contribution to this variable expression, we compared protein coding variation in the genomes of those with hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Methods and Results Nonsynonymous single‐nucleotide variants (nsSNVs) were ascertained using whole genome sequencing from familial cases of HCM (n=56) or DCM (n=70) and correlated with echocardiographic information. Focusing on nsSNVs in 102 genes linked to inherited cardiomyopathies, we correlated the number of nsSNVs per person with left ventricular measurements. Principal component analysis and generalized linear models were applied to identify the probability of cardiomyopathy type as it related to the number of nsSNVs in cardiomyopathy genes. The probability of having DCM significantly increased as the number of cardiomyopathy gene nsSNVs per person increased. The increase in nsSNVs in cardiomyopathy genes significantly associated with reduced left ventricular ejection fraction and increased left ventricular diameter for individuals carrying a DCM diagnosis, but not for those with HCM. Resampling was used to identify genes with aberrant cumulative allele frequencies, identifying potential modifier genes for cardiomyopathy. Conclusions Participants with DCM had more nsSNVs per person in cardiomyopathy genes than participants with HCM. The nsSNV burden in cardiomyopathy genes did not correlate with the probability or manifestation of left ventricular measures in HCM. These findings support the concept that increased variation in cardiomyopathy genes creates a genetic background that predisposes to DCM and increased disease severity.
- Published
- 2021
10. Chronobiology of Natriuretic Peptides and Blood Pressure in Lean and Obese Individuals
- Author
-
Karen L. Gamble, Vibhu Parcha, Orlando M. Gutiérrez, Nirav Patel, Thomas J. Wang, Thomas P. Cappola, Peng Li, Kenneth B. Margulies, Pankaj Arora, Garima Arora, and Kiran Musunuru
- Subjects
Adult ,Male ,medicine.medical_specialty ,Ambulatory blood pressure ,Adolescent ,medicine.drug_class ,Blood Pressure ,030204 cardiovascular system & hematology ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Interquartile range ,Internal medicine ,medicine ,Natriuretic peptide ,Humans ,030212 general & internal medicine ,Circadian rhythm ,Obesity ,Natriuretic Peptides ,Chronobiology Phenomena ,business.industry ,Light intensity ,Blood pressure ,Endocrinology ,Female ,Cardiology and Cardiovascular Medicine ,business ,Body mass index ,Blood drawing - Abstract
Background Diurnal variation of natriuretic peptide (NP) levels and its relationship with 24-h blood pressure (BP) rhythm has not been established. Obese individuals have a relative NP deficiency and disturbed BP rhythmicity. Objectives This clinical trial evaluated the diurnal rhythmicity of NPs (B-type natriuretic peptide [BNP], mid-regional pro-atrial natriuretic peptide [MR-proANP], N-terminal pro–B-type natriuretic peptide [NT-proBNP]) and the relationship of NP rhythm with 24-h BP rhythm in healthy lean and obese individuals. Methods On the background of a standardized diet, healthy, normotensive, lean (body mass index 18.5 to 25 kg/m2) and obese (body mass index 30 to 45 kg/m2) individuals, age 18 to 40 years, underwent 24-h inpatient protocol involving ambulatory BP monitoring starting 24 h prior to the visit, controlled light intensity, and repeated blood draws for assessment of analytes. Cosinor analysis of normalized NP levels (normalized to 24-h mean value) was conducted to assess the diurnal NP rhythm and its relationship with systolic BP. Results Among 52 participants screened, 40 participants (18 lean, 22 obese; 50% women; 65% Black) completed the study. The median range spread (percentage difference between the minimum and maximum values) over 24 h for MR-proANP, BNP, and NT-proBNP levels was 72.0% (interquartile range [IQR]: 50.9% to 119.6%), 75.5% (IQR: 50.7% to 106.8%), and 135.0% (IQR: 66.3% to 270.4%), respectively. A cosine wave-shaped 24-h oscillation of normalized NP levels (BNP, MR-proANP, and NT-proBNP) was noted both in lean and obese individuals (prhythmicity Conclusions This human physiological trial elucidates evidence of diurnal NP rhythmicity and the presence of an NP-BP rhythm axis. There exists a misalignment of the NP-BP diurnal rhythm in the obese, which may contribute to the disturbed diurnal BP pattern observed among obese individuals. (The Diurnal Rhythm in Natriuretic Peptide Levels; NCT03834168 )
- Published
- 2021
11. Quantitative proteomic analysis of diabetes mellitus in heart failure with preserved ejection fraction
- Author
-
Lei Zhao, Peter Schafer, David A. Gordon, Payman Zamani, Yi Jia, Francisco Ramirez-Valle, Julio A. Chirinos, Zhaoqing Wang, Dietmar A. Seiffert, Ali Javaheri, Ernst Rietzschel, Thomas C. Hanff, Leonard P. Adam, Alice M. Walsh, Thomas P. Cappola, Stuart B. Prenner, Jordana B. Cohen, Mary Ellen Cvijic, and Joseph Maranville
- Subjects
0301 basic medicine ,Cardiac function curve ,medicine.medical_specialty ,Apolipoprotein B ,HFpEF, heart failure with preserved ejection fraction ,heart failure ,030204 cardiovascular system & hematology ,Cardiovascular death ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,proteomics ,Clinical Research ,Diabetes mellitus ,Internal medicine ,DM, diabetes mellitus ,Medicine and Health Sciences ,apolipoprotein M ,Medicine ,In patient ,mediation analysis ,Aldosterone ,CILP2, cartilage intermediate layer protein 2 ,LASSO, least absolute shrinkage and selection operator ,biology ,diabetes ,business.industry ,medicine.disease ,HR, hazard ratio ,CI, confidence interval ,030104 developmental biology ,chemistry ,Heart failure ,biology.protein ,Cardiology ,ApoM, apolipoprotein M ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,business - Abstract
Visual Abstract, Highlights • DM is a significant risk factor for major adverse cardiovascular events in patients with HFpEF. • Patients with diabetes with HFpEF have a distinct proteome compared with patients without diabetes with HFpEF. • Proteomics analysis identified higher levels of alpha-1-microglobulin/bikunin precursor protein in patients with diabetes with HFpEF and lower levels of CILP2 and Apo M. • Lower Apo M levels mediate most of the association between diabetes and major adverse cardiovascular events in HFpEF., Summary Diabetes mellitus (DM) is associated with a higher risk of heart failure hospitalization and mortality in patients with heart failure with preserved ejection fraction (HFpEF). Using SomaScan assays and proteomics analysis of plasma from participants in the TOPCAT (Treatment of Preserved Cardiac Function Heart Failure with an Aldosterone Antagonist) trial and the Penn Heart Failure Study, this study identified 10 proteins with significantly different expression in patients with HFpEF and DM. Of these, apolipoprotein M was found to mediate 72% (95% CI: 36% to 100%; p
- Published
- 2021
12. Abstract 17325: Obesity, Blood Pressure, and Diurnal Variation in Natriuretic Peptides: A Clinical Trial
- Author
-
Vibhu Parcha, Peng Li, Thomas P. Cappola, Orlando M. Gutiérrez, Nirav Patel, Garima Arora, Thomas J. Wang, Kiran Musunuru, Pankaj Arora, and Kenneth B. Margulies
- Subjects
medicine.medical_specialty ,business.industry ,medicine.drug_class ,Diurnal temperature variation ,medicine.disease ,Obesity ,Clinical trial ,Blood pressure ,Endocrinology ,Physiology (medical) ,Internal medicine ,medicine ,Natriuretic peptide ,Cardiology and Cardiovascular Medicine ,business ,Hypertension experimental - Abstract
Background: Obese individuals have disturbed blood pressure (BP) rhythmicity and relative natriuretic peptide (NP) deficiency. The relationship of diurnal variation in NP levels and 24h BP rhythm is not known. Furthermore, mechanisms behind difference in circulating NPs in healthy obese and lean individuals has not been explored. We conducted a prospective clinical trial to evaluate 1) the diurnal rhythmicity of NPs and its relationship with 24-hour BP rhythm between healthy lean and obese individuals, and 2) elucidate mechanism behind NP deficiency in obese. Methods: Healthy, normotensive, lean (BMI:18.5-25 kg/m 2 ) and obese (BMI:30-45 kg/m 2 ) individuals aged 18-40 years, underwent 24-hour standardized inpatient protocol involving ambulatory BP monitoring (ABPM), controlled light intensity, and 10 blood draws, following 5-days of standardized diet. NP gene expression was evaluated in a cohort of 37 healthy donor heart tissues obtained from the MAGNet repository. Results: Among 52 participants screened, a total of 40 participants (18 lean; 22 obese) were enrolled. Diurnal variation in MRproANP levels was seen in both lean and obese individuals (p for rhythmicity=0.001). The mesor of the NP rhythm was 15% (8.5-21.6%) lower in obese. The diurnal variation in MRproANP was in antiphase with diurnal variation of systolic BP (pFigure ). Obese participants had lower 24h renin levels (p=0.06) and higher nocturnal sodium excretion (p=0.08). Among obese, there was lower expression of NP production genes ( NPPA, NPPB) (p, and higher expression of clearance gene ( NPR3 ) (p Conclusions: In a mechanistic human trial, we elucidate key neurohormonal differences in rhythm and evidence of poor salt handling in obese. Decreased production and increased clearance may contribute to the NP deficiency in obese. Targeting the diurnal NP-BP rhythm axis may reduce the cardiovascular risk burden, specifically in obese individuals.
- Published
- 2020
13. Studying Heart Failure Through the Lens of Gene Regulation
- Author
-
Thomas P. Cappola and Megan F. Burke
- Subjects
Heart Failure ,Regulation of gene expression ,business.industry ,MEDLINE ,Gene Expression ,Heart ,Genomics ,Bioinformatics ,medicine.disease ,Article ,Gene Expression Regulation ,Heart failure ,medicine ,Humans ,Promoter Regions, Genetic ,Cardiology and Cardiovascular Medicine ,business - Abstract
BACKGROUND: The failing heart is characterized by changes in gene expression. However, the regulatory regions of the genome that drive these gene expression changes have not been well defined in human hearts. METHODS: To define genomewide enhancer and promoter use in heart failure, Cap Analysis of Gene Expression (CAGE sequencing) was applied to three healthy and four failed human hearts to identify promoter and enhancer regions used in left ventricles. Healthy hearts were derived from donors unused for transplantation and failed hearts were obtained as discarded tissue after transplantation. RESULTS: CAGE sequencing identified a combined potential for ~23,000 promoters and ~5000 enhancers active in human left ventricles. Of these, 17,000 promoters and 1800 enhancers had additional support for their regulatory function. Comparing promoter usage between healthy and failed hearts highlighted promoter shifts which altered amino-terminal protein sequences. Enhancer usage between healthy and failed hearts identified a majority of differentially utilized heart failure enhancers were intronic and primarily localized within the first intron, revealing this position as a common feature associated with tissue-specific gene expression changes in the heart. CONCLUSIONS: This dataset defines the dynamic genomic regulatory landscape underlying heart failure and serves as an important resource for understanding genetic contributions to cardiac dysfunction. Additionally, regulatory changes contributing to heart failure are attractive therapeutic targets for controlling ventricular remodeling and clinical progression.
- Published
- 2020
14. Clinical and Proteomic Correlates of Plasma ACE2 (Angiotensin-Converting Enzyme 2) in Human Heart Failure
- Author
-
Lei Zhao, Nancy K. Sweitzer, Peter Schafer, Michael Basso, Dietmar A. Seiffert, Francisco Ramirez-Valle, Julio A. Chirinos, Michael Morley, Zhaoqing Wang, David A. Gordon, Vicente F. Corrales-Medina, Jeff Brandimarto, Christina Ebert, Ron Anmar, Payman Zamani, Priyanka Bhattacharya, Thomas C. Hanff, Thomas P. Cappola, James C. Fang, Yi Jia, and Jordana B. Cohen
- Subjects
0301 basic medicine ,Male ,Proteomics ,medicine.medical_specialty ,Pneumonia, Viral ,030204 cardiovascular system & hematology ,Peptidyl-Dipeptidase A ,Endocytosis ,Sensitivity and Specificity ,Severity of Illness Index ,Disease Outbreaks ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Renin–angiotensin system ,Internal Medicine ,medicine ,Humans ,Pandemics ,Proportional Hazards Models ,Retrospective Studies ,Heart Failure ,Academic Medical Centers ,Analysis of Variance ,business.industry ,COVID-19 ,Middle Aged ,Actin cytoskeleton ,medicine.disease ,Prognosis ,Blood proteins ,Protein ubiquitination ,United States ,030104 developmental biology ,Endocrinology ,Heart failure ,ACE inhibitor ,Angiotensin-converting enzyme 2 ,Disease Progression ,Linear Models ,Female ,Angiotensin-Converting Enzyme 2 ,business ,Coronavirus Infections ,hormones, hormone substitutes, and hormone antagonists ,Biomarkers ,medicine.drug - Abstract
ACE2 (angiotensin-converting enzyme 2) is a key component of the renin-angiotensin-aldosterone system. Yet, little is known about the clinical and biologic correlates of circulating ACE2 levels in humans. We assessed the clinical and proteomic correlates of plasma (soluble) ACE2 protein levels in human heart failure. We measured plasma ACE2 using a modified aptamer assay among PHFS (Penn Heart Failure Study) participants (n=2248). We performed an association study of ACE2 against ≈5000 other plasma proteins measured with the SomaScan platform. Plasma ACE2 was not associated with ACE inhibitor and angiotensin-receptor blocker use. Plasma ACE2 was associated with older age, male sex, diabetes mellitus, a lower estimated glomerular filtration rate, worse New York Heart Association class, a history of coronary artery bypass surgery, and higher pro-BNP (pro-B-type natriuretic peptide) levels. Plasma ACE2 exhibited associations with 1011 other plasma proteins. In pathway overrepresentation analyses, top canonical pathways associated with plasma ACE2 included clathrin-mediated endocytosis signaling, actin cytoskeleton signaling, mechanisms of viral exit from host cells, EIF2 (eukaryotic initiation factor 2) signaling, and the protein ubiquitination pathway. In conclusion, in humans with heart failure, plasma ACE2 is associated with various clinical factors known to be associated with severe coronavirus disease 2019 (COVID-19), including older age, male sex, and diabetes mellitus, but is not associated with ACE inhibitor and angiotensin-receptor blocker use. Plasma ACE2 protein levels are prominently associated with multiple cellular pathways involved in cellular endocytosis, exocytosis, and intracellular protein trafficking. Whether these have a causal relationship with ACE2 or are relevant to novel coronavirus-2 infection remains to be assessed in future studies.
- Published
- 2020
15. Assigning Distal Genomic Enhancers to Cardiac Disease-Causing Genes
- Author
-
Kenneth B. Margulies, Matias Ilmari Autio, Roger Foo, Arnaud Perrin, Bangfen Pan, Michael Morley, Wilson Lek Wen Tan, Bram Lim, Chang Jie Mick Lee, Chukwuemeka George Anene-Nzelu, Zheng Wenhao, Zenia Tiang, Thomas P. Cappola, Hui San Tan, Albert Dashi, and Eleanor Wong
- Subjects
Enhancer Elements ,Heart Diseases ,business.industry ,Human Embryonic Stem Cells ,Computational biology ,Disease ,Genome ,Article ,Chromatin ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Physiology (medical) ,Medicine ,Humans ,Myocytes, Cardiac ,Epigenetics ,Cardiology and Cardiovascular Medicine ,business ,Enhancer ,Gene - Published
- 2020
16. Reduced Apolipoprotein M and Adverse Outcomes Across the Spectrum of Human Heart Failure
- Author
-
Björn Dahlbäck, Douglas L. Mann, Nancy K. Sweitzer, Dietmar A. Seiffert, David A. Gordon, Christina Chistoffersen, James C. Fang, Lei Zhao, Swapnil V. Shewale, Thomas P. Cappola, Luigi Adamo, Kenneth B. Margulies, Daniel J. Rader, Jeff Brandimarto, Ali Javaheri, John S. Millar, Bruce D. Car, Cecilia Frej, Julio A. Chirinos, Zhaoqing Wang, Yi Jia, John S. Parks, and Benjamin French
- Subjects
Male ,Proteomics ,medicine.medical_specialty ,Time Factors ,Apolipoprotein B ,Proteome ,Adverse outcomes ,Down-Regulation ,Apolipoproteins M ,030204 cardiovascular system & hematology ,Risk Assessment ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Risk Factors ,Sphingosine ,Physiology (medical) ,Internal medicine ,medicine ,Humans ,Sphingosine-1-phosphate ,Registries ,030304 developmental biology ,Aged ,Randomized Controlled Trials as Topic ,Heart Failure ,0303 health sciences ,biology ,business.industry ,Human heart ,Physical interaction ,Middle Aged ,medicine.disease ,Prognosis ,United States ,Endocrinology ,chemistry ,Heart failure ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Female ,Lysophospholipids ,Cardiology and Cardiovascular Medicine ,business ,Lipoproteins, HDL ,Biomarkers ,Lipoprotein - Abstract
Background: Apo (apolipoprotein) M mediates the physical interaction between high-density lipoprotein (HDL) particles and sphingosine-1-phosphate (S1P). Apo M exerts anti-inflammatory and cardioprotective effects in animal models. Methods: In a subset of PHFS (Penn Heart Failure Study) participants (n=297), we measured apo M by Enzyme-Linked ImmunoSorbent Assay (ELISA). We also measured total S1P by liquid chromatography–mass spectrometry and isolated HDL particles to test the association between apo M and HDL-associated S1P. We confirmed the relationship between apo M and outcomes using modified aptamer-based apo M measurements among 2170 adults in the PHFS and 2 independent cohorts: the Washington University Heart Failure Registry (n=173) and a subset of TOPCAT (Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist Trial; n=218). Last, we examined the relationship between apo M and ≈5000 other proteins (SomaScan assay) to identify biological pathways associated with apo M in heart failure. Results: In the PHFS, apo M was inversely associated with the risk of death (standardized hazard ratio, 0.56 [95% CI, 0.51–0.61]; P P P P =0.001) in models that adjusted for multiple confounders. This association was present in both heart failure with reduced and preserved ejection fraction and was replicated in the Washington University cohort and a cohort with heart failure with preserved ejection fraction only (TOPCAT). The S1P and apo M content of isolated HDL particles strongly correlated ( R =0.81, P Conclusions: Reduced circulating apo M is independently associated with adverse outcomes across the spectrum of human heart failure. Further research is needed to assess whether the apo M/S1P axis is a suitable therapeutic target in heart failure.
- Published
- 2020
17. Aptamer-Based Proteomic Platform Identifies Novel Protein Predictors of Incident Heart Failure and Echocardiographic Traits
- Author
-
Janine F. Felix, Mark D. Benson, Dongxiao Shen, Martin G. Larson, Thomas P. Cappola, Thomas J. Wang, Philipp S. Wild, Torbjørn Omland, Meghan I. Short, Alanna C. Morrison, Humaira Rasheed, Michael Morley, Qiong Yang, Sumita Sinha, Christian Jonasson, Ramachandran S. Vasan, Michelle J. Keyes, Kristian Hveem, Honghuang Lin, Robert E. Gerszten, Ben Michael Brumpton, Debby Ngo, Matthew Nayor, and Epidemiology
- Subjects
Male ,Proteomics ,Aptamer ,Computational biology ,030204 cardiovascular system & hematology ,Risk Assessment ,03 medical and health sciences ,0302 clinical medicine ,Predictive Value of Tests ,Risk Factors ,Medicine ,Humans ,Genetic Predisposition to Disease ,Prospective Studies ,030304 developmental biology ,Aged ,Heart Failure ,0303 health sciences ,Ventricular Remodeling ,business.industry ,Novel protein ,Norway ,Incidence ,Genetic Variation ,Blood Proteins ,Aptamers, Nucleotide ,Middle Aged ,medicine.disease ,High-Throughput Screening Assays ,Circulating biomarkers ,Cross-Sectional Studies ,Phenotype ,Massachusetts ,Echocardiography ,Heart failure ,Female ,Cardiology and Cardiovascular Medicine ,business ,Biomarkers - Abstract
Background: We used a large-scale, high-throughput DNA aptamer-based discovery proteomic platform to identify circulating biomarkers of cardiac remodeling and incident heart failure (HF) in community-dwelling individuals. Methods: We evaluated 1895 FHS (Framingham Heart Study) participants (age 55±10 years, 54% women) who underwent proteomic profiling and echocardiography. Plasma levels of 1305 proteins were related to echocardiographic traits and to incident HF using multivariable regression. Statistically significant protein-HF associations were replicated in the HUNT (Nord-Trøndelag Health) study (n=2497, age 63±10 years, 43% women), and results were meta-analyzed. Genetic variants associated with circulating protein levels (pQTLs) were related to echocardiographic traits in the EchoGen (n=30 201) and to incident HF in the CHARGE (n=20 926) consortia. Results: Seventeen proteins associated with echocardiographic traits in cross-sectional analyses (false discovery rate P P − 5 ; 3 with higher risk: NT-proBNP [N-terminal proB-type natriuretic peptide], TSP2 [thrombospondin-2], MBL [mannose-binding lectin]; and 3 with lower risk: ErbB1 [epidermal growth factor receptor], GDF-11/8 [growth differentiation factor-11/8], and RGMC [hemojuvelin]). For 5 of the 6 proteins, pQTLs were associated with echocardiographic traits ( P P =0.001). Conclusions: A large-scale proteomics approach identified new predictors of cardiac remodeling and incident HF. Future studies are warranted to elucidate how biological pathways represented by these proteins may mediate cardiac remodeling and HF risk and to assess if these proteins can improve HF risk prediction.
- Published
- 2020
18. 42855 A Phenomics Approach to the Categorization and Refinement of Heart Failure
- Author
-
William P. Bone, Anurag Verma, Ashwin C. Murthy, Srinath Adusumalli, Macrylyn D. Ritchie, Srinivas Denduluri, Nosheen Reza, Thomas P. Cappola, and Pankhuri Singhal
- Subjects
Phenomics ,Categorization ,Computer science ,business.industry ,Heart failure ,medicine ,General Medicine ,Artificial intelligence ,Machine learning ,computer.software_genre ,medicine.disease ,business ,computer - Abstract
IMPACT: Measuring and analyzing qualitative and quantitative traits using phenomics approaches will yield previously unrecognized heart failure subphenotypes and has the potential to improve our knowledge of heart failure pathophysiology, identify novel biomarkers of disease, and guide the development of targeted therapeutics for heart failure. OBJECTIVES/GOALS: Current classification schemes fail to capture the broader pathophysiologic heterogeneity in heart failure. Phenomics offers a newer unbiased approach to identify subtypes of complex disease syndromes, like heart failure. The goal of this research is to use data-driven associations to redefine the classification of the heart failure syndrome. METHODS/STUDY POPULATION: We will identify < 10 subphenotypes of patients with heart failure using unsupervised machine learning approaches for dense multidimensional quantitative (i.e. demographics, comorbid conditions, physiologic measurements, clinical laboratory, imaging, and medication variables; disease diagnosis, procedure, and billing codes) and qualitative data extracted from an integrated health system electronic health record. The heart failure subphenotypes we identify from the integrated health system electronic health record will be replicated in other heart failure population datasets using unsupervised learning approaches. We will explore the potential to establish associations between identified subphenotypes and clinical outcomes (e.g. all-cause mortality, cardiovascular mortality). RESULTS/ANTICIPATED RESULTS: We expect to identify < 10 mutually exclusive phenogroups of patients with heart failure that have differential risk profiles and clinical trajectories. DISCUSSION/SIGNIFICANCE OF FINDINGS: We will attempt to derive and validate a data-driven unbiased approach to the categorization of novel phenogroups in heart failure. This has the potential to improve our knowledge of heart failure pathophysiology, identify novel biomarkers of disease, and guide the development of targeted therapeutics for heart failure.
- Published
- 2021
19. Mental Stress–Induced Myocardial Ischemia
- Author
-
Thomas P. Cappola and Paco E Bravo
- Subjects
medicine.medical_specialty ,Myocardial ischemia ,business.industry ,Myocardial Ischemia ,MEDLINE ,Heart ,Coronary Artery Disease ,General Medicine ,medicine.disease_cause ,Article ,Text mining ,Internal medicine ,Mental stress ,medicine ,Cardiology ,Humans ,Psychological stress ,business ,Stress, Psychological ,Original Investigation - Abstract
IMPORTANCE: Mental stress–induced myocardial ischemia is a recognized phenomenon in patients with coronary heart disease (CHD), but its clinical significance in the contemporary clinical era has not been investigated. OBJECTIVE: To compare the association of mental stress–induced or conventional stress–induced ischemia with adverse cardiovascular events in patients with CHD. DESIGN, SETTING, AND PARTICIPANTS: Pooled analysis of 2 prospective cohort studies of patients with stable CHD from a university-based hospital network in Atlanta, Georgia: the Mental Stress Ischemia Prognosis Study (MIPS) and the Myocardial Infarction and Mental Stress Study 2 (MIMS2). Participants were enrolled between June 2011 and March 2016 (last follow-up, February 2020). EXPOSURES: Provocation of myocardial ischemia with a standardized mental stress test (public speaking task) and with a conventional (exercise or pharmacological) stress test, using single-photon emission computed tomography. MAIN OUTCOMES AND MEASURES: The primary outcome was a composite of cardiovascular death or first or recurrent nonfatal myocardial infarction. The secondary end point additionally included hospitalizations for heart failure. RESULTS: Of the 918 patients in the total sample pool (mean age, 60 years; 34% women), 618 participated in MIPS and 300 in MIMS2. Of those, 147 patients (16%) had mental stress–induced ischemia, 281 (31%) conventional stress ischemia, and 96 (10%) had both. Over a 5-year median follow-up, the primary end point occurred in 156 participants. The pooled event rate was 6.9 per 100 patient-years among patients with and 2.6 per 100 patient-years among patients without mental stress–induced ischemia. The multivariable adjusted hazard ratio (HR) for patients with vs those without mental stress–induced ischemia was 2.5 (95% CI, 1.8-3.5). Compared with patients with no ischemia (event rate, 2.3 per 100 patient-years), patients with mental stress–induced ischemia alone had a significantly increased risk (event rate, 4.8 per 100 patient-years; HR, 2.0; 95% CI, 1.1-3.7) as did patients with both mental stress ischemia and conventional stress ischemia (event rate, 8.1 per 100 patient-years; HR, 3.8; 95% CI, 2.6-5.6). Patients with conventional stress ischemia alone did not have a significantly increased risk (event rate, 3.1 per 100 patient-years; HR, 1.4; 95% CI, 0.9-2.1). Patients with both mental stress ischemia and conventional stress ischemia had an elevated risk compared with patients with conventional stress ischemia alone (HR, 2.7; 95% CI, 1.7-4.3). The secondary end point occurred in 319 participants. The event rate was 12.6 per 100 patient-years for patients with and 5.6 per 100 patient-years for patients without mental stress–induced ischemia (adjusted HR, 2.0; 95% CI, 1.5-2.5). CONCLUSIONS AND RELEVANCE: Among patients with stable coronary heart disease, the presence of mental stress–induced ischemia, compared with no mental stress–induced ischemia, was significantly associated with an increased risk of cardiovascular death or nonfatal myocardial infarction. Although these findings may provide insights into mechanisms of myocardial ischemia, further research is needed to assess whether testing for mental stress–induced ischemia has clinical value.
- Published
- 2021
20. Effects of organic and inorganic nitrate on aortic and carotid haemodynamics in heart failure with preserved ejection fraction
- Author
-
Swapna Varakantam, Payman Zamani, Kenneth B. Margulies, Izzah Vasim, Timothy S Phan, Francisco Londono-Hoyos, Raymond R. Townsend, Melissa Beraun, Julio A. Chirinos, Patrick Segers, Thomas P. Cappola, and Philip Haines
- Subjects
Aorta ,medicine.medical_specialty ,business.industry ,Hemodynamics ,Vasodilation ,030204 cardiovascular system & hematology ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Blood pressure ,Heart failure ,medicine.artery ,Internal medicine ,Vascular resistance ,medicine ,Isosorbide mononitrate ,Cardiology ,030212 general & internal medicine ,Cardiology and Cardiovascular Medicine ,business ,Heart failure with preserved ejection fraction ,medicine.drug - Abstract
Aims To assess the haemodynamic effects of organic vs. inorganic nitrate administration among patients with heart failure with preserved ejection fraction (HFpEF). Methods and results We assessed carotid and aortic pressure–flow relations non-invasively before and after the administration of 0.4 mg of sublingual nitroglycerin (n = 26), and in a separate sub-study, in response to 12.9 mmoL of inorganic nitrate (n = 16). Nitroglycerin did not consistently reduce wave reflections arriving at the proximal aorta (change in real part of reflection coefficient, 1st harmonic: −0.09; P = 0.01; 2nd harmonic: −0.045, P = 0.16; 3rd harmonic: +0.087; P = 0.05), but produced profound vasodilatation in the carotid territory, with a significant reduction in systolic blood pressure (133.6 vs. 120.5 mmHg; P = 0.011) and a marked reduction in carotid bed vascular resistance (19 580 vs. 13 078 dynes · s/cm5; P = 0.001) and carotid characteristic impedance (3440 vs. 1923 dynes · s/cm5; P = 0.002). Inorganic nitrate, in contrast, consistently reduced wave reflections across the first three harmonics (change in real part of reflection coefficient, 1st harmonic: −0.12; P = 0.03; 2nd harmonic: −0.11, P = 0.01; 3rd harmonic: −0.087; P = 0.09) and did not reduce blood pressure, carotid bed vascular resistance, or carotid characteristic impedance (P = NS). Conclusions Nitroglycerin produces marked vasodilatation in the carotid circulation, with a pronounced reduction in blood pressure and inconsistent effects on central wave reflections. Inorganic nitrate, in contrast, produces consistent reductions in wave reflections, and unlike nitroglycerin, it does so without significant hypotension or cerebrovascular dilatation. These haemodynamic differences may underlie the different effects on exercise capacity and side effect profile of inorganic vs. organic nitrate in HFpEF.
- Published
- 2017
21. CYP3A4 genotype is associated with sildenafil concentrations in patients with heart failure with preserved ejection fraction
- Author
-
Douglas L. Mann, Ian Mongrain, S. de Denus, René Fouodjio, Thomas P. Cappola, Naveen L. Pereira, Marie-Pierre Dubé, Svati H. Shah, Jean L. Rouleau, and Gordon S. Huggins
- Subjects
0301 basic medicine ,Male ,Vasodilator Agents ,030226 pharmacology & pharmacy ,law.invention ,chemistry.chemical_compound ,0302 clinical medicine ,Randomized controlled trial ,law ,Medicine ,Cytochrome P-450 CYP3A ,pharmacogenetics ,education.field_of_study ,Ejection fraction ,Exercise Tolerance ,Middle Aged ,Cardiology ,cardiovascular system ,Molecular Medicine ,Female ,Cardiology and Cardiovascular Medicine ,pharmacokinetics ,medicine.medical_specialty ,Genotype ,Sildenafil ,Population ,Article ,Sildenafil Citrate ,03 medical and health sciences ,Pharmacokinetics ,Internal medicine ,Genetics ,Humans ,education ,Aged ,Pharmacology ,Heart Failure ,business.industry ,Repeated measures design ,Stroke Volume ,medicine.disease ,respiratory tract diseases ,030104 developmental biology ,Endocrinology ,chemistry ,Heart failure ,business ,Heart failure with preserved ejection fraction ,Pharmacogenetics - Abstract
Despite its established inter-individual variability, sildenafil has been the subject of only a few pharmacogenetic investigations, with limited data regarding the genetic modulators of its pharmacokinetics. We conducted a pharmacogenetic substudy of patients randomized to sildenafil (n = 85) in the RELAX trial, which investigated the impact of high-dose sildenafil in patients with heart failure with preserved left ventricular ejection fraction (HFpEF). In the overall population, the CYP3A4 inferred phenotype appeared associated with the dose-adjusted peak concentrations of sildenafil at week 12 and week 24 (adjusted P=0.045 for repeated measures analysis), although this P value did not meet our corrected significance threshold of 0.0167. In the more homogeneous Caucasian subgroup, this association was significant (adjusted P=0.0165 for repeated measures). Hence, CYP3A4 inferred phenotype is associated with peak sildenafil dose-adjusted concentrations in patients with HFpEF receiving high doses of sildenafil. The clinical impact of this association requires further investigation.
- Published
- 2017
22. Effect of Heart Failure With Preserved Ejection Fraction on Nitric Oxide Metabolites
- Author
-
Julio A. Chirinos, Raymond R. Townsend, Jeffrey Brandimarto, Thomas P. Cappola, Payman Zamani, Ali Javaheri, James C. Fang, Paschalis-Thomas Doulias, Kenneth B. Margulies, Benjamin French, Nancy K. Sweitzer, and Harry Ischiropoulos
- Subjects
Male ,medicine.medical_specialty ,Renal function ,030204 cardiovascular system & hematology ,Nitric Oxide ,Article ,Ventricular Function, Left ,Nitric oxide ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Matched cohort ,Interquartile range ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Renal Insufficiency, Chronic ,Aged ,Heart Failure ,Ejection fraction ,business.industry ,Smoking ,Age Factors ,Middle Aged ,medicine.disease ,Confidence interval ,chemistry ,Heart failure ,Linear Models ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,business ,Glomerular Filtration Rate - Abstract
Endothelial function may be deranged in heart failure with preserved ejection fraction (HFpEF). Serum NO-derived metabolites (NO m ) might provide a biochemical surrogate of endothelial function in patients with heart failure (HF). We measured serum NO m in 415 participants in the Penn HF Study. Participants with HFpEF (n = 82) and those whose EF had recovered (Recovered-HF, n = 125) were matched 1:1 to heart failure with reduced ejection fraction (HFrEF) participants based on age, gender, race, tobacco use, and eGFR. Serum NO m levels were quantified after chemical reduction coupled with gas-phase chemiluminescence detection. After adjustment for matching covariates and BMI, HFpEF (34.5 μM; interquartile range [IQR] 25.0, 51.5) participants had lower NO m levels than HFrEF (41.0 μM; IQR 28.3, 58.0; ratio of HFpEF:HFrEF 0.82, 95% confidence interval [CI] 0.67 to 0.99; p = 0.04), which further decreased when adjusted for covariates that affect endothelial function (ratio 0.79, 95% CI 0.65 to 0.98; p = 0.03). There were no differences between HFrEF (34.0; IQR 25.3, 49.0) and matched Recovered-HF (36.0 μM; IQR 25.0, 55.0) or HFpEF and Recovered-HF. Age (+21%/10-year increase, p m in HFpEF, whereas age (+11%/10-year increase, p = 0.03), current tobacco use (+67%, p = 0.01), and eGFR (p = 0.01) associated with NO m in Recovered-HF. In conclusion, HFpEF participants have reduced NO m compared with HFrEF in this matched cohort. This might suggest either compromised endothelial function or poor dietary intake. Black race was associated with lower NO m in HFpEF.
- Published
- 2016
23. Race, Natriuretic Peptides, and High-Carbohydrate Challenge: A Clinical Trial
- Author
-
Thomas J. Wang, Wenjian Lv, Thomas P. Cappola, Ganesh V. Halade, Kenneth B. Margulies, Vasundhara Kain, Pankaj Arora, Garima Arora, Kiran Musunuru, Griffin K. Russell, Orlando M. Gutiérrez, Nirav Patel, and Sumanth D. Prabhu
- Subjects
Adult ,Male ,medicine.medical_specialty ,High carbohydrate ,Time Factors ,Physiology ,medicine.drug_class ,Induced Pluripotent Stem Cells ,Down-Regulation ,030204 cardiovascular system & hematology ,White People ,Article ,Cell Line ,03 medical and health sciences ,Race (biology) ,0302 clinical medicine ,Internal medicine ,Gene expression ,microRNA ,Natriuretic Peptide, Brain ,medicine ,Natriuretic peptide ,Dietary Carbohydrates ,Humans ,Myocytes, Cardiac ,030212 general & internal medicine ,Prospective Studies ,business.industry ,Health Status Disparities ,Healthy Volunteers ,Peptide Fragments ,Race Factors ,Clinical trial ,Black or African American ,Endocrinology ,Alabama ,Female ,Cardiology and Cardiovascular Medicine ,business ,Atrial Natriuretic Factor ,Biomarkers - Abstract
Rationale: Lower NP (natriuretic peptide) levels may contribute to the development of cardiometabolic diseases. Blacks have lower NP levels than middle-aged and older white adults. A high-carbohydrate challenge causes an upregulation of a negative ANP regulator microRNA-425 ( miR-425 ), which reduces ANP (atrial-NP) levels in whites. Objectives: We designed a prospective trial to study racial differences in (1) NP levels among young adults, (2) NP response to a high-carbohydrate challenge, and (3) explore underlying mechanisms for race-based differences. Methods and Results: Healthy self-identified blacks and whites received 3 days of study diet followed by a high-carbohydrate challenge. Gene expression from whole blood RNA was assessed in the trial participants. Additionally, atrial and ventricular tissue samples from the Myocardial Applied Genomics Network repository were examined for NP system gene expression. Among 72 healthy participants, we found that B-type-NP, NT-proBNP (N-terminal-pro-B-type NP), and MRproANP (midregional-pro-ANP) levels were 30%, 47%, and 18% lower in blacks compared with whites ( P ≤0.01), respectively. The decrease in MRproANP levels in response to a high-carbohydrate challenge differed by race (blacks 23% [95% CI, 19%–27%] versus whites 34% [95% CI, 31%–38]; P interaction NPPA/NPPB ) or NP signaling ( NPR1 ) in atrial and ventricular tissues. NP processing ( corin ), clearance ( NPR3 ), and regulation ( miR-425 ) genes were ≈3.5-, ≈2.5-, and ≈2-fold higher in blacks than whites in atrial tissues, respectively. We also found a 2-and 8-fold higher whole blood RNA expression of gene encoding for Neprilysin ( MME ) and miR-425 among blacks than whites. Conclusions: Racial differences in NP levels are evident in young, healthy adults suggesting a state of NP deficiency exists in blacks. Impaired NP processing and clearance may contribute to race-based NP differences. Higher miR-425 levels in blacks motivate additional studies to understand differences in NP downregulation after physiological perturbations. Clinical Trial Registration: URL: https://clinicaltrials.gov/ct2/show/NCT03072602 . Unique identifier: NCT03072602.
- Published
- 2019
24. Genome-wide association study provides new insights into the genetic architecture and pathogenesis of heart failure
- Author
-
Stefan Gross, Honghuang Lin, Diane T. Smelser, März W, Niek Verweij, Alexander Niessner, Peter M. Visscher, Ruth C. Lovering, Erik Ingelsson, Nicholas L. Smith, Lars Lind, Jerome I. Rotter, Anna Helgadottir, David J. Stott, Michelle L. O'Donoghue, Daniel I. Chasman, Karoline Kuchenbaecker, Morris Ad, Michael W. Nagle, Morris Ap, Steven A. Lubitz, Giedraitis, Abirami Veluchamy, Paul M. Ridker, David J. Carey, Benoit Tyl, Carolina Roselli, Laura M. Yerges-Armstrong, Michael R. Brown, Faiez Zannad, Aroon D. Hingorani, Åsa K. Hedman, Nilesh J. Samani, Daníel F. Guðbjartsson, Maris Teder-Laving, Stephan B. Felix, Christopher Newton-Cheh, Marcus Dörr, Thomas P. Cappola, Daniel I. Swerdlow, Leonard Buckbinder, Xiaosong Wang, Alex S. F. Doney, C. Andersson, Jonathan H. Chung, Joshua D. Backman, Tõnu Esko, Mohsen Ghanbari, Joop Jukema, Ian Ford, Gunnar Engström, Kent D. Taylor, Johan Ärnlöv, P.E Weeke, Kenneth B. Margulies, Peter Svensson, Folkert W. Asselbergs, Lars Køber, Michael V. Holmes, Anders Mälarstig, Chim C. Lang, Krishna G. Aragam, Samuel C. Dudley, Christian Torp-Pedersen, Bruce M. Psaty, Alexander Teumer, John J.V. McMurray, Raul Weiss, Smith Jg, Patrick T. Ellinor, Anjali T. Owens, Ify R. Mordi, G Sveinbjörnsson, Luca A. Lotta, John S. Gottdiener, Christopher M. Haggerty, Christopher P. Nelson, C M Lindgren, Hilma Holm, Michael E. Dunn, Albert Henry, Helen M. Parry, Salomaa, Huilin Xing, Kenneth Rice, Marcus E. Kleber, Spiros Denaxas, Chris Finan, Xu Chen, Unnur Thorsteinsdottir, Naveed Sattar, Bing Yu, Kerri L. Wiggins, Alaa Shalaby, Romaine Spr., Patrik K. E. Magnusson, Adriaan A. Voors, Lumbers Rt, Stella Trompet, Maryam Kavousi, Kari Stefansson, Jian'an Luan, Harry Hemingway, van Setten J, Anubha Mahajan, Olle Melander, Uwe Völker, Peter Almgren, Chaffin, Palmer Cna., Eliana Portilla-Fernandez, Stefan Stender, Markus Perola, Guðmundur Thorgeirsson, Ghazaleh Fatemifar, Teemu J. Niiranen, Nicholas J. Wareham, Andrea Koekemoer, James P. Cook, Jeff Brandimarto, Jian Yang, Mary L. Biggs, Claudia Langenberg, Perttu Salo, Sonia Shah, A.G. Uitterlinden, Mari-Liis Tammesoo, Jemma B. Wilk, Craig L. Hyde, Graciela E. Delgado, van der Harst P, Alanna C. Morrison, Kay-Tee Khaw, Barry London, Rebecca Gutmann, Thomas M. Morgan, Franco Giulianini, Abbas Dehghan, Heather L. Bloom, William A. Chutkow, Dawn M. Waterworth, Ramachandran S. Vasan, Jing Hua Zhao, Morley Mp, and Sahar Ghasemi
- Subjects
2. Zero hunger ,0303 health sciences ,business.industry ,Atrial fibrillation ,Genome-wide association study ,030204 cardiovascular system & hematology ,Quantitative trait locus ,medicine.disease ,Bioinformatics ,Genetic architecture ,3. Good health ,Coronary artery disease ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Heart failure ,medicine ,Mendelian inheritance ,symbols ,business ,030304 developmental biology ,Genetic association - Abstract
Heart failure (HF) is a leading cause of morbidity and mortality worldwide1. A small proportion of HF cases are attributable to monogenic cardiomyopathies and existing genome-wide association studies (GWAS) have yielded only limited insights, leaving the observed heritability of HF largely unexplained2–4. We report the largest GWAS meta-analysis of HF to-date, comprising 47,309 cases and 930,014 controls. We identify 12 independent variant associations with HF at 11 genomic loci, all of which demonstrate one or more associations with coronary artery disease (CAD), atrial fibrillation, or reduced left ventricular function suggesting shared genetic aetiology. Expression quantitative trait analysis of non-CAD-associated loci implicate genes involved in cardiac development (MYOZ1, SYNPO2L), protein homeostasis (BAG3), and cellular senescence (CDKN1A). Using Mendelian randomisation analysis we provide new evidence supporting previously equivocal causal roles for several HF risk factors identified in observational studies, and demonstrate CAD-independent effects for atrial fibrillation, body mass index, hypertension and triglycerides. These findings extend our knowledge of the genes and pathways underlying HF and may inform the development of new therapeutic approaches.
- Published
- 2019
- Full Text
- View/download PDF
25. Genomics-First Evaluation of Heart Disease Associated With Titin-Truncating Variants
- Author
-
Joseph B. Leader, Diane T. Smelser, Kenneth B. Margulies, Aris Baras, Aeron Small, Heather Williams, Zoltan Arany, Alicia Golden, Chris McDermott-Roe, Michael E. Hall, Frederick E. Dewey, Richard C. Stahl, Thomas P. Cappola, Rachel L. Kember, Marylyn D. Ritchie, Daniel J. Rader, David Birtwell, Scott M. Damrauer, Adolfo Correa, Yirui Hu, Xinyuang Zhang, Apoorva Babu, David J. Carey, Melissa A. Kelly, Michael Morley, Arichanah Pulenthiran, James G. Wilson, H. Lester Kirchner, Michael G. Levin, Dustin N. Hartzel, Michael F. Murray, Zachariah Nealy, Lusha Liang, Thomas N. Person, Renae Judy, Amy C. Sturm, Christopher M. Haggerty, Brandon K. Fornwalt, and Anurag Verma
- Subjects
Adult ,Male ,Heart disease ,Heart Diseases ,Genomics ,030204 cardiovascular system & hematology ,Bioinformatics ,White People ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Idiopathic dilated cardiomyopathy ,medicine ,Electronic Health Records ,Humans ,Connectin ,Longitudinal Studies ,030304 developmental biology ,Aged ,0303 health sciences ,biology ,business.industry ,Genetic Variation ,Dilated cardiomyopathy ,Middle Aged ,medicine.disease ,biology.protein ,Titin ,Female ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background: Truncating variants in the Titin gene (TTNtvs) are common in individuals with idiopathic dilated cardiomyopathy (DCM). However, a comprehensive genomics-first evaluation of the impact of TTNtvs in different clinical contexts, and the evaluation of modifiers such as genetic ancestry, has not been performed. Methods: We reviewed whole exome sequence data for >71 000 individuals (61 040 from the Geisinger MyCode Community Health Initiative (2007 to present) and 10 273 from the PennMedicine BioBank (2013 to present) to identify anyone with TTNtvs. We further selected individuals with TTNtvs in exons highly expressed in the heart (proportion spliced in [PSI] >0.9). Using linked electronic health records, we evaluated associations of TTNtvs with diagnoses and quantitative echocardiographic measures, including subanalyses for individuals with and without DCM diagnoses. We also reviewed data from the Jackson Heart Study to validate specific analyses for individuals of African ancestry. Results: Identified with a TTNtv in a highly expressed exon (hiPSI) were 1.2% individuals in PennMedicine BioBank and 0.6% at Geisinger. The presence of a hiPSI TTNtv was associated with increased odds of DCM in individuals of European ancestry (odds ratio [95% CI]: 18.7 [9.1–39.4] {PennMedicine BioBank} and 10.8 [7.0–16.0] {Geisinger}). hiPSI TTNtvs were not associated with DCM in individuals of African ancestry, despite a high DCM prevalence (odds ratio, 1.8 [0.2–13.7]; P =0.57). Among 244 individuals of European ancestry with DCM in PennMedicine BioBank, hiPSI TTNtv carriers had lower left ventricular ejection fraction (β=–12%, P =3×10 –7 ), and increased left ventricular diameter (β=0.65 cm, P =9×10 –3 ). In the Geisinger cohort, hiPSI TTNtv carriers without a cardiomyopathy diagnosis had more atrial fibrillation (odds ratio, 2.4 [1.6–3.6]) and heart failure (odds ratio, 3.8 [2.4–6.0]), and lower left ventricular ejection fraction (β=–3.4%, P =1×10 –7 ). Conclusions: Individuals of European ancestry with hiPSI TTNtv have an abnormal cardiac phenotype characterized by lower left ventricular ejection fraction, irrespective of the clinical manifestation of cardiomyopathy. Associations with arrhythmias, including atrial fibrillation, were observed even when controlling for cardiomyopathy diagnosis. In contrast, no association between hiPSI TTNtvs and DCM was discerned among individuals of African ancestry. Given these findings, clinical identification of hiPSI TTNtv carriers may alter clinical management strategies.
- Published
- 2019
26. The genetic makeup of the electrocardiogram
- Author
-
Michael Morley, Teresa Trenkwalder, Alexander Teumer, Yordi J. van de Vegte, J W Benjamins, Thomas P. Cappola, Pim van der Harst, Wibke Reinhard, Niek Verweij, and Cardiovascular Centre (CVC)
- Subjects
Histology ,In silico ,Genome-wide association study ,Disease ,Computational biology ,030204 cardiovascular system & hematology ,Genome ,Polymorphism, Single Nucleotide ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,Electrocardiography ,0302 clinical medicine ,Cardiac conduction ,medicine ,Humans ,cardiovascular diseases ,Cardiac disorders ,030304 developmental biology ,Functional validation ,0303 health sciences ,CLCNKA ,biology ,business.industry ,Genetic variants ,Dilated cardiomyopathy ,Cell Biology ,Integrated approach ,medicine.disease ,Biobank ,Phenotype ,Cohort ,biology.protein ,Snapshot (computer storage) ,business ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Since its original description in 1893 by Willem van Einthoven, the electrocardiogram (ECG) has been instrumental in the recognition of a wide array of cardiac disorders1,2. Although many electrocardiographic patterns have been well described, the underlying biology is incompletely understood. Genetic associations of particular features of the ECG have been identified by genome wide studies. This snapshot approach only provides fragmented information of the underlying genetic makeup of the ECG. Here, we follow the effecs of individual genetic variants through the complete cardiac cycle the ECG represents. We found that genetic variants have unique morphological signatures not identfied by previous analyses. By exploiting identified abberations of these morphological signatures, we show that novel genetic loci can be identified for cardiac disorders. Our results demonstrate how an integrated approach to analyse high-dimensional data can further our understanding of the ECG, adding to the earlier undertaken snapshot analyses of individual ECG components. We anticipate that our comprehensive resource will fuelin silicoexplorations of the biological mechanisms underlying cardiac traits and disorders represented on the ECG. For example, known disease causing variants can be used to identify novel morphological ECG signatures, which in turn can be utilized to prioritize genetic variants or genes for functional validation. Furthermore, the ECG plays a major role in the development of drugs, a genetic assessment of the entire ECG can drive such developments.
- Published
- 2019
- Full Text
- View/download PDF
27. Cardioprotective Effects of MTSS1 Enhancer Variants
- Author
-
Michael Morley, Ray Hu, Kiran Musunuru, Pim van der Harst, Thomas P. Cappola, Janine F. Felix, Xiao Wang, Patrick T. Ellinor, Nathan R. Tucker, Kenneth B. Margulies, Jeffrey Brandimarto, Nicholas L. Smith, Cardiovascular Centre (CVC), and Epidemiology
- Subjects
myocardial biology ,Embryo, Nonmammalian ,Heart Ventricles ,Quantitative Trait Loci ,Cardiomyopathy ,heart failure ,Genomics ,Quantitative trait locus ,Transfection ,Polymorphism, Single Nucleotide ,Article ,Mice ,Polymorphism (computer science) ,Physiology (medical) ,genomics ,medicine ,Animals ,genetics ,Enhancer ,Zebrafish ,Regulation of gene expression ,Genetics ,Mice, Knockout ,business.industry ,translational studies ,gene expression and regulation ,Microfilament Proteins ,Gene Expression Regulation, Developmental ,Embryo ,Stroke Volume ,medicine.disease ,Neoplasm Proteins ,Enhancer Elements, Genetic ,Cardiology and Cardiovascular Medicine ,business ,cardiomyopathy - Published
- 2019
28. Response by Cappola and Cappola to Letter Regarding Article, 'Thyroid Dysfunction in Heart Failure and Cardiovascular Outcomes'
- Author
-
Anne R. Cappola and Thomas P. Cappola
- Subjects
Heart Failure ,medicine.medical_specialty ,business.industry ,MEDLINE ,medicine.disease ,Cardiovascular System ,Thyroid Diseases ,Thyroid dysfunction ,Heart failure ,medicine ,Humans ,Cardiology and Cardiovascular Medicine ,Intensive care medicine ,business ,Cardiovascular outcomes - Published
- 2019
29. Thyroid Dysfunction in Heart Failure and Cardiovascular Outcomes
- Author
-
Lakshmi Kannan, Nancy K. Sweitzer, James C. Fang, Pamela A. Shaw, Michael Morley, Jeffrey Brandimarto, Thomas P. Cappola, and Anne R. Cappola
- Subjects
Heart transplantation ,medicine.medical_specialty ,endocrine system ,endocrine system diseases ,business.industry ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Atrial fibrillation ,030204 cardiovascular system & hematology ,medicine.disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Thyroid dysfunction ,Internal medicine ,Heart failure ,medicine ,Cardiology ,In patient ,Cardiology and Cardiovascular Medicine ,business ,Prospective cohort study ,Cardiovascular outcomes ,hormones, hormone substitutes, and hormone antagonists ,Euthyroid sick syndrome - Abstract
Background: The effects of thyroid dysfunction in patients with preexisting heart failure have not been adequately studied. We examined the prevalence of thyroid dysfunction and associations with cardiovascular outcomes in a large, prospective cohort of outpatients with preexisting heart failure. Methods and Results: We examined associations between thyroid dysfunction and New York Heart Association class, atrial fibrillation, and a composite end point of ventricular assist device placement, heart transplantation, or death in 1365 participants with heart failure enrolled in the Penn Heart Failure Study. Mean age was 57 years, 35% were women, and the majority had New York Heart Association class II (45%) or III (32%) symptoms. More severe heart failure was associated with higher thyroid-stimulating hormone (TSH), higher free thyroxine (FT4), and lower total triiodothyronine (TT3) concentrations ( P P ≤0.01 all models). There were 462 composite end points over a median 4.2 years of follow-up. In adjusted models, compared with euthyroidism, subclinical hypothyroidism (TSH 4.51–19.99 mIU/L with normal FT4) was associated with an increased risk of the composite end point overall (hazard ratio, 1.82; 95% CI, 1.27–2.61; P =0.001) and in the subgroup with TSH ≥7.00 mIU/L (hazard ratio, 3.25; 95% CI, 1.96–5.39; P P =0.34). Isolated low T3 was also associated with the composite end point (hazard ratio, 2.12; 95% CI, 1.65–2.72; P Conclusions: In patients with preexisting heart failure, subclinical hypothyroidism with TSH ≥7 mIU/L and isolated low T3 levels are associated with poor prognosis. Clinical trials are needed to explore therapeutic effects of T4 and T3 administration in heart failure.
- Published
- 2018
30. From statistical significance to clinical relevance: A simple algorithm to integrate brain natriuretic peptide and the Seattle Heart Failure Model for risk stratification in heart failure
- Author
-
Nancy K. Sweitzer, Wayne C. Levy, Benjamin French, Allan S. Jaffe, James C. Fang, Sultan A. Mirzoyev, Omar F. AbouEzzeddine, Margaret M. Redfield, and Thomas P. Cappola
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Pediatrics ,medicine.medical_treatment ,030204 cardiovascular system & hematology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Natriuretic Peptide, Brain ,medicine ,Humans ,Clinical significance ,Prospective Studies ,030212 general & internal medicine ,Prospective cohort study ,Retrospective Studies ,Heart Failure ,Transplantation ,Ejection fraction ,business.industry ,Retrospective cohort study ,Prognosis ,Brain natriuretic peptide ,medicine.disease ,Ventricular assist device ,Heart failure ,Cardiology ,Surgery ,Cardiology and Cardiovascular Medicine ,business ,Algorithms - Abstract
Background Heart failure (HF) guidelines recommend brain natriuretic peptide (BNP) and multivariable risk scores, such as the Seattle Heart Failure Model (SHFM), to predict risk in HF with reduced ejection fraction (HFrEF). A practical way to integrate information from these 2 prognostic tools is lacking. We sought to establish a SHFM+BNP risk-stratification algorithm. Methods The retrospective derivation cohort included consecutive patients with HFrEF at the Mayo Clinic. One-year outcome (death, transplantation or ventricular assist device) was assessed. The SHFM+BNP algorithm was derived by stratifying patients within SHFM-predicted risk categories (≤2.5%, 2.6% to ≤10%, >10%) according to BNP above or below 700 pg/ml and comparing SHFM-predicted and observed event rates within each SHFM+BNP category. The algorithm was validated in a prospective, multicenter HFrEF registry (Penn HF Study). Results Derivation ( n = 441; 1-year event rate 17%) and validation ( n = 1,513; 1-year event rate 12%) cohorts differed with the former being older and more likely ischemic with worse symptoms, lower EF, worse renal function and higher BNP and SHFM scores. In both cohorts, across the 3 SHFM-predicted risk strata, a BNP >700 pg/ml consistently identified patients with approximately 3-fold the risk that the SHFM would have otherwise estimated, regardless of stage of HF, intensity and duration of HF therapy and comorbidities. Conversely, the SHFM was appropriately calibrated in patients with a BNP Conclusion The simple SHFM+BNP algorithm displays stable performance across diverse HFrEF cohorts and may enhance risk stratification to enable appropriate decision-making regarding HF therapeutic or palliative strategies.
- Published
- 2016
31. Prognostic Value of Galectin-3 for Adverse Outcomes in Chronic Heart Failure
- Author
-
Le Wang, Bonnie Ky, Anupam Basuray, Nancy K. Sweitzer, Thomas P. Cappola, Benjamin French, James C. Fang, and Jeffrey Brandimarto
- Subjects
Male ,medicine.medical_specialty ,Galectin 3 ,Galectins ,medicine.medical_treatment ,030204 cardiovascular system & hematology ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Troponin I ,medicine ,Humans ,Prospective Studies ,Mortality ,Aged ,Heart Failure ,Ejection fraction ,business.industry ,Proportional hazards model ,Hazard ratio ,Stroke Volume ,Blood Proteins ,Stroke volume ,Middle Aged ,Prognosis ,medicine.disease ,Transplantation ,Treatment Outcome ,Heart failure ,Ventricular assist device ,Chronic Disease ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business ,Biomarkers ,030217 neurology & neurosurgery ,Follow-Up Studies - Abstract
Clinical studies have suggested the prognostic value of galectin-3, a marker of fibrosis, in chronic heart failure. However, the specific role of galectin-3, compared with established biomarkers, remains uncertain.The Penn Heart Failure Study was an ambulatory heart failure cohort that included 1385 participants with reduced (1141), preserved (106), and recovered (138) left ventricular ejection fraction (LVEF). Cox regression models determined the association between galectin-3 and risk of all-cause mortality, cardiac transplantation, or placement of a ventricular assist device. Receiver operating characteristic curves compared the prognostic accuracy of galectin-3, high-sensitivity soluble Toll-like receptor 2 (ST2), troponin I, and B-type natriuretic peptide (BNP) at 1 and 5 years. Higher galectin-3 levels were associated with an increased risk of adverse events (adjusted hazard ratio of 1.96 for each doubling in galectin-3; P .001). This association was most pronounced among participants with preserved LVEF (adjusted hazard ratio 3.30; P .001). At 5 years, galectin-3 was the most accurate discriminator of risk among participants with preserved LVEF (area under the curve 0.782; P = .81 vs high-sensitivity ST2; P = .029 vs troponin I; P = .35 vs BNP). BNP was most accurate among participants with reduced and recovered LVEF (areas under the curves 0.716 and 0.728, respectively).Galectin-3 could have prognostic value for long-term events among patients with heart failure and preserved ejection fraction.
- Published
- 2016
32. A pharmacogenetic investigation of intravenous furosemide in decompensated heart failure: a meta-analysis of three clinical trials
- Author
-
Ian Mongrain, Sylvie Provost, Yassamin Feroz Zada, Julianna Keleti, Douglas L. Mann, Thomas P. Cappola, S. de Denus, M-P Dubé, Jean-Lucien Rouleau, A Bardhadi, Svati H. Shah, Michael S. Phillips, Gordon S. Huggins, and Valérie Normand
- Subjects
medicine.medical_specialty ,Apolipoprotein L1 ,medicine.medical_treatment ,030204 cardiovascular system & hematology ,Pharmacology ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Internal medicine ,Genetics ,medicine ,030212 general & internal medicine ,biology ,business.industry ,Weight change ,Furosemide ,medicine.disease ,Regimen ,Heart failure ,biology.protein ,Cardiology ,Molecular Medicine ,Diuretic ,business ,Pharmacogenetics ,medicine.drug - Abstract
We conducted a meta-analysis of pharmacogenomic substudies of three randomized trials conducted in patients with decompensated heart failure (HF) that were led by National Heart Lung and Blood Institute (NHLBI)-funded HF Network to test the hypothesis that candidate genes modulate net fluid loss and weight change in patients with decompensated HF treated with a furosemide-based diuretic regimen. Although none of the genetic variants previously shown to modulate the effects of loop diuretics in healthy individuals were associated with net fluid loss after 72 h of treatment, a set of rare variants in the APOL1 gene, which codes for apolipoprotein L1 (P=0.0005 in the random effects model), was associated with this end point. Moreover, a common variant in the multidrug resistance protein-4 coding gene (ABCC4, rs17268282) was associated with weight loss with furosemide use (P=0.0001). Our results suggest that both common and rare genetic variants modulate the response to a furosemide-based diuretic regimen in patients with decompensated HF.
- Published
- 2016
33. Systems Genomics Identifies a Key Role for Hypocretin/Orexin Receptor-2 in Human Heart Failure
- Author
-
Roda P. Konadhode, Jing Liu, Heidi Salisbury, Jeffrey Brandimarto, Matthew T. Wheeler, Euan A. Ashley, Aleksandra Pavlovic, Rupak Mukherjee, Porama Koy Thanaporn, Ching Shang, Devin Absher, Thomas E. Scammell, Daniel Sedehi, Thomas P. Cappola, Thomas Quertermous, Marco V Perez, Stephen Pan, Clint L. Miller, Frederick E. Dewey, Richard M. Myers, and Khin Chan
- Subjects
Cardiac function curve ,Adult ,Male ,Candidate gene ,medicine.medical_specialty ,Genome-wide association study ,Article ,Cohort Studies ,Hypocretin (orexin) receptor 2 ,Mice ,Orexin Receptors ,Internal medicine ,medicine ,Animals ,Humans ,Aged ,Genetics ,Heart Failure ,Ejection fraction ,business.industry ,Stroke Volume ,Middle Aged ,medicine.disease ,Orexin receptor ,Minor allele frequency ,Disease Models, Animal ,Heart failure ,Cardiology ,Female ,business ,Cardiology and Cardiovascular Medicine ,Genome-Wide Association Study - Abstract
Background The genetic determinants of heart failure (HF) and response to medical therapy remain unknown. We hypothesized that identifying genetic variants of HF that associate with response to medical therapy would elucidate the genetic basis of cardiac function. Objectives This study sought to identify genetic variations associated with response to HF therapy. Methods This study compared extremes of response to medical therapy in 866 HF patients using a genome-wide approach that informed the systems-based design of a customized single nucleotide variant array. The effect of genotype on gene expression was measured using allele-specific luciferase reporter assays. Candidate gene transcription-deficient mice underwent echocardiography and treadmill exercise. The ability of the target gene agonist to rescue mice from chemically-induced HF was assessed with echocardiography. Results Of 866 HF patients, 136 had an ejection fraction improvement of 20% attributed to resynchronization (n = 83), revascularization (n = 7), tachycardia resolution (n = 2), alcohol cessation (n = 1), or medications (n = 43). Those with the minor allele for rs7767652, upstream of hypocretin (orexin) receptor-2 (HCRTR2), were less likely to have improved left ventricular function (odds ratio: 0.40 per minor allele; p = 3.29 × 10−5). In a replication cohort of 798 patients, those with a minor allele for rs7767652 had a lower prevalence of ejection fraction >35% (odds ratio: 0.769 per minor allele; p = 0.021). In an HF model, HCRTR2-deficient mice exhibited poorer cardiac function, worse treadmill exercise capacity, and greater myocardial scarring. Orexin, an HCRTR2 agonist, rescued function in this HF mouse model. Conclusions A systems approach identified a novel genetic contribution to human HF and a promising therapeutic agent efficacious in an HF model.
- Published
- 2015
- Full Text
- View/download PDF
34. Genomic Risk Stratification Predicts All-Cause Mortality After Cardiac Catheterization
- Author
-
Daniel J. Rader, Marie A Guerraty, David Birtwell, Heather Williams, Jay Giri, Thomas P. Cappola, Zolt Arany, Michael G. Levin, Scott M. Damrauer, Rachel L. Kember, Renae Judy, and Jinbo Chen
- Subjects
Male ,0301 basic medicine ,Cardiac Catheterization ,medicine.medical_specialty ,medicine.medical_treatment ,Myocardial Infarction ,Coronary Artery Disease ,030204 cardiovascular system & hematology ,Coronary Angiography ,Risk Assessment ,Article ,Disease-Free Survival ,Cohort Studies ,Coronary artery disease ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,Genetic variation ,Humans ,Medicine ,Myocardial infarction ,Aged ,Cardiac catheterization ,Aged, 80 and over ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Human genetics ,Survival Rate ,030104 developmental biology ,Risk stratification ,Cardiology ,Female ,Polygenic risk score ,business ,All cause mortality - Abstract
Background: Coronary artery disease (CAD) is influenced by genetic variation and traditional risk factors. Polygenic risk scores (PRS), which can be ascertained before the development of traditional risk factors, have been shown to identify individuals at elevated risk of CAD. Here, we demonstrate that a genome-wide PRS for CAD predicts all-cause mortality after accounting for not only traditional cardiovascular risk factors but also angiographic CAD itself. Methods: Individuals who underwent coronary angiography and were enrolled in an institutional biobank were included; those with prior myocardial infarction or heart transplant were excluded. Using a pruning-and-thresholding approach, a genome-wide PRS comprised of 139 239 variants was calculated for 1503 participants who underwent coronary angiography and genotyping. Individuals were categorized into high PRS (hiPRS) and low-PRS control groups using the maximally selected rank statistic. Stratified analysis based on angiographic findings was also performed. The primary outcome was all-cause mortality following the index coronary angiogram. Results: Individuals with hiPRS were younger than controls (66 years versus 69 years; P =2.1×10 -5 ) but did not differ by sex, body mass index, or traditional risk-factor profiles. Individuals with hiPRS were at significantly increased risk of all-cause mortality after cardiac catheterization, adjusting for traditional risk factors and angiographic extent of CAD (hazard ratio, 1.6; 95% CI, 1.2–2.2; P =0.004). The strongest increase in risk of all-cause mortality conferred by hiPRS was seen among individuals without angiographic CAD (hazard ratio, 2.4; 95% CI, 1.1–5.5; P =0.04). In the overall cohort, adding hiPRS to traditional risk assessment improved prediction of 5-year all-cause mortality (area under the receiver-operating curve 0.70; 95% CI, 0.66–0.75 versus 0.66; 95% CI, 0.61–0.70; P =0.001). Conclusions: A genome-wide PRS improves risk stratification when added to traditional risk factors and coronary angiography. Individuals without angiographic CAD but with hiPRS remain at significantly elevated risk of mortality.
- Published
- 2018
35. Genome-Wide Associations of Global Electrical Heterogeneity ECG Phenotype: The ARIC (Atherosclerosis Risk in Communities) Study and CHS (Cardiovascular Health Study)
- Author
-
Colleen M. Sitlani, Tor Biering-Sørensen, Wendy S. Post, Larisa G. Tereshchenko, Mary L. Biggs, Scott D. Solomon, Dan E. Arking, Michael Morley, Elsayed Z. Soliman, Muammar M. Kabir, Nona Sotoodehnia, David S. Siscovick, Alfred E. Buxton, Foram N. Ashar, Jonathan W. Waks, and Thomas P. Cappola
- Subjects
0301 basic medicine ,Male ,Genome-wide association study ,sum absolute QRST integral ,030204 cardiovascular system & hematology ,Sudden cardiac death ,spatial ventricular gradient ,0302 clinical medicine ,Risk Factors ,Arrhythmia and Electrophysiology ,Prospective Studies ,genome wide association study ,Original Research ,2. Zero hunger ,medicine.diagnostic_test ,Incidence ,Middle Aged ,Phenotype ,Survival Rate ,Electrophysiology ,medicine.anatomical_structure ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,medicine.medical_specialty ,global electrical heterogeneity ,electrocardiography ,Disease cluster ,Risk Assessment ,03 medical and health sciences ,Genetic, Association Studies ,Internal medicine ,Linear regression ,medicine ,Humans ,Electrocardiology (ECG) ,business.industry ,ECG ,medicine.disease ,Atherosclerosis ,United States ,030104 developmental biology ,Death, Sudden, Cardiac ,Ventricle ,Genetic Loci ,business ,Body mass index ,Electrocardiography ,Follow-Up Studies ,Genome-Wide Association Study - Abstract
Background ECG global electrical heterogeneity ( GEH ) is associated with sudden cardiac death. We hypothesized that a genome‐wide association study would identify genetic loci related to GEH . Methods and Results We tested genotyped and imputed variants in black (N=3057) and white (N=10 769) participants in the ARIC (Atherosclerosis Risk in Communities) study and CHS (Cardiovascular Health Study). GEH ( QRS ‐T angle, sum absolute QRST integral, spatial ventricular gradient magnitude, elevation, azimuth) was measured on 12‐lead ECG s. Linear regression models were constructed with each GEH variable as an outcome, adjusted for age, sex, height, body mass index, study site, and principal components to account for ancestry. GWAS identified 10 loci that showed genome‐wide significant association with GEH in whites or joint ancestry. The strongest signal (rs7301677, near TBX 3 ) was associated with QRS ‐T angle (white standardized β+0.16 [95% CI 0.13–0.19]; P =1.5×10 −26 ), spatial ventricular gradient elevation (+0.11 [0.08–0.14]; P =2.1×10 −12 ), and spatial ventricular gradient magnitude (−0.12 [95% CI −0.15 to −0.09]; P =5.9×10 −15 ). Altogether, GEH ‐ SNP s explained 1.1% to 1.6% of GEH variance. Loci on chromosomes 4 (near HMCN 2 ), 5 ( IGF 1R ), 11 (11p11.2 region cluster), and 7 (near ACTB ) are novel ECG phenotype‐associated loci. Several loci significantly associated with gene expression in the left ventricle ( HMCN 2 locus—with HMCN 2 ; IGF 1R locus—with IGF 1R ), and atria ( RP 11‐481J2.2 locus—with expression of a long non‐coding RNA and NDRG 4 ). Conclusions We identified 10 genetic loci associated with ECG GEH . Replication of GEH GWAS findings in independent cohorts is warranted. Further studies of GEH ‐loci may uncover mechanisms of arrhythmogenic remodeling in response to cardiovascular risk factors.
- Published
- 2018
36. Effects of Xanthine Oxidase Inhibition in Hyperuricemic Heart Failure Patients
- Author
-
Douglas L. Mann, Haissam Haddad, W.H. Wilson Tang, Monica R. Shah, Kevin J. Anstrom, Martin M. LeWinter, Mitchell T. Saltzberg, Elizabeth Ofili, Steven McNulty, Adrian F. Hernandez, Anita Deswal, Margaret M. Redfield, Javed Butler, Thomas P. Cappola, Steven R. Goldsmith, Kerry L. Lee, Michael M. Givertz, Marc J. Semigran, Christopher M. O'Connor, Mark E. Dunlap, G. Michael Felker, Kenneth B. Margulies, and Marvin A. Konstam
- Subjects
medicine.medical_specialty ,Ejection fraction ,business.industry ,Allopurinol ,medicine.disease ,Placebo ,Clinical trial ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Physiology (medical) ,Heart failure ,Internal medicine ,Multicenter trial ,medicine ,Cardiology ,Uric acid ,Cardiology and Cardiovascular Medicine ,Xanthine oxidase ,business ,medicine.drug - Abstract
Background— Oxidative stress may contribute to heart failure (HF) progression. Inhibiting xanthine oxidase in hyperuricemic HF patients may improve outcomes. Methods and Results— We randomly assigned 253 patients with symptomatic HF, left ventricular ejection fraction ≤40%, and serum uric acid levels ≥9.5 mg/dL to receive allopurinol (target dose, 600 mg daily) or placebo in a double-blind, multicenter trial. The primary composite end point at 24 weeks was based on survival, worsening HF, and patient global assessment. Secondary end points included change in quality of life, submaximal exercise capacity, and left ventricular ejection fraction. Uric acid levels were significantly reduced with allopurinol in comparison with placebo (treatment difference, –4.2 [–4.9, –3.5] mg/dL and –3.5 [–4.2, –2.7] mg/dL at 12 and 24 weeks, respectively, both P P =0.68). At 12 and 24 weeks, there was no significant difference in change in Kansas City Cardiomyopathy Questionnaire scores or 6-minute walk distances between the 2 groups. At 24 weeks, left ventricular ejection fraction did not change in either group or between groups. Rash occurred more frequently with allopurinol (10% versus 2%, P =0.01), but there was no difference in serious adverse event rates between the groups (20% versus 15%, P =0.36). Conclusions— In high-risk HF patients with reduced ejection fraction and elevated uric acid levels, xanthine oxidase inhibition with allopurinol failed to improve clinical status, exercise capacity, quality of life, or left ventricular ejection fraction at 24 weeks. Clinical Trial Registration— URL: http://www.clinicaltrials.gov . Unique identifier: NCT00987415.
- Published
- 2015
37. Quality of Anticoagulation Control in Preventing Adverse Events in Patients With Heart Failure in Sinus Rhythm
- Author
-
S. Shaw, Conrado J. Estol, Biykem Bozkurt, B. Donelly, R. Berkowitz, L. Berente, M. Jurczok, J. Simon, Sami Khella, A. Travis, M. Schoenauer, Ronald S. Freudenberger, Anikó Ilona Nagy, G. Marcinek, M. Hajkova, T. Siebert, R. Breedveld, W. Baker, P. Avellana, S. Kozhukhov, M. Diek, J. Jaros, R. Vicari, S. Timar, C. Schanz, J. Povolny, M. van Zagten, Shelley Zieroth, G. Rex, M. Apelian, M. Dzaiy, K. Hayward, A. Warner, L. Baliko, Piotr Ponikowski, S. Mathus, Arthur J. Labovitz, C. DeMers, E. Péterfai, Mariana Bolos, B. Stephens, F. Tito, Denise L. Janosik, N. Molakala, J. Dizes, R. van der Loo, A. van Bujisen-Nutters, K. Wolf, S. Nawaz, A. Boguschewski, D. Ferguson, A. McPhail, M. Rohwedder, M. Malkowski, R. Prodhan, P. Miekus, R. Pellegrino, M. Rossi, Michelle Bierig, E. Wirkowski, Zbigniew Gaciong, F. Guerlloy, D. Chupka, O. Kovtun, M. Padour, D. Horak, Elly M.C.J. Wajon, J. Carda, P. Gregor, M. Tilem, P. Kosolcharoen, R. Wensel, V. Garman, N. Torok, B. Benczur, E. Peña, G. Moe, B.M. Massie, S. Slomiak, C. Benesch, V. Orlyk, K. Greenan, M. Ogorek, I. Jedlinski, Shun Kohsaka, R. Haynes, Z. Lorenc, M. Kuchar, R. Pauer, T. Dugan, A. Juszczak, T. Schrader, A. Henriquez, K. Toth, M. Varga, J. Herman, M. Lee, M. Dunaj, L. Krizova, M. Bonora, P. Loviska, D. Aubin, M. Kokles, J. D. Easton, J. Meschia, A. Bruno, Steven Goldman, S. Voigt, D. Malchik, A. Tierney, Demetrios J. Sahlas, N. Elzebroek, Richard J. Hobbs, M. Charlet, B. Hattler, L. Regos, Dennis Wolf, A. Peljto, C. Lindsey, R. Winkler, J. Arredondo, B. Oze, C. Schuler, D. Bruck, E. Jones, G. Torre, S. Nabhan, G. Mallis, Jindřich Špinar, W. Graettinger, A. Ruiz, N. Holwerda, R. Katz, J. Gonzalez, Christine Gerula, P. Jansky, H. V. Anderson, B. Krizova, Andreas König, M. Moussavian, M. Konarzewski, G. Jakab, M. Liston, Elizabeth O. Ofili, L. Nikolaidis, Susan E. Ammon, Rebekka K. Schneider, M. Krauze-Wielicka, M. Zimmermann, J. Lynch, J. Minuk, F. Sokn, Douglas L. Mann, R. Sawyer, J. Partridge, D. Leifer, M. Bohdanowicz-Zazula, D. Barratt, C. Dewar, S. Kolomiets, T. Noonan, D. Beran, M. Jeserich, J. Wong, R. Bessoudo, M. Lichtenberger, E. Bednarova, R. Serafin, M. Scullin, I. Kosa, M. Hranai, E. Duverger, L. Joseph, A. M. Sindilar, I. Edes, E. Yakimenko, J. Hobbs-Williams, F. Novoa, A. Szczepanska, R. Wachter, P. A. de Milliano, L. Golan, Gregory W. Albers, K. Ammerman, Alexandra R. Sanford, W. Burgin, A. Richmond, A. Kleinrok, B. Dandapani, Laurie Gutmann, M. Houra, Udho Thadani, P. Schygiel, A. van Hessen, Christian Weber, S. Mehešová, P. Stein-Beal, E. Flanagan, R. Khadouri, P. Chang, Thomas P. Cappola, L. Konczarek, B. Mangariello, L. Atkins, C. McKay, J. Svoboda, O. Lesniak, L. Westbrook, S. J. Kim, C. Moy, M. Kuch, M. Nemec, M. Krobot, B. Kozlowski, M. Applegate, M. Kiorwantsi, J. Thierer, K. Craig, A. Slim, Y. Besaga, R. Kuba, P. Fulop, K. Crotto, R. Porcile, E. Falgout, E. Olgren, K. Kuc, Marcus F. Stoddard, W. Almeida, L. Pas, L. Williams, I. Sorokina, J. Rodl, S. Sparr, A. Coppes, E. Ronner, A. Jurczyk, S. Gass, John R. Teerlink, P. Kucera, H. J. Barnett, B. McGinnis, L. Wilson, Y. Tutov, R. MacFadyen, Gregory Y.H. Lip, V. Sorrell, T. Ochalek, J. Turner, M. Modzelewski, Matthew Wilson, A. Ogorodnichuk, C. Anderson, P. de Kort, B. Palossy, Alan B. Miller, T. Giles, H. Brown, Andrew H. Baker, P. Czaja, Roman Szełemej, J. Hanna, P. Gilbert, A. Metcalf, R. Piotrowski, D. Yip, B. Coull, P. Gitelman, Burkert Pieske, C. Rapallo, M. Morgil, M. Resch, A. Zachar, Jan Biegus, W. Watson, J. A. Hinchey, E. Polland, L. Caufield, D. Kopcik, E. Pechackova, M. Calderon, Stefan D. Anker, V. Virkud, R. Rothbart, L. Rudenko, W. Wicha, B. Jacques, Susan Graham, J. Donaldson, O. Girina, L. Guillory, S. Khoury, I. Padourova, Haissam Haddad, L. Kowalczyk, J. A. Swain, G. Prokop-Lewicka, R. Mattessich, K. Remmel, S. Tikhonova, S. Klochkov, J. Leppo, K. C. Johnston, D. Gohs, Peter K. Smith, Eric E. Smith, V. Hart, J. Vanyi, L. Voronkov, G. Allam, M. Klapholz, T. Varadyova, S. Daugherty, L. Witkin, K. Panizzon, B. Drachman, S. Locke, Ann‐Katrin Mojica Munoz, J. Love, T. Winder, P. Bailey, T. Huynh, G. A. Verheul, Tomasz J. Pasierski, J. Borbola, R. Liu, A. Elizalde, C. Walker, R. Kelley, Robert Côté, P. Frey, J. McGee, Peter E. Carson, T. Bator, Donald L. Patrick, K. Karsay, J. Plomp, O. Novikova, J. Vuijsters, Jay P. Mohr, A. Parkhomenko, A. Ducharme, C. Alteri, S. Borden, Siqin Ye, W. Felton, K. Peterson, M. Satori, N. Polenova, D. Karia, G. Turhan, R. Nagy, K. Amosova, J. Michalska, R. Libman, E. Frey, O. Najmanova, R. Yufe, O. Montaña, S. Bailey, M. Bodi, A. Ellis, J. Tarchalski, L. Sitwell, M. Del Valle, John P. Boehmer, J. Marler, P. Romia, K. Tea, E. Hartmann, R. Lebedova, V. Yurlov, O. Karpenko, Malcolm Arnold, P. Berkowski, J. Johnson, P. Ramappa, R. Ferkl, Dirk J. Lok, N. Bayer, S. Bezucha, A. Mercando, H. Tworek, R. Longaker, P. Jinkins, J. Kirmani, L. Svoboda, David Spence, Min Qian, John L.P. Thompson, N. Brodi, Y. Prokopovych, Sedat Sen, M. Nanna, S. T. Palmeri, M. Michalova, L. Giron, K. Wolkowska, D. Borts, K. Hamroui, G. Linssen, L. Arcement, A. McNulty, C. Jakobs, F. Bleyer, W. Lo, J. Bisognano, J. Kosits, Steven R. Levine, G. Berry, P. Heidrich, G. Kiss, G. Tullio, J. Yasen, G. Ortiz, B. O'Hare, P. Jackson, T. Rennie, Z. Davidovits, G. v Buchem-Damming, H. Dvorakova, W. Hermans, Zbigniew Kalarus, H. Morgil, C. Harris, M. Vissiennon, Shunichi Homma, André P. Gabriel, J. Aiub, I. Katzan, C. Zaidman, S. Sassone, A. Duszanska, J. Litvinova, P. Kralicek, M. Natour, E. Nagy, E. Nishime, M. J. Bos, S. Nowakowska, J. Beebe, B. Watson, N. Jacek, Ralph L. Sacco, A. Cwynar, S. Pezzella, B. George, B. Hott, T. Vegh, V. Mejia, E. Janzen, M. Eliasziw, Heribert Schunkert, L. Swydan, J. Gora, D. Drazek, C. Landau, L. Roffidal, L. Casazza, Andrew M Penn, Richard L. Hughes, V. Schumann, R. Santi, I. Sakharchuk, A. Adler, D. Taylor, C. F. Peerenboom-Fey, M. Dluzniewski, L. Cape, Attila Kovacs, E. Ziekenhuis, A. Bujdoso, L. Fischer, Richard Buchsbaum, Petr Arenberger, L. Pollak, J. Vosmerova, B. Donley, Patrick M. Pullicino, B. Darrow, A. Minagar, N. Jarmukli, J. Dissin, M. Daniels, L. Csuros, G. J. del Zoppo, E. Anthony, B. Metzkier-Wyrwa, A. Ronaszeki, I. Malek, R. Arbing, Gilberto Levy, D. Mauceri, Carlos J. Rodriguez, V. Kovalenko, J. Smith, O. Yaremenko, R. de Graaf Gasthuis, C. Donato, L. Spinarova, Martin M. Brown, F. Padour, O. Lovasz, Marco R. Di Tullio, K. Balaban, S. Donovan, S. Genth-Zotz, M. Maruskova, I. Varga, T. Drasnar, V. Berchou, R. Davies, Robert G. Hart, M. Lebedynska, G. Hageman, D. Disantis, W. Schneider, M. Frankel, A. Hajnalne, S. Baumann, P. Karpati, L. C. Pettigrew, Johnston Grier, A. Ellenberg, Bruce Levin, and Stephanie Hope Dunlap
- Subjects
Intracerebral hemorrhage ,medicine.medical_specialty ,Aspirin ,Ejection fraction ,business.industry ,Warfarin ,030204 cardiovascular system & hematology ,medicine.disease ,3. Good health ,Surgery ,03 medical and health sciences ,0302 clinical medicine ,Interquartile range ,Heart failure ,Internal medicine ,medicine ,Cardiology ,Sinus rhythm ,030212 general & internal medicine ,Cardiology and Cardiovascular Medicine ,business ,Stroke ,medicine.drug - Abstract
Background— The aim of this study is to examine the relationship between time in the therapeutic range (TTR) and clinical outcomes in heart failure patients in sinus rhythm treated with warfarin. Methods and Results— We used data from the Warfarin versus Aspirin in Reduced Cardiac Ejection Fraction (WARCEF) trial to assess the relationship of TTR with the WARCEF primary outcome (ischemic stroke, intracerebral hemorrhage, or death), with death alone, ischemic stroke alone, major hemorrhage alone, and net clinical benefit (primary outcome and major hemorrhage combined). Multivariable Cox models were used to examine how the event risk changed with TTR and to compare the high TTR, low TTR, and aspirin-treated patients, with TTR being treated as a time-dependent covariate. A total of 2217 patients were included in the analyses; among whom 1067 were randomized to warfarin and 1150 were randomized to aspirin. The median (interquartile range) follow-up duration was 3.6 (2.0–5.0) years. Mean (±SD) age was 61±11.3 years, with 80% being men. The mean (±SD) TTR was 57% (±28.5%). Increasing TTR was significantly associated with reduction in primary outcome (adjusted P P =0.001), and improved net clinical benefit (adjusted P P =0.082 for ischemic stroke and adjusted P =0.109 for major hemorrhage). Conclusions— In patients with heart failure in sinus rhythm, increasing TTR is associated with better outcome and improved net clinical benefit. Patients in whom good quality anticoagulation can be achieved may benefit from the use of anticoagulants. Clinical Trial Registration— URL: http://www.clinicaltrials.gov . Unique identifier: NCT00041938.
- Published
- 2015
38. Effect of Inorganic Nitrate on Exercise Capacity in Heart Failure With Preserved Ejection Fraction
- Author
-
Kenneth B. Margulies, David C. Poole, Harry Ischiropoulos, Deepa Rawat, Julio A. Chirinos, Paschalis-Thomas Doulias, Raymond R. Townsend, Prithvi Shiva-Kumar, Prasad Konda, Rushik Bhuva, Salvatore Geraci, Thomas P. Cappola, and Payman Zamani
- Subjects
medicine.medical_specialty ,Cardiac output ,business.industry ,Vasodilation ,Stroke volume ,Oxygenation ,medicine.disease ,Nitric oxide ,Surgery ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Physiology (medical) ,Internal medicine ,Heart failure ,medicine ,Vascular resistance ,Cardiology ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,business - Abstract
Background— Inorganic nitrate (NO 3 − ), abundant in certain vegetables, is converted to nitrite by bacteria in the oral cavity. Nitrite can be converted to nitric oxide in the setting of hypoxia. We tested the hypothesis that NO 3 − supplementation improves exercise capacity in heart failure with preserved ejection fraction via specific adaptations to exercise. Methods and Results— Seventeen subjects participated in this randomized, double-blind, crossover study comparing a single dose of NO 3 -rich beetroot juice (NO 3 − , 12.9 mmol) with an identical nitrate-depleted placebo. Subjects performed supine-cycle maximal-effort cardiopulmonary exercise tests, with measurements of cardiac output and skeletal muscle oxygenation. We also assessed skeletal muscle oxidative function. Study end points included exercise efficiency (total work/total oxygen consumed), peak o 2 , total work performed, vasodilatory reserve, forearm mitochondrial oxidative function, and augmentation index (a marker of arterial wave reflections, measured via radial arterial tonometry). Supplementation increased plasma nitric oxide metabolites (median, 326 versus 10 μmol/L; P =0.0003), peak o 2 (12.6±3.7 versus 11.6±3.1 mL O 2 ·min −1 ·kg −1 ; P =0.005), and total work performed (55.6±35.3 versus 49.2±28.9 kJ; P =0.04). However, efficiency was unchanged. NO 3 − led to greater reductions in systemic vascular resistance (−42.4±16.6% versus −31.8±20.3%; P =0.03) and increases in cardiac output (121.2±59.9% versus 88.7±53.3%; P =0.006) with exercise. NO 3 − reduced aortic augmentation index (132.2±16.7% versus 141.4±21.9%; P =0.03) and tended to improve mitochondrial oxidative function. Conclusions— NO 3 − increased exercise capacity in heart failure with preserved ejection fraction by targeting peripheral abnormalities. Efficiency did not change as a result of parallel increases in total work and o 2. NO 3 − increased exercise vasodilatory and cardiac output reserves. NO 3 − also reduced arterial wave reflections, which are linked to left ventricular diastolic dysfunction and remodeling. Clinical Trial Registration— URL: www.clinicaltrials.gov . Unique identifier: NCT01919177.
- Published
- 2015
39. Effect of Renal Function on Prognosis in Chronic Heart Failure
- Author
-
Scott Hetzel, Brad C. Astor, Nancy K. Sweitzer, Andrew O. Kadlec, James C. Fang, Thomas P. Cappola, and Adrián Ignacio Löffler
- Subjects
Male ,medicine.medical_specialty ,Time Factors ,Renal function ,Risk Assessment ,Article ,Risk Factors ,Internal medicine ,medicine ,Humans ,Prospective Studies ,Renal Insufficiency ,Prospective cohort study ,Survival rate ,Heart Failure ,business.industry ,Incidence ,Incidence (epidemiology) ,Middle Aged ,Pennsylvania ,Prognosis ,medicine.disease ,Confidence interval ,Survival Rate ,Heart failure ,Cohort ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business ,Follow-Up Studies ,Glomerular Filtration Rate ,Cohort study - Abstract
Renal dysfunction (RD) is associated with increased mortality in heart failure (HF). The aim of this study was to identify whether worsened or improved renal function during mid-term follow-up is associated with worsened outcomes in patients with chronic HF. A total of 892 participants from a multicenter cohort study of chronic HF were followed over 3.1 ± 1.9 years of enrollment. Worsened and improved renal functions were tested with multivariate models as independent predictors of HF hospitalization and mortality. Although 12% of subjects experienced a ≥25% decrease in estimated glomerular filtration rate (eGFR), 17% experienced a ≥25% increase in eGFR, and there was stability of kidney function observed in the cohort as a whole. The quartile with the worst RD at any point in time had increased risk of HF hospitalization and mortality. Worsened eGFR was associated with HF outcomes in the unadjusted (hazard ratio = 1.71, 95% confidence interval 1.04 to 2.81, p = 0.035), but not the adjusted analysis. Improvement in eGFR was not associated with outcome (p = 0.453). In chronic HF, the severity of RD predicts risk of poor outcome better than changes in renal function during mid-term follow-up. This suggests that in patients with appropriately treated chronic HF, worsening renal function in itself does not yield useful prognostic information and may not reflect poor outcome.
- Published
- 2015
40. The Diastolic Pulmonary Gradient Does Not Predict Survival in Patients With Pulmonary Hypertension Due to Left Heart Disease
- Author
-
Ryan J. Tedford, Stuart D. Russell, Teresa A De Marco, Edward K. Kasper, Van N. Selby, Emmanouil Tampakakis, Michael Felker, Thomas P. Cappola, and Peter J. Leary
- Subjects
medicine.medical_specialty ,business.industry ,Diastole ,food and beverages ,medicine.disease ,Pulmonary hypertension ,Heart failure ,Internal medicine ,medicine ,Cardiology ,In patient ,Left heart disease ,Cardiology and Cardiovascular Medicine ,business - Abstract
Objectives: This study sought to evaluate if diastolic pulmonary gradient (DPG) can predict survival in patients with pulmonary hypertension due to left heart disease (PH-LHD).Background: P...
- Published
- 2015
41. Recreational Exercise in Hypertrophic Cardiomyopathy
- Author
-
Thomas P. Cappola and Anjali T. Owens
- Subjects
medicine.medical_specialty ,Exercise intolerance ,030204 cardiovascular system & hematology ,Left ventricular hypertrophy ,Sudden death ,Asymptomatic ,Sudden cardiac death ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Palpitations ,Humans ,030212 general & internal medicine ,Exercise ,biology ,business.industry ,Hypertrophic cardiomyopathy ,General Medicine ,Cardiomyopathy, Hypertrophic ,medicine.disease ,Troponin ,biology.protein ,Cardiology ,Exercise Test ,Recreation ,medicine.symptom ,business - Abstract
Hypertrophic cardiomyopathy, defined as unexplained left ventricular hypertrophy, has an estimated prevalence of at least 1 in 500 and is among the most common forms of inherited heart disease.1 The pathophysiology of hypertrophic cardiomyopathy is complex and involves the interplay between diastolic dysfunction, myocardial ischemia, systolic anterior motion of the mitral valve resulting in outflow tract obstruction, and arrhythmia.2 Clinical presentations are heterogeneous and range from asymptomatic to palpitations, dyspnea, exercise intolerance, chest pain, syncope, and sudden death. Although the absolute risk of cardiac mortality is less than 1% per year,3 hypertrophic cardiomyopathy remains a common cause of sudden cardiac death in the young, especially in competitive athletes.4,5 Since hypertrophic cardiomyopathy has autosomal dominant inheritance, relatives of affected individuals have substantial risk of disease. Clinical evaluation of family members and cascade screening with genetic testing can identify those affected, many of whom are asymptomatic. When properly treated, the majority of patients with hypertrophic cardiomyopathy will have a normal life span.3
- Published
- 2017
42. Biomarker Predictors of Cardiac Hospitalization in Chronic Heart Failure: A Recurrent Event Analysis
- Author
-
Thomas P. Cappola, Nancy K. Sweitzer, Esther Vorovich, Bonnie Ky, Lee R. Goldberg, Benjamin French, and James C. Fang
- Subjects
Male ,medicine.medical_specialty ,Exacerbation ,medicine.drug_class ,Article ,chemistry.chemical_compound ,Recurrence ,Internal medicine ,Natriuretic Peptide, Brain ,Troponin I ,Natriuretic peptide ,Humans ,Medicine ,Decompensation ,Intensive care medicine ,Heart Failure ,Creatinine ,Vascular Endothelial Growth Factor Receptor-1 ,biology ,business.industry ,Hazard ratio ,Middle Aged ,Prognosis ,medicine.disease ,Troponin ,United States ,Hospitalization ,chemistry ,Heart failure ,biology.protein ,Cardiology ,Female ,Morbidity ,Cardiology and Cardiovascular Medicine ,business ,Biomarkers ,Follow-Up Studies - Abstract
Background Identification of heart failure (HF) patients at risk for hospitalization may improve care and reduce costs. We evaluated 9 biomarkers as predictors of cardiac hospitalization in chronic HF. Methods and Results In a multicenter cohort of 1,512 chronic HF outpatients, we assessed the association between 9 biomarkers and cardiac hospitalization with the use of a recurrent events approach. Over a median follow-up of 4 years, 843 participants experienced ≥1 hospitalizations (total 2,178 hospitalizations). B-type natriuretic peptide (BNP) and troponin I (TnI) exhibited the strongest associations with risk of hospitalization (hazard ratio [HR] 3.8 [95% confidence interval (CI) 2.9-4.9] and HR 3.3 [95% CI 2.8-3.9]; 3rd vs 1st tertiles). Soluble Fms-like tyrosine kinase receptor 1 (sFlt-1) exhibited the next strongest association (HR 2.8 [95% CI 2.4–3.4]), followed by soluble Toll-like receptor 2 (HR 2.3 [95% CI 2.0–2.8]) and creatinine (HR 1.9 [95% CI 1.6–2.4]). Within ischemic/nonischemic subgroups, BNP and TnI remained most strongly associated. Except for creatinine, HRs for all biomarkers studied were smaller within the ischemic subgroup, suggesting greater importance of cardiorenal interactions in decompensation of ischemic HF. Conclusion Although BNP and TnI exhibited the strongest associations with hospitalization, etiology-dependent associations for the remaining biomarkers suggest etiology-specific mechanisms for HF exacerbation. sFlt-1 exhibited a strong association with cardiac hospitalization, highlighting its potential role as a biomarker of HF morbidity.
- Published
- 2014
43. SOLUBLE NEPRILYSIN IN PATIENTS WITH HEART FAILURE WITH PRESERVED EJECTION FRACTION
- Author
-
Melissa A. Lyle, Yogesh N.V. Reddy, Thomas P. Cappola, Naveen L. Pereira, Seethalakshmi R. Iyer, Steven McNulty, John R. Burnett, Margaret M. Redfield, Adrian F. Hernandez, Christopher G. Scott, and G. Michael Felker
- Subjects
medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,Cardiology ,In patient ,Cardiology and Cardiovascular Medicine ,Heart failure with preserved ejection fraction ,business ,Neprilysin - Published
- 2019
44. Associations of Conventional Echocardiographic Measures with Incident Heart Failure and Mortality: The Chronic Renal Insufficiency Cohort
- Author
-
Matthew J. Budoff, Radhakrishna R. Kallem, Ruth F. Dubin, Peter C. Yang, Jackson T. Wright, Jason Roy, Rajat Deo, Raymond R. Townsend, Martin G. Keane, Jiang He, Daohang Sha, Thomas P. Cappola, Amanda H. Anderson, Nisha Bansal, Shaista Malik, Mahboob Rahman, Michael G. Shlipak, Anna Porter, and Alan S. Go
- Subjects
Male ,medicine.medical_specialty ,Epidemiology ,Heart Ventricles ,030232 urology & nephrology ,Diastole ,030204 cardiovascular system & hematology ,Critical Care and Intensive Care Medicine ,Left ventricular hypertrophy ,03 medical and health sciences ,0302 clinical medicine ,Interquartile range ,Internal medicine ,Cause of Death ,medicine ,Humans ,Renal Insufficiency, Chronic ,Aged ,Heart Failure ,Transplantation ,Ejection fraction ,business.industry ,Incidence ,Hazard ratio ,Stroke Volume ,Original Articles ,Organ Size ,Middle Aged ,medicine.disease ,Confidence interval ,Quartile ,Nephrology ,Echocardiography ,Heart failure ,Cardiology ,Female ,Hypertrophy, Left Ventricular ,business ,Follow-Up Studies - Abstract
Background and objectives Heart failure is the most frequent cardiac complication of CKD. Left ventricular hypertrophy is common and develops early in CKD, but studies have not adequately evaluated the association of left ventricular mass index with heart failure incidence among men and women with CKD. Design, setting, participants, & measurements We evaluated echocardiograms of 2567 participants without self–reported heart failure enrolled in the Chronic Renal Insufficiency Cohort Study. Two-dimensional echocardiograms were performed at the year 1 study visit and interpreted at a central core laboratory. Left ventricular mass index was calculated using the linear method, indexed to height 2.7 , and analyzed using sex-specific quartiles. The primary outcomes of incident heart failure and all-cause mortality were adjudicated over a median of 6.6 (interquartile range, 5.7–7.6) years. Results Among 2567 participants, 45% were women, and 54% were nonwhite race; mean (SD) age was 59±11 years old, and mean eGFR was 44±17 ml/min per 1.73 m 2 . During a median follow-up period of 6.6 years, 262 participants developed heart failure, and 470 participants died. Compared with participants in the first quartile of left ventricular mass index, those in the highest quartile had higher rates of incident heart failure (hazard ratio, 3.96; 95% confidence interval, 1.96 to 8.02) and mortality (hazard ratio, 1.86; 95% confidence interval, 1.22 to 2.85), even after adjustment for B–type natriuretic peptide, troponin T, mineral metabolism markers, and other cardiovascular disease risk factors. Those in the lowest quartile of ejection fraction had higher rates of incident heart failure (hazard ratio, 3.01; 95% confidence interval, 1.94 to 4.67) but similar mortality rates (hazard ratio, 1.18; 95% confidence interval, 0.89 to 1.57) compared with those in the highest quartile. Diastolic dysfunction was not significantly associated with heart failure or death. Conclusions Among persons with CKD and without history of cardiovascular disease, left ventricular mass index is strongly associated with incident heart failure, even after adjustment for major cardiovascular risk factors and biomarkers.
- Published
- 2016
45. Heart Failure, Left Ventricular Remodeling, and Circulating Nitric Oxide Metabolites
- Author
-
Harry Ischiropoulos, Scott Akers, Payman Zamani, Maheswara R Koppula, Paschalis-Thomas Doulias, Haideliza Soto-Calderon, Kenneth B. Margulies, Julio A. Chirinos, Thomas P. Cappola, Ali Tariq, Amer Ahmed Syed, Raymond R. Townsend, Izzah Vasim, and Lien Trieu
- Subjects
Male ,Magnetic Resonance Imaging (MRI) ,030204 cardiovascular system & hematology ,chemistry.chemical_compound ,No bioavailability ,0302 clinical medicine ,Nitrate ,030212 general & internal medicine ,Prospective Studies ,Nitrite ,Original Research ,2. Zero hunger ,hypertrophy/remodeling ,Ventricular Remodeling ,Diastolic heart failure ,Heart ,Organ Size ,Middle Aged ,Magnetic Resonance Imaging ,humanities ,3. Good health ,United States Department of Veterans Affairs ,Cardiology ,Female ,Hypertrophy, Left Ventricular ,Cardiology and Cardiovascular Medicine ,medicine.medical_specialty ,Nitric Oxide ,Nitric oxide ,03 medical and health sciences ,myocardial structure ,Internal medicine ,medicine ,Humans ,Ventricular remodeling ,Aged ,Heart Failure ,business.industry ,diastolic heart failure ,Myocardium ,Correction ,Stroke Volume ,medicine.disease ,Fibrosis ,United States ,Endocrinology ,chemistry ,Heart failure ,Case-Control Studies ,Myocardial fibrosis ,myocardial fibrosis ,business ,Biomarkers - Abstract
Background Stable plasma nitric oxide ( NO ) metabolites ( NO M ), composed predominantly of nitrate and nitrite, are attractive biomarkers of NO bioavailability. NO M levels integrate the influence of NO ‐synthase‐derived NO production/metabolism, dietary intake of inorganic nitrate/nitrite, and clearance of NO M . Furthermore, nitrate and nitrite, the most abundant NO M , can be reduced to NO via the nitrate‐nitrite‐ NO pathway. Methods and Results We compared serum NO M among subjects without heart failure (n=126), subjects with heart failure and preserved ejection fraction ( HF p EF ; n=43), and subjects with heart failure and reduced ejection fraction ( HF r EF ; n=32). LV mass and extracellular volume fraction were measured with cardiac MRI . Plasma NO M levels were measured after reduction to NO via reaction with vanadium ( III )/hydrochloric acid. Subjects with HF p EF demonstrated significantly lower unadjusted levels of NO M (8.0 μmol/L; 95% CI 6.2–10.4 μmol/L; ANOVA P =0.013) than subjects without HF (12.0 μmol/L; 95% CI 10.4–13.9 μmol/L) or those with HF r EF (13.5 μmol/L; 95% CI 9.7–18.9 μmol/L). There were no significant differences in NO M between subjects with HF r EF and subjects without HF . In a multivariable model that adjusted for age, sex, race, diabetes mellitus, body mass index, current smoking, systolic blood pressure, and glomerular filtration rate, HF p EF remained a predictor of lower NO M (β=−0.43; P =0.013). NO M did not correlate with LV mass, or LV diffuse fibrosis. Conclusions HF p EF , but not HF r EF , is associated with reduced plasma NO M , suggesting greater endothelial dysfunction, enhanced clearance, or deficient dietary ingestion of inorganic nitrate. Our findings may underlie the salutary effects of inorganic nitrate supplementation demonstrated in recent clinical trials in HF p EF .
- Published
- 2016
46. Pharmacokinetics and Pharmacodynamics of Inorganic Nitrate in Heart Failure With Preserved Ejection Fraction
- Author
-
Melissa Beraun, Paschalis-Thomas Doulias, Victor X Tan, Jeffrey Brandimarto, Raymond R. Townsend, Haideliza Soto-Calderon, Jesse Chittams, Kenneth B. Margulies, Julio A. Chirinos, Thomas P. Cappola, David C. Poole, Lien Trieu, Payman Zamani, Harry Ischiropoulos, and Swapna Varakantam
- Subjects
Male ,medicine.medical_specialty ,Supine position ,Physiology ,Qi ,Potassium Compounds ,030204 cardiovascular system & hematology ,Methemoglobinemia ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,Interquartile range ,law ,medicine ,Humans ,Exercise ,Aged ,Heart Failure ,Exercise Tolerance ,Nitrates ,business.industry ,Stroke Volume ,Middle Aged ,medicine.disease ,Confidence interval ,Surgery ,Blood pressure ,Heart failure ,Anesthesia ,Quality of Life ,Female ,Cardiology and Cardiovascular Medicine ,business ,Heart failure with preserved ejection fraction ,030217 neurology & neurosurgery - Abstract
Rationale: Nitrate-rich beetroot juice has been shown to improve exercise capacity in heart failure with preserved ejection fraction, but studies using pharmacological preparations of inorganic nitrate are lacking. Objectives: To determine (1) the dose–response effect of potassium nitrate (KNO 3 ) on exercise capacity; (2) the population-specific pharmacokinetic and safety profile of KNO 3 in heart failure with preserved ejection fraction. Methods and Results: We randomized 12 subjects with heart failure with preserved ejection fraction to oral KNO 3 (n=9) or potassium chloride (n=3). Subjects received 6 mmol twice daily during week 1, followed by 6 mmol thrice daily during week 2. Supine cycle ergometry was performed at baseline (visit 1) and after each week (visits 2 and 3). Quality of life was assessed with the Kansas City Cardiomyopathy Questionnaire. The primary efficacy outcome, peak O 2 -uptake, did not significantly improve ( P =0.13). Exploratory outcomes included exercise duration and quality of life. Exercise duration increased significantly with KNO 3 (visit 1: 9.87, 95% confidence interval [CI] 9.31–10.43 minutes; visit 2: 10.73, 95% CI 10.13–11.33 minute; visit 3: 11.61, 95% CI 11.05–12.17 minutes; P =0.002). Improvements in the Kansas City Cardiomyopathy Questionnaire total symptom (visit 1: 58.0, 95% CI 52.5–63.5; visit 2: 66.8, 95% CI 61.3–72.3; visit 3: 70.8, 95% CI 65.3–76.3; P =0.016) and functional status scores (visit 1: 62.2, 95% CI 58.5–66.0; visit 2: 68.6, 95% CI 64.9–72.3; visit 3: 71.1, 95% CI 67.3–74.8; P =0.01) were seen after KNO 3 . Pronounced elevations in trough levels of nitric oxide metabolites occurred with KNO 3 (visit 2: 199.5, 95% CI 98.7–300.2 μmol/L; visit 3: 471.8, 95% CI 377.8–565.8 μmol/L) versus baseline (visit 1: 38.0, 95% CI 0.00–132.0 μmol/L; P 3 did not lead to clinically significant hypotension or methemoglobinemia. After 6 mmol of KNO 3 , systolic blood pressure was reduced by a maximum of 17.9 (95% CI −28.3 to −7.6) mm Hg 3.75 hours later. Peak nitric oxide metabolites concentrations were 259.3 (95% CI 176.2–342.4) μmol/L 3.5 hours after ingestion, and the median half-life was 73.0 (interquartile range 33.4–232.0) minutes. Conclusions: KNO 3 is potentially well tolerated and improves exercise duration and quality of life in heart failure with preserved ejection fraction. This study reinforces the efficacy of KNO 3 and suggests that larger randomized trials are warranted. Clinical Trial Registration : URL: http://www.clinicaltrials.gov . Unique identifier: NCT02256345
- Published
- 2016
47. Effects of Liraglutide on Clinical Stability Among Patients With Advanced Heart Failure and Reduced Ejection Fraction: A Randomized Clinical Trial
- Author
-
Kenneth B, Margulies, Adrian F, Hernandez, Margaret M, Redfield, Michael M, Givertz, Guilherme H, Oliveira, Robert, Cole, Douglas L, Mann, David J, Whellan, Michael S, Kiernan, G Michael, Felker, Steven E, McNulty, Kevin J, Anstrom, Monica R, Shah, Eugene, Braunwald, Thomas P, Cappola, and M, Shah
- Subjects
Male ,medicine.medical_specialty ,030209 endocrinology & metabolism ,030204 cardiovascular system & hematology ,Placebo ,Patient Readmission ,Article ,law.invention ,03 medical and health sciences ,Ventricular Dysfunction, Left ,0302 clinical medicine ,Randomized controlled trial ,Double-Blind Method ,Interquartile range ,law ,Glucagon-Like Peptide 1 ,Internal medicine ,medicine ,Clinical endpoint ,Odds Ratio ,Humans ,Hypoglycemic Agents ,Aged ,Heart Failure ,Ejection fraction ,business.industry ,Liraglutide ,Hazard ratio ,Stroke Volume ,General Medicine ,Middle Aged ,medicine.disease ,United States ,Endocrinology ,Treatment Outcome ,Diabetes Mellitus, Type 2 ,Heart failure ,Cardiology ,Quality of Life ,Female ,business ,medicine.drug - Abstract
Importance Abnormal cardiac metabolism contributes to the pathophysiology of advanced heart failure with reduced left ventricular ejection fraction (LVEF). Glucagon-like peptide 1 (GLP-1) agonists have shown cardioprotective effects in early clinical studies of patients with advanced heart failure, irrespective of type 2 diabetes status. Objective To test whether therapy with a GLP-1 agonist improves clinical stability following hospitalization for acute heart failure. Design, Setting, and Participants Phase 2, double-blind, placebo-controlled randomized clinical trial of patients with established heart failure and reduced LVEF who were recently hospitalized. Patients were enrolled between August 2013 and March 2015 at 24 US sites. Interventions The GLP-1 agonist liraglutide (n = 154) or placebo (n = 146) via a daily subcutaneous injection; study drug was advanced to a dosage of 1.8 mg/d during the first 30 days as tolerated and continued for 180 days. Main Outcomes and Measures The primary end point was a global rank score in which all patients, regardless of treatment assignment, were ranked across 3 hierarchical tiers: time to death, time to rehospitalization for heart failure, and time-averaged proportional change in N-terminal pro-B-type natriuretic peptide level from baseline to 180 days. Higher values indicate better health (stability). Exploratory secondary outcomes included primary end point components, cardiac structure and function, 6-minute walk distance, quality of life, and combined events. Results Among the 300 patients who were randomized (median age, 61 years [interquartile range {IQR}, 52-68 years]; 64 [21%] women; 178 [59%] with type 2 diabetes; median LVEF of 25% [IQR, 19%-33%]; median N-terminal pro-B-type natriuretic peptide level of 2049 pg/mL [IQR, 1054-4235 pg/mL]), 271 completed the study. Compared with placebo, liraglutide had no significant effect on the primary end point (mean rank of 146 for the liraglutide group vs 156 for the placebo group, P = .31). There were no significant between-group differences in the number of deaths (19 [12%] in the liraglutide group vs 16 [11%] in the placebo group; hazard ratio, 1.10 [95% CI, 0.57-2.14]; P = .78) or rehospitalizations for heart failure (63 [41%] vs 50 [34%], respectively; hazard ratio, 1.30 [95% CI, 0.89-1.88]; P = .17) or for the exploratory secondary end points. Prespecified subgroup analyses in patients with diabetes did not reveal any significant between-group differences. The number of investigator-reported hyperglycemic events was 16 (10%) in the liraglutide group vs 27 (18%) in the placebo group and hypoglycemic events were infrequent (2 [1%] vs 4 [3%], respectively). Conclusions and Relevance Among patients recently hospitalized with heart failure and reduced LVEF, the use of liraglutide did not lead to greater posthospitalization clinical stability. These findings do not support the use of liraglutide in this clinical situation. Trial Registration clinicaltrials.gov Identifier:NCT01800968
- Published
- 2016
48. Transcriptomic Biomarkers of Cardiovascular Disease
- Author
-
Michael Morley, Thomas P. Cappola, and Dawn M. Pedrotty
- Subjects
Genetic Markers ,business.industry ,Gene Expression Profiling ,Computational biology ,Disease ,Prognosis ,Article ,Transcriptome ,Phenotype ,Gene Expression Regulation ,Cardiovascular Diseases ,Predictive Value of Tests ,Quantitative assessment ,Humans ,Medicine ,Genetic Predisposition to Disease ,Precision Medicine ,Quantitative Real-Time Polymerase Chain Reaction ,Cardiology and Cardiovascular Medicine ,business - Abstract
Transcriptomics is the study of how our genes are regulated and expressed in different biological settings. Technical advances now enable quantitative assessment of all expressed genes (ie, the entire “transcriptome”) in a given tissue at a given time. These approaches provide a powerful tool for understanding complex biological systems and for developing novel biomarkers. This chapter will introduce basic concepts in transcriptomics and available technologies for developing transcriptomic biomarkers. We will then review current and emerging applications in cardiovascular medicine.
- Published
- 2012
49. Multiple Biomarkers for Risk Prediction in Chronic Heart Failure
- Author
-
Bonnie Ky, Nancy K. Sweitzer, Lee R. Goldberg, Thomas P. Cappola, Wayne C. Levy, Benjamin French, James C. Fang, Mariell Jessup, and Alan H.B. Wu
- Subjects
Male ,medicine.medical_specialty ,medicine.medical_treatment ,Risk Assessment ,Severity of Illness Index ,Article ,Risk Factors ,Internal medicine ,Outpatients ,Confidence Intervals ,medicine ,Humans ,Intensive care medicine ,Proportional Hazards Models ,Heart Failure ,Immunoassay ,Receiver operating characteristic ,Proportional hazards model ,business.industry ,Hazard ratio ,Middle Aged ,Prognosis ,medicine.disease ,United States ,Confidence interval ,Survival Rate ,Transplantation ,ROC Curve ,Heart failure ,Ventricular assist device ,Disease Progression ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,Risk assessment ,business ,Biomarkers ,Follow-Up Studies - Abstract
Background— Prior studies have suggested using a panel of biomarkers that measure diverse biological processes as a prognostic tool in chronic heart failure. Whether this approach improves risk prediction beyond clinical evaluation is unknown. Methods and Results— In a multicenter cohort of 1513 chronic systolic heart failure patients, we measured a contemporary biomarker panel consisting of high-sensitivity C-reactive protein, myeloperoxidase, B-type natriuretic peptide, soluble fms-like tyrosine kinase receptor-1, troponin I, soluble toll-like receptor-2, creatinine, and uric acid. From this panel, we calculated a parsimonious multimarker score and assessed its performance in predicting risk of death, cardiac transplantation, or ventricular assist device placement in comparison to an established clinical risk score, the Seattle Heart Failure Model (SHFM). During a median follow-up of 2.5 years, there were 317 outcomes: 187 patients died; 99 were transplanted; and 31 had a ventricular assist device placed. In unadjusted Cox models, patients in the highest tertile of the multimarker score had a 13.7-fold increased risk of adverse outcomes compared with the lowest tertile (95% confidence interval, 8.75–21.5). These effects were independent of the SHFM (adjusted hazard ratio, 6.80; 95% confidence interval, 4.18–11.1). Addition of the multimarker score to the SHFM led to a significantly improved area under the receiver operating characteristic curve of 0.803 versus 0.756 ( P =0.003) and appropriately reclassified a significant number of patients who had the outcome into a higher risk category (net reclassification improvement, 25.2%; 95% confidence interval, 14.2–36.2%; P Conclusions— In ambulatory chronic heart failure patients, a score derived from multiple biomarkers integrating diverse biological pathways substantially improves prediction of adverse events beyond current metrics.
- Published
- 2012
50. Clinical Considerations of Heritable Factors in Common Heart Failure
- Author
-
Thomas P. Cappola and Gerald W. Dorn
- Subjects
medicine.medical_specialty ,Candidate gene ,Cardiac output ,Genome-wide association study ,Polymorphism, Single Nucleotide ,Article ,Diabetes mellitus ,Internal medicine ,Epidemiology ,Genetics ,medicine ,Animals ,Humans ,Intensive care medicine ,Adverse effect ,Genetics (clinical) ,Heart Failure ,business.industry ,Genetic Variation ,Dilated cardiomyopathy ,medicine.disease ,Heart failure ,Cardiology ,Cardiology and Cardiovascular Medicine ,business ,Signal Transduction - Abstract
Heart failure is a common condition responsible for at least 290 000 deaths each year in the United States alone.1 A small minority of heart failure cases are attributed to Mendelian or familial cardiomyopathies. The majority of systolic heart failure cases are not familial but represent the end result of 1 or many conditions that primarily injure the myocardium sufficiently to diminish cardiac output in the absence of compensatory mechanisms. Paradoxically, because they also injure the myocardium, it is the chronic actions of the compensatory mechanisms that in many instances contribute to the progression from simple cardiac injury to dilated cardiomyopathy and overt heart failure. Thus, the epidemiology of common heart failure appears to be just as sporadic as its major antecedent conditions (atherosclerosis, diabetes, hypertension, and viral myocarditis). Familial trends in preclinical cardiac remodeling2 and risk of developing heart failure3 reveal an important role for genetic modifiers in addition to clinical and environmental factors. Candidate gene studies performed over the past 10 years have identified a few polymorphic gene variants that modify risk or progression of common heart failure.4 Whole-genome sequencing will lead to the discovery of other genetic modifiers that were not candidates.5 The imminent availability of individual whole-genome sequences at a cost competitive with available genetic tests for familial cardiomyopathy will no doubt further expand the list of putative genetic heart failure modifiers. Heart failure risk alleles along with traditional clinical factors will need to be considered by clinical cardiologists in their design of optimal disease surveillance and prevention programs and in individually tailoring heart failure management. The use of individual genetic make-up is likely to have the earliest and greatest impact on managing patients with heart failure by tailoring available pharmacotherapeutics to optimize patient response and minimize adverse effects (ie, the …
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.