311 results on '"Margulies KB"'
Search Results
2. Development of dilated cardiomyopathy and impaired calcium homeostasis with cardiac-specific deletion of ESRRβ
- Author
-
Rowe, GC, Asimaki, A, Graham, EL, Martin, KD, Margulies, KB, Das, S, Saffitz, J, and Arany, Z
- Subjects
cardiovascular system ,cardiovascular diseases - Abstract
Mechanisms underlying the development of idiopathic dilated cardiomyopathy (DCM) remain poorly understood. Using transcription factor expression profiling, we identified estrogen-related receptor-β (ESRRβ), a member of the nuclear receptor family of transcription factors, as highly expressed in murine hearts and other highly oxidative striated muscle beds. Mice bearing cardiac-specific deletion of ESRRβ (MHC-ERRB KO) develop DCM and sudden death at ~10 mo of age. Isolated adult cardiomyocytes from the MHC-ERRB KO mice showed an increase in calcium sensitivity and impaired cardiomyocyte contractility, which preceded echocardiographic cardiac remodeling and dysfunction by several months. Histological analyses of myocardial biopsies from patients with various cardiomyopathies revealed that ESRRβ protein is absent from the nucleus of cardiomyocytes from patients with DCM but not other forms of cardiomyopathy (ischemic, hypertrophic, and arrhythmogenic right ventricular cardiomyopathy). Taken together these observations suggest that ESRRβ is a critical component in the onset of DCM by affecting contractility and calcium balance.NEW & NOTEWORTHY Estrogen-related receptor-β (ESRRβ) is highly expressed in the heart and cardiac-specific deletion results in the development of a dilated cardiomyopathy (DCM). ESRRβ is mislocalized in human myocardium samples with DCM, suggesting a possible role for ESRRβ in the pathogenesis of DCM in humans.
- Published
- 2017
3. Transcription Factor 7-like 2 Mediates Canonical Wnt/beta-Catenin Signaling and c-Myc Upregulation in Heart Failure
- Author
-
Hou, N, Ye, B, Li, X, Margulies, KB, Xu, H, Wang, X, and Li, F
- Subjects
desmin ,heart failure ,cell signaling/signal transduction ,cardiomyopathy ,acetylation - Published
- 2016
4. CaMKII phosphorylation of NaV1.5: Novel in vitro sites identified by mass spectrometry and reduced s516 phosphorylation in human heart failure
- Author
-
Herren, AW, Weber, DM, Rigor, RR, Margulies, KB, Phinney, BS, and Bers, DM
- Subjects
enzymes and coenzymes (carbohydrates) ,parasitic diseases ,environment and public health - Abstract
© 2015 American Chemical Society. The cardiac voltage-gated sodium channel, NaV1.5, drives the upstroke of the cardiac action potential and is a critical determinant of myocyte excitability. Recently, calcium (Ca2+)/calmodulin(CaM)-dependent protein kinase II (CaMKII) has emerged as a critical regulator of NaV1.5 function through phosphorylation of multiple residues including S516, T594, and S571, and these phosphorylation events may be important for the genesis of acquired arrhythmias, which occur in heart failure. However, phosphorylation of full-length human NaV1.5 has not been systematically analyzed and NaV1.5 phosphorylation in human heart failure is incompletely understood. In the present study, we used label-free mass spectrometry to assess phosphorylation of human NaV1.5 purified from HEK293 cells with full coverage of phosphorylatable sites and identified 23 sites that were phosphorylated by CaMKII in vitro. We confirmed phosphorylation of S516 and S571 by LC-MS/MS and found a decrease in S516 phosphorylation in human heart failure, using a novel phospho-specific antibody. This work furthers our understanding of the phosphorylation of NaV1.5 by CaMKII under normal and disease conditions, provides novel CaMKII target sites for functional validation, and provides the first phospho-proteomic map of full-length human NaV1.5.
- Published
- 2015
5. Elevated myocardial and lymphocyte beta ARK1 levels in human heart failure
- Author
-
Iaccarino, G, Barbato, E, Deamicis, V, Esposito, A, Silvestri, S, Margulies, Kb, Trimarco, B, and Koch, Wj
- Published
- 1999
6. Contribution of late sodium current (I(Na-L)) to rate adaptation of ventricular repolarization and reverse use-dependence of QT-prolonging agents.
- Author
-
Guo D, Lian J, Liu T, Cox R, Margulies KB, Kowey PR, Yan GX, Guo, Donglin, Lian, Jianfang, Liu, Tengxian, Cox, Robert, Margulies, Kenneth B, Kowey, Peter R, and Yan, Gan-Xin
- Abstract
Background: Abnormal rate adaptation of ventricular repolarization is arrhythmogenic. There is controversy on the underlying ionic mechanisms for rate-dependent change in repolarization.Objective: The purpose of this study was to examine the role of the late sodium current (I(Na-L)) in normal rate-dependence of ventricular repolarization and reverse use-dependence of QT-prolonging agents.Methods: The effects of I(Na-L) blockade, I(Na-L) enhancement, I(Kr) blockade, and changes in extracellular potassium concentration ([K(+)](o)) on rate adaptation of the QT interval and action potential duration (APD) were examined in isolated rabbit ventricular wedges and single myocytes. Rate dependence of I(Na-L), delayed rectifier potassium current (I(K)), and L-type calcium current (I(Ca)) was determined using a whole-cell, voltage clamp technique.Results: At control, APD exhibited rate-dependent changes in the multicellular preparations as well as in the isolated single ventricular myocytes when [K(+)](o) remained constant. The rate dependence of APD was significantly enhanced by reduction of [K(+)](o) from 4 to 1 mM or by I(Na-L) enhancement but was markedly blunted by the selective sodium channel blocker tetrodotoxin. The I(Kr) blocker dofetilide (3 nM) amplified the QT to basic cycle length slope (71.2 ± 13.1 ms/s vs 35.1 ± 8.8 ms/s in control, n = 4, P <.05). This reverse use-dependence was abolished by tetrodotoxin at 5 μM (11.4 ± 4.3 ms/s, n = 4, P <.01). There were no significant differences in I(Ca) or I(K) over the range of basic cycle lengths from 2,000 to 500 ms. However, I(Na-L) exhibited a significant rate-dependent reduction.Conclusion: I(Na-L) is sensitive to rate change due to its slow inactivation and recovery kinetics and plays a central role in the rate dependence of APD/QT and in the reverse use-dependence of select APD/QT-prolonging agents. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
7. Reciprocal regulation of myocardial microRNAs and messenger RNA in human cardiomyopathy and reversal of the microRNA signature by biomechanical support.
- Author
-
Matkovich SJ, Van Booven DJ, Youker KA, Torre-Amione G, Diwan A, Eschenbacher WH, Dorn LE, Watson MA, Margulies KB, Dorn GW 2nd, Matkovich, Scot J, Van Booven, Derek J, Youker, Keith A, Torre-Amione, Guillermo, Diwan, Abhinav, Eschenbacher, William H, Dorn, Lisa E, Watson, Mark A, Margulies, Kenneth B, and Dorn, Gerald W 2nd
- Published
- 2009
- Full Text
- View/download PDF
8. Transcriptional genomics associates FOX transcription factors with human heart failure.
- Author
-
Hannenhalli S, Putt ME, Gilmore JM, Wang J, Parmacek MS, Epstein JA, Morrisey EE, Margulies KB, and Cappola TP
- Published
- 2006
9. S100A1: Another Step Toward Therapeutic Development for Heart Failure.
- Author
-
Belmonte SL, Margulies KB, and Blaxall BC
- Published
- 2011
- Full Text
- View/download PDF
10. Altered myocardial Ca2+ cycling after left ventricular assist device support in the failing human heart.
- Author
-
Chaudhary KW, Rossman EI, Piacentino V III, Kenessey A, Weber C, Gaughan JP, Ojamaa K, Klein I, Bers DM, Houser SR, and Margulies KB
- Published
- 2004
- Full Text
- View/download PDF
11. Elevated myocardial and lymphocyte GRK2 expression and activity in human heart failure
- Author
-
Vincenzo De Amicis, Guido Iaccarino, Emanuele Barbato, Ersilia Cipolletta, Kenneth B. Margulies, Bruno Trimarco, Walter J. Koch, Dario Leosco, Iaccarino, G, Barbato, E, Cipolletta, E, DE AMICIS, Vincenzo, Margulies, Kb, Leosco, Dario, Trimarco, Bruno, Koch, W. J., Iaccarino, Guido, Emanuele, Barbato, Ersilia, Cipolletta, Kenneth B., Margulie, and Walter J., Koch
- Subjects
Male ,medicine.medical_specialty ,Heart disease ,G-Protein-Coupled Receptor Kinase 2 ,Lymphocyte ,medicine.medical_treatment ,enzymology ,Severity of Illness Index ,Internal medicine ,Receptors, Adrenergic, beta ,Receptors ,medicine ,Cyclic AMP ,Humans ,Lymphocytes ,Kinase activity ,enzymology/physiopathology/surgery ,Aged ,Biological Markers ,metabolism ,biosynthesis ,Female ,Heart Failure ,Heart Transplantation ,Middle Aged ,Myocardium ,Adrenergic ,beta ,physiology ,Signal Transduction ,Stroke Volume ,beta-Adrenergic Receptor Kinases ,Heart transplantation ,Ejection fraction ,biology ,business.industry ,Beta adrenergic receptor kinase ,medicine.disease ,Endocrinology ,medicine.anatomical_structure ,Heart failure ,Circulatory system ,biology.protein ,Cardiology and Cardiovascular Medicine ,business ,Biomarkers - Abstract
Aims The G protein-coupled receptor kinase-2 (GRK2 or b-ARK1) regulates b-adrenergic receptors (b-ARs) in the heart, and its cardiac expression is elevated in human heart failure (HF). We sought to determine whether myocardial levels and activity of GRK2 could be monitored using white blood cells, which have been used to study cardiac b-ARs. Moreover, we were interested in determining whether GRK2 levels in myocardium and lymphocytes may be associated with b-AR dysfunction and HF severity. Methods and results In myocardial biopsies from explanted failing human hearts, GRK activity was inversely correlated with b-AR-mediated cAMP production (R 2 ¼ 20.215, P , 0.05, n ¼ 24). Multiple regression analysis confirmed that GRK activity participates with b-AR density to regulate catecholaminesensitive cAMP responses. Importantly, there was a direct correlation between myocardial and lymphocytes GRK2 activity (R 2 ¼ 0.5686, P , 0.05, n ¼ 10). Lymphocyte GRK activity was assessed in HF patients with various ejection fractions (EFs) (n ¼ 33), and kinase activity was significantly higher in patients with lower EFs and was higher with increasing NYHA class (P , 0.001). Conclusion Myocardial GRK2 expression and activity are mirrored by lymphocyte levels of this kinase, and its elevation in HF is associated with the loss of b-AR responsiveness and appears to increase with disease severity. Therefore, lymphocytes may provide a surrogate for monitoring cardiac GRK2 in human HF.
- Published
- 2008
12. Genome-wide association analysis provides insights into the molecular etiology of dilated cardiomyopathy.
- Author
-
Zheng SL, Henry A, Cannie D, Lee M, Miller D, McGurk KA, Bond I, Xu X, Issa H, Francis C, De Marvao A, Theotokis PI, Buchan RJ, Speed D, Abner E, Adams L, Aragam KG, Ärnlöv J, Raja AA, Backman JD, Baksi J, Barton PJR, Biddinger KJ, Boersma E, Brandimarto J, Brunak S, Bundgaard H, Carey DJ, Charron P, Cook JP, Cook SA, Denaxas S, Deleuze JF, Doney AS, Elliott P, Erikstrup C, Esko T, Farber-Eger EH, Finan C, Garnier S, Ghouse J, Giedraitis V, Guðbjartsson DF, Haggerty CM, Halliday BP, Helgadottir A, Hemingway H, Hillege HL, Kardys I, Lind L, Lindgren CM, Lowery BD, Manisty C, Margulies KB, Moon JC, Mordi IR, Morley MP, Morris AD, Morris AP, Morton L, Noursadeghi M, Ostrowski SR, Owens AT, Palmer CNA, Pantazis A, Pedersen OBV, Prasad SK, Shekhar A, Smelser DT, Srinivasan S, Stefansson K, Sveinbjörnsson G, Syrris P, Tammesoo ML, Tayal U, Teder-Laving M, Thorgeirsson G, Thorsteinsdottir U, Tragante V, Trégouët DA, Treibel TA, Ullum H, Valdes AM, van Setten J, van Vugt M, Veluchamy A, Verschuren WMM, Villard E, Yang Y, Asselbergs FW, Cappola TP, Dube MP, Dunn ME, Ellinor PT, Hingorani AD, Lang CC, Samani NJ, Shah SH, Smith JG, Vasan RS, O'Regan DP, Holm H, Noseda M, Wells Q, Ware JS, and Lumbers RT
- Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure and cardiac transplantation. We report a genome-wide association study and multi-trait analysis of DCM (14,256 cases) and three left ventricular traits (36,203 UK Biobank participants). We identified 80 genomic risk loci and prioritized 62 putative effector genes, including several with rare variant DCM associations (MAP3K7, NEDD4L and SSPN). Using single-nucleus transcriptomics, we identify cellular states, biological pathways, and intracellular communications that drive pathogenesis. We demonstrate that polygenic scores predict DCM in the general population and modify penetrance in carriers of rare DCM variants. Our findings may inform the design of genetic testing strategies that incorporate polygenic background. They also provide insights into the molecular etiology of DCM that may facilitate the development of targeted therapeutics., Competing Interests: Competing interests: S.L.Z. has acted as a consultant for Health Lumen. A.H. and R.T.L. have received funding from Pfizer Inc. R.T.L. has performed paid consultancy for Health Lumen and Fitfile Ltd. J.S.W. has acted as a consultant for MyoKardia, Pfizer, Foresite Labs and Health Lumen and received institutional support from Bristol Myers Squibb and Pfizer Inc. P.C. has received personal fees for consultancies, outside the present work, for Amicus, Pfizer Inc., Owkin and Bristol Myers Squibb. M.-P.D. declares holding equity in Dalcor Pharmaceuticals, unrelated to this work. The authors who are affiliated with deCODE genetics/Amgen Inc. and Regeneron Pharmaceuticals declare competing financial interests as employees. The other authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Large-scale single-nuclei profiling identifies role for ATRNL1 in atrial fibrillation.
- Author
-
Hill MC, Simonson B, Roselli C, Xiao L, Herndon CN, Chaffin M, Mantineo H, Atwa O, Bhasin H, Guedira Y, Bedi KC Jr, Margulies KB, Klattenhoff CA, Tucker NR, and Ellinor PT
- Subjects
- Humans, Macrophages metabolism, Animals, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics, Male, Action Potentials, Cell Nucleus metabolism, Heart Atria metabolism, Female, Gene Expression Profiling, RNA-Seq, Mice, Middle Aged, Atrial Fibrillation genetics, Atrial Fibrillation metabolism, Myocytes, Cardiac metabolism
- Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in humans, yet the molecular basis of AF remains incompletely understood. To determine the cell type-specific transcriptional changes underlying AF, we perform single-nucleus RNA-seq (snRNA-seq) on left atrial (LA) samples from patients with AF and controls. From more than 175,000 nuclei we find that only cardiomyocytes (CMs) and macrophages (MΦs) have a significant number of differentially expressed genes in patients with AF. Attractin Like 1 (ATRNL1) was overexpressed in CMs among patients with AF and localized to the intercalated disks. Further, in both knockdown and overexpression experiments we identify a potent role for ATRNL1 in cell stress response, and in the modulation of the cardiac action potential. Finally, we detect an unexpected expression pattern for a leading AF candidate gene, KCNN3. In sum, we uncover a role for ATRNL1 which may serve as potential therapeutic target for this common arrhythmia., Competing Interests: Competing interests C.A.K. is an employee of Bayer US LLC (a subsidiary of Bayer AG) and may own stock in Bayer. The Precision Cardiology Laboratory is joint effort between the Broad Institute and Bayer AG. P.T.E. has received sponsored research support from Bayer AG, Novo-Nordisk, Bristol Myers Squibb and Pfizer; he has also served on advisory boards or consulted for Bayer AG. C.R. is a full-time employee at GSK as of July 2024. All remaining authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
14. Unipolar Voltage Mapping to Predict Recovery of Left Ventricular Ejection Fraction in Patients With Recent-Onset Nonischemic Cardiomyopathy.
- Author
-
Chaumont C, Peyster E, Siontis KC, Muser D, Kapa S, Markman TM, Pathak RK, Oraii A, Rodriguez-Queralto O, Anselme F, Margulies KB, Marchlinski FE, and Frankel DS
- Abstract
Background: The ability to predict recovery of left ventricular ejection fraction (LVEF) in response to guideline-directed therapy among patients with nonischemic cardiomyopathy is desired. We sought to determine whether left ventricular endocardial unipolar voltage measured during invasive electroanatomic mapping could be used to predict LVEF recovery among those with recent-onset nonischemic cardiomyopathy., Methods: We analyzed the left ventricular voltage maps of patients included in the eMAP trial (Electrogram-Guided Myocardial Advanced Phenotyping; NCT03293381), a prospective, nonrandomized, interventional trial conducted at 2 institutions between 2017 and 2020. Patients had recent-onset nonischemic cardiomyopathy defined by LVEF ≤45% and development of symptoms or signs of heart failure within the past 6 months. Detailed voltage maps of the left ventricular endocardium were generated using the CARTO electroanatomic mapping system. Abnormal unipolar amplitude was defined as <8.27 mV. The primary end point was recovery of LVEF (Recovery) defined by a 1-year LVEF ≥50% or ≥45% with ≥10% increase from baseline., Results: Of the 29 enrolled patients (median age, 49 years [25th percentile, 39; 75th percentile, 59], 8 females [27.6%]), LVEF recovered in 13 (44.8%) by 1-year follow-up. The percentage of total endocardial surface area with unipolar voltage abnormality (AUA) was significantly lower among Recovery patients than No Recovery patients (18.2% [6.4, 22.4] versus 80.0% [29.5, 90.9]; P =0.004). Percent AUA was associated with lower likelihood of Recovery (odds ratio, 0.64 per 10% increase in AUA; 95% CI, 0.47-0.88; P =0.006). A 28% cutoff value for percent AUA was 92% sensitive and 75% specific with an area under the receiver operating characteristic curve of 0.81 (95% CI, 0.63-0.99; P =0.001) for predicting recovery versus no recovery. The majority of patients (12 of 13; 92.3%) with a percent AUA >28% did not recover., Conclusions: Left ventricular unipolar voltage abnormality is a potent predictor of LVEF recovery among patients recently diagnosed with nonischemic cardiomyopathy. Detailed left ventricular unipolar voltage mapping could therefore be used as a valuable prognostic tool in guiding treatment decisions.
- Published
- 2024
- Full Text
- View/download PDF
15. Metabolic Effects of the SGLT2 Inhibitor Dapagliflozin in Heart Failure Across the Spectrum of Ejection Fraction.
- Author
-
Selvaraj S, Patel S, Sauer AJ, McGarrah RW, Jones P, Kwee LC, Windsor SL, Ilkayeva O, Muehlbauer MJ, Newgard CB, Borlaug BA, Kitzman DW, Shah SJ, Margulies KB, Husain M, Inzucchi SE, McGuire DK, Lanfear DE, Javaheri A, Umpierrez G, Mentz RJ, Sharma K, Kosiborod MN, and Shah SH
- Subjects
- Humans, Female, Male, Aged, Middle Aged, Ventricular Function, Left drug effects, Treatment Outcome, Metabolomics, Biomarkers blood, Fatty Acids metabolism, Benzhydryl Compounds therapeutic use, Glucosides therapeutic use, Sodium-Glucose Transporter 2 Inhibitors therapeutic use, Heart Failure drug therapy, Heart Failure physiopathology, Heart Failure metabolism, Stroke Volume drug effects, Stroke Volume physiology
- Abstract
Background: Mechanisms of benefit with SGLT2is (sodium-glucose cotransporter-2 inhibitors) in heart failure (HF) remain incompletely characterized. Dapagliflozin alters ketone and fatty acid metabolism in HF with reduced ejection fraction though similar effects have not been observed in HF with preserved ejection fraction. We explore whether metabolic effects of SGLT2is vary across the left ventricular ejection fraction spectrum and their relationship with cardiometabolic end points in 2 randomized trials of dapagliflozin in HF., Methods: Metabolomic profiling of 61 metabolites was performed in 527 participants from DEFINE-HF (Dapagliflozin Effects on Biomarkers, Symptoms and Functional Status in Patients With HF With Reduced Ejection Fraction) and PRESERVED-HF (Dapagliflozin in PRESERVED Ejection Fraction HF; 12-week, placebo-controlled trials of dapagliflozin in HF with reduced ejection fraction and HF with preserved ejection fraction, respectively). Linear regression was used to assess changes in principal components analysis-defined metabolite factors with treatment from baseline to 12 weeks, as well as the relationship between changes in metabolite clusters and HF-related end points., Results: The mean age was 66±11 years, 43% were female, and 33% were self-identified as Black. Two principal components analysis-derived metabolite factors (which were comprised of ketone and short-/medium-chain acylcarnitines) increased with dapagliflozin compared with placebo. Ketosis (defined as 3-hydroxybutyrate >500 μM) was achieved in 4.5% with dapagliflozin versus 1.2% with placebo ( P =0.03). There were no appreciable treatment effects on amino acids, including branched-chain amino acids. Increases in several acylcarnitines were consistent across LVEF ( P
interaction >0.10), whereas the ketogenic effect diminished at higher LVEF ( Pinteraction =0.01 for 3-hydroxybutyrate). Increases in metabolites reflecting mitochondrial dysfunction (particularly long-chain acylcarnitines) and aromatic amino acids and decreases in branched-chain amino acids were associated with worse HF-related outcomes in the overall cohort, with consistency across treatment and LVEF., Conclusions: SGLT2is demonstrate common (fatty acid) and distinct (ketogenic) metabolic signatures across the LVEF spectrum. Changes in key pathways related to fatty acid and amino acid metabolism are associated with HF-related end points and may serve as therapeutic targets across HF subtypes., Registration: URL: https://www.clinicaltrials.gov; Unique Identifiers: NCT03030235 and NCT02653482., Competing Interests: Dr Selvaraj receives research support from the National Heart, Lung, and Blood Institute (grant K23HL161348), the Doris Duke Foundation (grant 2020061), the American Heart Association (grant 935275), the Mandel Foundation, the Duke Heart Center Leadership Council, the Institute for Translational Medicine and Therapeutics, and the Foundation for Sarcoidosis Research. Dr Selvaraj has participated in advisory boards for AstraZeneca for unrelated work. Dr Sauer performs consulting and advising or receives research funding from Abbott, Boston Scientific, Biotronik, Bayer, Amgen, CSL Vifor, AstraZeneca, Acorai, Story Health, FIRE1, uLink, General Prognostics, Impulse Dynamics, Edwards Lifesciences, 35Pharma, Rivus, and Pfizer, and he owns stock as a senior advisor to ISHI, a private digital health company. Dr McGarrah has been a consultant for AstraZeneca and M3 and received research funding from Eli Lilly. Dr Newgard is a member of the Eli Lilly Global Diabetes Advisory Board. Dr Borlaug receives research support from the National Institutes of Health (NIH) and the US Department of Defense and research grant funding from AstraZeneca, Axon, GlaxoSmithKline, Medtronic, Mesoblast, Novo Nordisk, and Tenax Therapeutics. Dr Borlaug has served as a consultant for Actelion, Amgen, Aria, BD, Boehringer Ingelheim, Cytokinetics, Edwards Lifesciences, Eli Lilly, Janssen, Merck, and Novo Nordisk. Dr Borlaug and Shah are named inventors (US Patent 10,307,179) for the tools and approach for a minimally invasive pericardial modification procedure to treat heart failure. Dr Kitzman has been a consultant for AstraZeneca, Pfizer, Corvia Medical, Bayer, Boehringer Ingelheim, Novo Nordisk, Rivus, and St. Luke’s Medical Center; received grant support from Novartis, AstraZeneca, Bayer, Pfizer, Novo Nordisk, Rivus, and St. Luke’s Medical Center; and owns stock in Gilead Sciences. Dr Sanjiv Shah reports support from research grants from the NIH (U54 HL160273, R01 HL140731, and R01 HL149423), Pfizer, and AstraZeneca and consulting fees from Abbott, Alleviant, AstraZeneca, Amgen, Aria CV, Axon Therapies, Bayer, Boehringer Ingelheim, Boston Scientific, Bristol Myers Squibb, Cyclerion, Corvia, Cytokinetics, Edwards Lifesciences, Eidos, Imara, Impulse Dynamics, Intellia, Ionis, Lilly, Merck, Metabolic Flux, MyoKardia, NGM Biopharmaceuticals, Novartis, Novo Nordisk, Pfizer, Prothena, Regeneron, Rivus, Sardocor, Shifamed, Tenax, Tenaya, Ultromics, and United Therapeutics. Dr Margulies reports sponsored research funding from Amgen, Bristol Myers Squibb, and Lexicon Pharmaceuticals and advisory board activities for Amgen and Bristol Myers Squibb. Dr David Lanfear reports support from NIH grants (P50MD017351 and R01HL132154) and Illumina; consulting fees from Janssen, AstraZeneca, and Abbot Laboratories (via ACI clinical); and clinical trial participation with Lilly, Pfizer, AstraZeneca, Bayer, Illumina, and Janssen. Dr Kosiborod reports research grant support from AstraZeneca, Boehringer Ingelheim, and Pfizer; is on the consultant/advisory board of 35Pharma, Alnylam, Amgen, Applied Therapeutics, AstraZeneca, Bayer, Boehringer Ingelheim, Cytokinetics, Dexcom, Eli Lilly, Esperion Therapeutics, Imbria Pharmaceuticals, Janssen, Lexicon Pharmaceuticals, Merck (Diabetes and Cardiovascular), Novo Nordisk, Pharmacosmos, Pfizer, Sanofi, scPharmaceuticals, Structure Therapeutics, Vifor Pharma, and Youngene Therapeutics; other research support from AstraZeneca; honoraria from AstraZeneca, Boehringer Ingelheim, and Novo Nordisk; and stock options in Artera Health and Saghmos Therapeutics. Dr Svati Shah reports research funding through sponsored research agreements to Duke University from AstraZeneca, Lilly Inc, Verily Inc, and nference and is a co-inventor of unlicensed patents held by Duke University. The other authors report no conflicts.- Published
- 2024
- Full Text
- View/download PDF
16. Cardiac NAD + depletion in mice promotes hypertrophic cardiomyopathy and arrhythmias prior to impaired bioenergetics.
- Author
-
Doan KV, Luongo TS, Ts'olo TT, Lee WD, Frederick DW, Mukherjee S, Adzika GK, Perry CE, Gaspar RB, Walker N, Blair MC, Bye N, Davis JG, Holman CD, Chu Q, Wang L, Rabinowitz JD, Kelly DP, Cappola TP, Margulies KB, and Baur JA
- Subjects
- Animals, Disease Models, Animal, Cytokines metabolism, Mice, Knockout, Mice, Inbred C57BL, Pyridinium Compounds, Male, Death, Sudden, Cardiac etiology, Death, Sudden, Cardiac pathology, Mice, Niacinamide analogs & derivatives, Niacinamide pharmacology, Niacinamide therapeutic use, Niacinamide metabolism, Electrocardiography, Nicotinamide Phosphoribosyltransferase metabolism, Nicotinamide Phosphoribosyltransferase genetics, NAD metabolism, Energy Metabolism, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Arrhythmias, Cardiac metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology
- Abstract
Nicotinamide adenine dinucleotide (NAD
+ ) is an essential co-factor in metabolic reactions and co-substrate for signaling enzymes. Failing human hearts display decreased expression of the major NAD+ biosynthetic enzyme nicotinamide phosphoribosyltransferase (Nampt) and lower NAD+ levels, and supplementation with NAD+ precursors is protective in preclinical models. Here we show that Nampt loss in adult cardiomyocytes caused depletion of NAD+ along with marked metabolic derangements, hypertrophic remodeling and sudden cardiac deaths, despite unchanged ejection fraction, endurance and mitochondrial respiratory capacity. These effects were directly attributable to NAD+ loss as all were ameliorated by restoring cardiac NAD+ levels with the NAD+ precursor nicotinamide riboside (NR). Electrocardiograms revealed that loss of myocardial Nampt caused a shortening of QT intervals with spontaneous lethal arrhythmias causing sudden cardiac death. Thus, changes in NAD+ concentration can have a profound influence on cardiac physiology even at levels sufficient to maintain energetics., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
17. Virally delivered CMYA5 enhances the assembly of cardiac dyads.
- Author
-
Lu F, Liou C, Ma Q, Wu Z, Xue B, Xia Y, Xia S, Trembley MA, Ponek A, Xie W, Shani K, Bortolin RH, Prondzynski M, Berkson P, Zhang X, Naya FJ, Bedi KC, Margulies KB, Zhang D, Parker KK, and Pu WT
- Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) lack nanoscale structures essential for efficient excitation-contraction coupling. Such nanostructures, known as dyads, are frequently disrupted in heart failure. Here we show that the reduced expression of cardiomyopathy-associated 5 (CMYA5), a master protein that establishes dyads, contributes to dyad disorganization in heart failure and to impaired dyad assembly in hiPSC-CMs, and that a miniaturized form of CMYA5 suitable for delivery via an adeno-associated virus substantially improved dyad architecture and normalized cardiac function under pressure overload. In hiPSC-CMs, the miniaturized form of CMYA5 increased contractile forces, improved Ca
2+ handling and enhanced the alignment of sarcomere Z-lines with ryanodine receptor 2, a protein that mediates the sarcoplasmic release of stored Ca2+ . Our findings clarify the mechanisms responsible for impaired dyad structure in diseased cardiomyocytes, and suggest strategies for promoting dyad assembly and stability in heart disease and during the derivation of hiPSC-CMs., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
18. Phosphorylation of CRYAB Induces a Condensatopathy to Worsen Post-Myocardial Infarction Left Ventricular Remodeling.
- Author
-
Islam M, Rawnsley DR, Ma X, Navid W, Zhao C, Foroughi L, Murphy JT, Navid H, Weinheimer CJ, Kovacs A, Nigro J, Diwan A, Chang R, Kumari M, Young ME, Razani B, Margulies KB, Abdellatif M, Sedej S, Javaheri A, Covey DF, Mani K, and Diwan A
- Abstract
Protein aggregates are emerging therapeutic targets in rare monogenic causes of cardiomyopathy and amyloid heart disease, but their role in more prevalent heart failure syndromes remains mechanistically unexamined. We observed mis-localization of desmin and sarcomeric proteins to aggregates in human myocardium with ischemic cardiomyopathy and in mouse hearts with post-myocardial infarction ventricular remodeling, mimicking findings of autosomal-dominant cardiomyopathy induced by R120G mutation in the cognate chaperone protein, CRYAB. In both syndromes, we demonstrate increased partitioning of CRYAB phosphorylated on serine-59 to NP40-insoluble aggregate-rich biochemical fraction. While CRYAB undergoes phase separation to form condensates, the phospho-mimetic mutation of serine-59 to aspartate (S59D) in CRYAB mimics R120G-CRYAB mutants with reduced condensate fluidity, formation of protein aggregates and increased cell death. Conversely, changing serine to alanine (phosphorylation-deficient mutation) at position 59 (S59A) restored condensate fluidity, and reduced both R120G-CRYAB aggregates and cell death. In mice, S59D CRYAB knock-in was sufficient to induce desmin mis-localization and myocardial protein aggregates, while S59A CRYAB knock-in rescued left ventricular systolic dysfunction post-myocardial infarction and preserved desmin localization with reduced myocardial protein aggregates. 25-Hydroxycholesterol attenuated CRYAB serine-59 phosphorylation and rescued post-myocardial infarction adverse remodeling. Thus, targeting CRYAB phosphorylation-induced condensatopathy is an attractive strategy to counter ischemic cardiomyopathy.
- Published
- 2024
- Full Text
- View/download PDF
19. Pathophysiological insights into HFpEF from studies of human cardiac tissue.
- Author
-
Fayyaz AU, Eltony M, Prokop LJ, Koepp KE, Borlaug BA, Dasari S, Bois MC, Margulies KB, Maleszewski JJ, Wang Y, and Redfield MM
- Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major, worldwide health-care problem. Few therapies for HFpEF exist because the pathophysiology of this condition is poorly defined and, increasingly, postulated to be diverse. Although perturbations in other organs contribute to the clinical profile in HFpEF, altered cardiac structure, function or both are the primary causes of this heart failure syndrome. Therefore, studying myocardial tissue is fundamental to improve pathophysiological insights and therapeutic discovery in HFpEF. Most studies of myocardial changes in HFpEF have relied on cardiac tissue from animal models without (or with limited) confirmatory studies in human cardiac tissue. Animal models of HFpEF have evolved based on theoretical HFpEF aetiologies, but these models might not reflect the complex pathophysiology of human HFpEF. The focus of this Review is the pathophysiological insights gained from studies of human HFpEF myocardium. We outline the rationale for these studies, the challenges and opportunities in obtaining myocardial tissue from patients with HFpEF and relevant comparator groups, the analytical approaches, the pathophysiological insights gained to date and the remaining knowledge gaps. Our objective is to provide a roadmap for future studies of cardiac tissue from diverse cohorts of patients with HFpEF, coupling discovery biology with measures to account for pathophysiological diversity., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
20. Endogenous adenine is a potential driver of the cardiovascular-kidney-metabolic syndrome.
- Author
-
Tamayo I, Lee HJ, Aslam MI, Liu JJ, Ragi N, Karanam V, Maity S, Saliba A, Treviño E, Zheng H, Lim SC, Lanzer JD, Bjornstad P, Tuttle K, Bedi KC Jr, Margulies KB, Ramachandran V, Abdel-Latif A, Saez-Rodriguez J, Iyengar R, Bopassa JC, and Sharma K
- Abstract
Mechanisms underlying the cardiovascular-kidney-metabolic (CKM) syndrome are unknown, although key small molecule metabolites may be involved. Bulk and spatial metabolomics identified adenine to be upregulated and specifically enriched in coronary blood vessels in hearts from patients with diabetes and left ventricular hypertrophy. Single nucleus gene expression studies revealed that endothelial methylthioadenosine phosphorylase (MTAP) was increased in human hearts with hypertrophic cardiomyopathy. The urine adenine/creatinine ratio in patients was predictive of incident heart failure with preserved ejection fraction. Heart adenine and MTAP gene expression was increased in a 2-hit mouse model of hypertrophic heart disease and in a model of diastolic dysfunction with diabetes. Inhibition of MTAP blocked adenine accumulation in the heart, restored heart dysfunction in mice with type 2 diabetes and prevented ischemic heart damage in a rat model of myocardial infarction. Mechanistically, adenine-induced impaired mitophagy was reversed by reduction of mTOR. These studies indicate that endogenous adenine is in a causal pathway for heart failure and ischemic heart disease in the context of CKM syndrome., Competing Interests: Competing interests: Dr. Margulies holds research grants from Amgen and serves as a scientific consultant/advisory board member for Bristol Myers Squibb and Amgen. Dr. Sharma serves on the data safety board for Cara Therapeutics and holds equity in SygnaMap. All other authors declare that they have no competing interests. Dr. Tuttle has received investigator-initiated grant support (to Providence Inland Northwest Health) from Travere and Bayer outside of the submitted work; consultancy fees from AstraZeneca, Boehringer Ingelheim, Eli Lilly and Company, Novo Nordisk and Travere; speaker fees from AstraZeneca, Eli Lilly, and Novo Nordisk. Dr. Julia Saez-Rodriguez reports funding from GSK, Pfizer and Sanofi & fees/honoraria from Travere Therapeutics, Stadapharm, Astex, Owkin, Pfizer and Grunenthal.
- Published
- 2024
- Full Text
- View/download PDF
21. Myocardial ultrastructure of human heart failure with preserved ejection fraction.
- Author
-
Meddeb M, Koleini N, Binek A, Keykhaei M, Darehgazani R, Kwon S, Aboaf C, Margulies KB, Bedi KC Jr, Lehar M, Sharma K, Hahn VS, Van Eyk JE, Drachenberg CI, and Kass DA
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Microscopy, Electron, Transmission, Ventricular Function, Left physiology, Sarcomeres ultrastructure, Sarcomeres metabolism, Sarcomeres pathology, Biopsy, Proteomics, Obesity pathology, Obesity metabolism, Lipid Droplets metabolism, Comorbidity, Heart Failure pathology, Heart Failure physiopathology, Heart Failure metabolism, Stroke Volume, Myocardium pathology, Myocardium metabolism, Myocardium ultrastructure, Mitochondria, Heart ultrastructure, Mitochondria, Heart pathology, Mitochondria, Heart metabolism
- Abstract
Over half of patients with heart failure have a preserved ejection fraction (>50%, called HFpEF), a syndrome with substantial morbidity/mortality and few effective therapies
1 . Its dominant comorbidity is now obesity, which worsens disease and prognosis1-3 . Myocardial data from patients with morbid obesity and HFpEF show depressed myocyte calcium-stimulated tension4 and disrupted gene expression of mitochondrial and lipid metabolic pathways5,6 , abnormalities shared by human HF with a reduced EF but less so in HFpEF without severe obesity. The impact of severe obesity on human HFpEF myocardial ultrastructure remains unexplored. Here we assessed the myocardial ultrastructure in septal biopsies from patients with HFpEF using transmission electron microscopy. We observed sarcomere disruption and sarcolysis, mitochondrial swelling with cristae separation and dissolution and lipid droplet accumulation that was more prominent in the most obese patients with HFpEF and not dependent on comorbid diabetes. Myocardial proteomics revealed associated reduction in fatty acid uptake, processing and oxidation and mitochondrial respiration proteins, particularly in very obese patients with HFpEF., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
22. Vasohibin inhibition improves myocardial relaxation in a rat model of heart failure with preserved ejection fraction.
- Author
-
Eaton DM, Lee BW, Caporizzo MA, Iyengar A, Chen CY, Uchida K, Marcellin G, Lannay Y, Vite A, Bedi KC Jr, Brady CF, Smolyak JN, Meldrum D, Dominic J, Weingarten N, Patel M, Belec A, Hached K, Atluri P, Van Der Laan S, Prosser BL, and Margulies KB
- Subjects
- Animals, Humans, Male, Rats, Cell Cycle Proteins metabolism, Cell Cycle Proteins antagonists & inhibitors, Diastole drug effects, Myocardium pathology, Myocardium metabolism, Obesity drug therapy, Obesity physiopathology, Rats, Inbred WKY, Tubulin metabolism, Disease Models, Animal, Heart Failure drug therapy, Heart Failure physiopathology, Heart Failure pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Stroke Volume drug effects
- Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome associated with increased myocardial stiffness and cardiac filling abnormalities. Prior studies implicated increased α-tubulin detyrosination, which is catalyzed by the vasohibin enzymes, as a contributor to increased stabilization of the cardiomyocyte microtubule network (MTN) and stiffness in failing human hearts. We explored whether increased MTN detyrosination contributed to impaired diastolic function in the ZSF1 obese rat model of HFpEF and designed a small-molecule vasohibin inhibitor to ablate MTN detyrosination in vivo. Compared with ZSF1 lean and Wistar Kyoto rats, obese rats exhibited increased tubulin detyrosination concomitant with diastolic dysfunction, left atrial enlargement, and cardiac hypertrophy with a preserved left ventricle ejection fraction, consistent with an HFpEF phenotype. Ex vivo myocardial phenotyping assessed cardiomyocyte mechanics and contractility. Vasohibin inhibitor treatment of isolated cardiomyocytes from obese rats resulted in reduced stiffness and faster relaxation. Acute in vivo treatment with vasohibin inhibitor improved diastolic relaxation in ZSF1 obese rats compared with ZSF1 lean and Wistar Kyoto rats. Vasohibin inhibition also improved relaxation in isolated human cardiomyocytes from both failing and nonfailing hearts. Our data suggest the therapeutic potential for vasohibin inhibition to reduce myocardial stiffness and improve relaxation in HFpEF.
- Published
- 2024
- Full Text
- View/download PDF
23. Nuclear ATP-citrate lyase regulates chromatin-dependent activation and maintenance of the myofibroblast gene program.
- Author
-
Lazaropoulos MP, Gibb AA, Chapski DJ, Nair AA, Reiter AN, Roy R, Eaton DM, Bedi KC Jr, Margulies KB, Wellen KE, Estarás C, Vondriska TM, and Elrod JW
- Subjects
- Animals, Acetylation drug effects, Cell Nucleus metabolism, Cell Nucleus drug effects, Smad3 Protein metabolism, Smad3 Protein genetics, Cells, Cultured, Chromatin metabolism, Mice, Knockout, Transforming Growth Factor beta metabolism, Disease Models, Animal, Signal Transduction, Mice, Inbred C57BL, Male, Mice, Gene Expression Regulation drug effects, Myofibroblasts metabolism, Myofibroblasts drug effects, ATP Citrate (pro-S)-Lyase metabolism, ATP Citrate (pro-S)-Lyase genetics, Fibrosis metabolism, Cell Differentiation drug effects, Histones metabolism, Smad2 Protein metabolism, Smad2 Protein genetics
- Abstract
Differentiation of cardiac fibroblasts to myofibroblasts is necessary for matrix remodeling and fibrosis in heart failure. We previously reported that mitochondrial calcium signaling drives α-ketoglutarate-dependent histone demethylation, promoting myofibroblast formation. Here we investigate the role of ATP-citrate lyase (ACLY), a key enzyme for acetyl-CoA biosynthesis, in histone acetylation regulating myofibroblast fate and persistence in cardiac fibrosis. We show that inactivation of ACLY prevents myofibroblast differentiation and reverses myofibroblasts towards quiescence. Genetic deletion of Acly in post-activated myofibroblasts prevents fibrosis and preserves cardiac function in pressure-overload heart failure. TGFβ stimulation enhances ACLY nuclear localization and ACLY-SMAD2/3 interaction, and increases H3K27ac at fibrotic gene loci. Pharmacological inhibition of ACLY or forced nuclear expression of a dominant-negative ACLY mutant prevents myofibroblast formation and H3K27ac. Our data indicate that nuclear ACLY activity is necessary for myofibroblast differentiation and persistence by maintaining histone acetylation at TGFβ-induced myofibroblast genes. These findings provide targets to prevent and reverse pathological fibrosis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
24. Integrating Clinical Phenotype With Multiomics Analyses of Human Cardiac Tissue Unveils Divergent Metabolic Remodeling in Genotype-Positive and Genotype-Negative Patients With Hypertrophic Cardiomyopathy.
- Author
-
Nollet EE, Schuldt M, Sequeira V, Binek A, Pham TV, Schoonvelde SAC, Jansen M, Schomakers BV, van Weeghel M, Vaz FM, Houtkooper RH, Van Eyk JE, Jimenez CR, Michels M, Bedi KC Jr, Margulies KB, Dos Remedios CG, Kuster DWD, and van der Velden J
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Myocardium metabolism, Myocardium pathology, Metabolomics, Proteomics, Lipidomics, Lipid Metabolism genetics, Sarcomeres metabolism, Sarcomeres genetics, Energy Metabolism genetics, Aged, Multiomics, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Cardiomyopathy, Hypertrophic pathology, Genotype, Phenotype
- Abstract
Background: Hypertrophic cardiomyopathy (HCM) is caused by sarcomere gene mutations (genotype-positive HCM) in ≈50% of patients and occurs in the absence of mutations (genotype-negative HCM) in the other half of patients. We explored how alterations in the metabolomic and lipidomic landscape are involved in cardiac remodeling in both patient groups., Methods: We performed proteomics, metabolomics, and lipidomics on myectomy samples (genotype-positive N=19; genotype-negative N=22; and genotype unknown N=6) from clinically well-phenotyped patients with HCM and on cardiac tissue samples from sex- and age-matched and body mass index-matched nonfailing donors (N=20). These data sets were integrated to comprehensively map changes in lipid-handling and energy metabolism pathways. By linking metabolomic and lipidomic data to variability in clinical data, we explored patient group-specific associations between cardiac and metabolic remodeling., Results: HCM myectomy samples exhibited (1) increased glucose and glycogen metabolism, (2) downregulation of fatty acid oxidation, and (3) reduced ceramide formation and lipid storage. In genotype-negative patients, septal hypertrophy and diastolic dysfunction correlated with lowering of acylcarnitines, redox metabolites, amino acids, pentose phosphate pathway intermediates, purines, and pyrimidines. In contrast, redox metabolites, amino acids, pentose phosphate pathway intermediates, purines, and pyrimidines were positively associated with septal hypertrophy and diastolic impairment in genotype-positive patients., Conclusions: We provide novel insights into both general and genotype-specific metabolic changes in HCM. Distinct metabolic alterations underlie cardiac disease progression in genotype-negative and genotype-positive patients with HCM., Competing Interests: Disclosures Dr Margulies is a consultant for Bristol Myers Squibb and receives research support from Amgen.
- Published
- 2024
- Full Text
- View/download PDF
25. Perinuclear damage from nuclear envelope deterioration elicits stress responses that contribute to LMNA cardiomyopathy.
- Author
-
Sikder K, Phillips E, Zhong Z, Wang N, Saunders J, Mothy D, Kossenkov A, Schneider T, Nichtova Z, Csordas G, Margulies KB, and Choi JC
- Subjects
- Animals, Mice, Autophagy, Stress, Physiological, Disease Models, Animal, Endoplasmic Reticulum Stress, Golgi Apparatus metabolism, Mice, Knockout, Lamin Type A metabolism, Lamin Type A genetics, Nuclear Envelope metabolism, Cardiomyopathies metabolism, Cardiomyopathies etiology, Cardiomyopathies pathology, Cardiomyopathies genetics, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology
- Abstract
Mutations in the LMNA gene encoding lamins A/C cause an array of tissue-selective diseases, with the heart being the most commonly affected organ. Despite progress in understanding the perturbations emanating from LMNA mutations, an integrative understanding of the pathogenesis underlying cardiac dysfunction remains elusive. Using a novel conditional deletion model capable of translatome profiling, we observed that cardiomyocyte-specific Lmna deletion in adult mice led to rapid cardiomyopathy with pathological remodeling. Before cardiac dysfunction, Lmna -deleted cardiomyocytes displayed nuclear abnormalities, Golgi dilation/fragmentation, and CREB3-mediated stress activation. Translatome profiling identified MED25 activation, a transcriptional cofactor that regulates Golgi stress. Autophagy is disrupted in the hearts of these mice, which can be recapitulated by disrupting the Golgi. Systemic administration of modulators of autophagy or ER stress significantly delayed cardiac dysfunction and prolonged survival. These studies support a hypothesis wherein stress responses emanating from the perinuclear space contribute to the LMNA cardiomyopathy development.
- Published
- 2024
- Full Text
- View/download PDF
26. Epistasis regulates genetic control of cardiac hypertrophy.
- Author
-
Wang Q, Tang TM, Youlton N, Weldy CS, Kenney AM, Ronen O, Weston Hughes J, Chin ET, Sutton SC, Agarwal A, Li X, Behr M, Kumbier K, Moravec CS, Wilson Tang WH, Margulies KB, Cappola TP, Butte AJ, Arnaout R, Brown JB, Priest JR, Parikh VN, Yu B, and Ashley EA
- Abstract
The combinatorial effect of genetic variants is often assumed to be additive. Although genetic variation can clearly interact non-additively, methods to uncover epistatic relationships remain in their infancy. We develop low-signal signed iterative random forests to elucidate the complex genetic architecture of cardiac hypertrophy. We derive deep learning-based estimates of left ventricular mass from the cardiac MRI scans of 29,661 individuals enrolled in the UK Biobank. We report epistatic genetic variation including variants close to CCDC141 , IGF1R , TTN , and TNKS. Several loci where variants were deemed insignificant in univariate genome-wide association analyses are identified. Functional genomic and integrative enrichment analyses reveal a complex gene regulatory network in which genes mapped from these loci share biological processes and myogenic regulatory factors. Through a network analysis of transcriptomic data from 313 explanted human hearts, we found strong gene co-expression correlations between these statistical epistasis contributors in healthy hearts and a significant connectivity decrease in failing hearts. We assess causality of epistatic effects via RNA silencing of gene-gene interactions in human induced pluripotent stem cell-derived cardiomyocytes. Finally, single-cell morphology analysis using a novel high-throughput microfluidic system shows that cardiomyocyte hypertrophy is non-additively modifiable by specific pairwise interactions between CCDC141 and both TTN and IGF1R . Our results expand the scope of genetic regulation of cardiac structure to epistasis.
- Published
- 2024
- Full Text
- View/download PDF
27. Distinct cytoskeletal regulators of mechanical memory in cardiac fibroblasts and cardiomyocytes.
- Author
-
Bouhrira N, Vite A, and Margulies KB
- Subjects
- Humans, Actins metabolism, Mechanotransduction, Cellular, Cell Differentiation physiology, Fibroblasts metabolism, Myocytes, Cardiac metabolism, Induced Pluripotent Stem Cells
- Abstract
Recognizing that cells "feel" and respond to their mechanical environment, recent studies demonstrate that many cells exhibit a phenomenon of "mechanical memory" in which features induced by prior mechanical cues persist after the mechanical stimulus has ceased. While there is a general recognition that different cell types exhibit different responses to changes in extracellular matrix stiffening, the phenomenon of mechanical memory within myocardial cell types has received little attention to date. To probe the dynamics of mechanical memory in cardiac fibroblasts (CFs) and cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CMs), we employed a magnetorheological elastomer (MRE) cell culture substrate with tunable and reversible stiffness spanning the range from normal to diseased myocardium. In CFs, using increased cell area and increases in α-smooth muscle actin as markers of cellular responses to matrix stiffening, we found that induction of mechanical memory required seven days of stiff priming. Both induction and maintenance of persistent CF activation were blocked with the F-actin inhibitor cytochalasin D, while inhibitors of microtubule detyrosination had no impact on CFs. In iPSC-CMs, mechanical memory was invoked after only 24 h of stiff priming. Moreover, mechanical memory induction and maintenance were microtubule-dependent in CMs with no dependence on F-actin. Overall, these results identify the distinct temporal dynamics of mechanical memory in CFs and iPSC-CMs with different cytoskeletal mediators responsible for inducing and maintaining the stiffness-activated phenotype. Due to its flexibility, this model is broadly applicable to future studies interrogating mechanotransduction and mechanical memory in the heart and might inform strategies for attenuating the impact of load-induced pathology and excess myocardial stiffness., (© 2024. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany.)
- Published
- 2024
- Full Text
- View/download PDF
28. The HFSA Advanced Heart Failure and Transplant Cardiology Fellowship Consensus Conference.
- Author
-
Drazner MH, Ambardekar AV, Berlacher K, Blumer V, Chatur S, Cheng R, Cheng RK, Grandin EW, Gorodeski EZ, Kataria R, Katz JN, Kittleson MM, Krishnamoorthy A, Lala A, Lenneman AJ, Lohr NL, Margulies KB, Mentz RJ, Reza N, Wilcox J, Youmans QR, Zieroth S, and Teerlink JR
- Subjects
- Humans, Fellowships and Scholarships, Quality of Life, Consensus, Heart Failure diagnosis, Heart Failure surgery, Cardiology
- Abstract
There is waning interest among cardiology trainees in pursuing an Advanced Heart Failure/Transplant Cardiology (AHFTC) fellowship as evidenced by fewer applicants in the National Resident Matching Program match to this specialty. This trend has generated considerable attention across the heart failure community. In response, the Heart Failure Society of America convened the AHFTC Fellowship Task Force with a charge to develop strategies to increase the value proposition of an AHFTC fellowship. Subsequently, the HFSA sponsored the AHFTC Fellowship Consensus Conference April 26-27, 2023. Before the conference, interviews of 44 expert stakeholders diverse across geography, site of practice (traditional academic medical center or other centers), specialty/area of expertise, sex, and stage of career were conducted virtually. Based on these interviews, potential solutions to address the declining interest in AHFTC fellowship were categorized into five themes: (1) alternative training pathways, (2) regulatory and compensation, (3) educational improvements, (4) exposure and marketing for pipeline development, and (5) quality of life and mental health. These themes provided structure to the deliberations of the AHFTC Fellowship Consensus Conference. The recommendations from the Consensus Conference were subsequently presented to the HFSA Board of Directors to inform strategic plans and interventions. The HFSA Board of Directors later reviewed and approved submission of this document. The purpose of this communication is to provide the HF community with an update summarizing the processes used and concepts that emerged from the work of the HFSA AHFTC Fellowship Task Force and Consensus Conference., Competing Interests: Disclosures The authors have no conflicts of interest to declare., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
29. Failing to Make the Grade: Conventional Cardiac Allograft Rejection Grading Criteria Are Inadequate for Predicting Rejection Severity.
- Author
-
Arabyarmohammadi S, Yuan C, Viswanathan VS, Lal P, Feldman MD, Fu P, Margulies KB, Madabhushi A, and Peyster EG
- Subjects
- Humans, Myocardium pathology, Heart, Allografts, Graft Rejection diagnosis, Biopsy, Heart Transplantation adverse effects, Heart Failure pathology
- Abstract
Background: Cardiac allograft rejection is the leading cause of early graft failure and is a major focus of postheart transplant patient care. While histological grading of endomyocardial biopsy samples remains the diagnostic standard for acute rejection, this standard has limited diagnostic accuracy. Discordance between biopsy rejection grade and patient clinical trajectory frequently leads to both overtreatment of indolent processes and delayed treatment of aggressive ones, spurring the need to investigate the adequacy of the current histological criteria for assessing clinically important rejection outcomes., Methods: N=2900 endomyocardial biopsy images were assigned a rejection grade label (high versus low grade) and a clinical trajectory label (evident versus silent rejection). Using an image analysis approach, n=370 quantitative morphology features describing the lymphocytes and stroma were extracted from each slide. Two models were constructed to compare the subset of features associated with rejection grades versus those associated with clinical trajectories. A proof-of-principle machine learning pipeline-the cardiac allograft rejection evaluator-was then developed to test the feasibility of identifying the clinical severity of a rejection event., Results: The histopathologic findings associated with conventional rejection grades differ substantially from those associated with clinically evident allograft injury. Quantitative assessment of a small set of well-defined morphological features can be leveraged to more accurately reflect the severity of rejection compared with that achieved by the International Society of Heart and Lung Transplantation grades., Conclusions: Conventional endomyocardial samples contain morphological information that enables accurate identification of clinically evident rejection events, and this information is incompletely captured by the current, guideline-endorsed, rejection grading criteria., Competing Interests: Disclosures Dr Feldman is an equity holder and has technology licensed to both Elucid Bioimaging and Inspirata Inc. Dr Feldman is a scientific advisory consultant for Inspirata Inc and is on its scientific advisory board. Dr Feldman is also a consultant for Phillips Healthcare, XFIN, and Verbio. Dr Margulies holds research grants from Amgen and serves as a scientific consultant/advisory board member of Bristol Myers Squibb. Dr Madabhushi is an equity holder in Elucid Bioimaging, Inspirata Inc, and Picture Health. He has served as a scientific advisory board member of Picture Health, SimbioSys, and Aiforia Inc. He also has sponsored research agreements with AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, and Eli Lilly. His technology has been licensed to Elucid Bioimaging and Picture Health. He is also involved in 3 different National Institutes of Health R01 grants with Inspirata Inc.
- Published
- 2024
- Full Text
- View/download PDF
30. Truncated titin protein in dilated cardiomyopathy incorporates into the sarcomere and transmits force.
- Author
-
McAfee Q, Caporizzo MA, Uchida K, Bedi KC Jr, Margulies KB, Arany Z, and Prosser BL
- Subjects
- Humans, Connectin genetics, Connectin metabolism, Sarcomeres metabolism, Muscle Proteins metabolism, Cardiomyopathy, Dilated genetics, Cardiomyopathy, Dilated metabolism
- Published
- 2024
- Full Text
- View/download PDF
31. Epistasis regulates genetic control of cardiac hypertrophy.
- Author
-
Wang Q, Tang TM, Youlton N, Weldy CS, Kenney AM, Ronen O, Hughes JW, Chin ET, Sutton SC, Agarwal A, Li X, Behr M, Kumbier K, Moravec CS, Tang WHW, Margulies KB, Cappola TP, Butte AJ, Arnaout R, Brown JB, Priest JR, Parikh VN, Yu B, and Ashley EA
- Abstract
The combinatorial effect of genetic variants is often assumed to be additive. Although genetic variation can clearly interact non-additively, methods to uncover epistatic relationships remain in their infancy. We develop low-signal signed iterative random forests to elucidate the complex genetic architecture of cardiac hypertrophy. We derive deep learning-based estimates of left ventricular mass from the cardiac MRI scans of 29,661 individuals enrolled in the UK Biobank. We report epistatic genetic variation including variants close to CCDC141 , IGF1R , TTN , and TNKS. Several loci not prioritized by univariate genome-wide association analysis are identified. Functional genomic and integrative enrichment analyses reveal a complex gene regulatory network in which genes mapped from these loci share biological processes and myogenic regulatory factors. Through a network analysis of transcriptomic data from 313 explanted human hearts, we show that these interactions are preserved at the level of the cardiac transcriptome. We assess causality of epistatic effects via RNA silencing of gene-gene interactions in human induced pluripotent stem cell-derived cardiomyocytes. Finally, single-cell morphology analysis using a novel high-throughput microfluidic system shows that cardiomyocyte hypertrophy is non-additively modifiable by specific pairwise interactions between CCDC141 and both TTN and IGF1R . Our results expand the scope of genetic regulation of cardiac structure to epistasis., Competing Interests: Competing interests E.A.A. is a Founder of Personalis, Deepcell, Svexa, RCD Co, and Parameter Health; Advisor to Oxford Nanopore, SequenceBio, and Pacific Biosciences; and a non-executive director for AstraZeneca. C.S.W. is a consultant for Tensixteen Bio and Renovacor. V.N.P. is an SAB member for and receives research support from BioMarin, Inc, and is a consultant for Constantiam, Inc. and viz.ai. The remaining authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
32. Discovery adductomics provides a comprehensive portrait of tissue-, age- and sex-specific DNA modifications in rodents and humans.
- Author
-
Guilbaud A, Ghanegolmohammadi F, Wang Y, Leng J, Kreymerman A, Gamboa Varela J, Garbern J, Elwell H, Cao F, Ricci-Blair EM, Liang C, Balamkundu S, Vidoudez C, DeMott MS, Bedi K, Margulies KB, Bennett DA, Palmer AA, Barkley-Levenson A, Lee RT, and Dedon PC
- Subjects
- Animals, Female, Humans, Male, Rats, Chromatography, Liquid methods, Rodentia, Tandem Mass Spectrometry methods, DNA chemistry, DNA Adducts genetics
- Abstract
DNA damage causes genomic instability underlying many diseases, with traditional analytical approaches providing minimal insight into the spectrum of DNA lesions in vivo. Here we used untargeted chromatography-coupled tandem mass spectrometry-based adductomics (LC-MS/MS) to begin to define the landscape of DNA modifications in rat and human tissues. A basis set of 114 putative DNA adducts was identified in heart, liver, brain, and kidney in 1-26-month-old rats and 111 in human heart and brain by 'stepped MRM' LC-MS/MS. Subsequent targeted analysis of these species revealed species-, tissue-, age- and sex-biases. Structural characterization of 10 selected adductomic signals as known DNA modifications validated the method and established confidence in the DNA origins of the signals. Along with strong tissue biases, we observed significant age-dependence for 36 adducts, including N2-CMdG, 5-HMdC and 8-Oxo-dG in rats and 1,N6-ϵdA in human heart, as well as sex biases for 67 adducts in rat tissues. These results demonstrate the potential of adductomics for discovering the true spectrum of disease-driving DNA adducts. Our dataset of 114 putative adducts serves as a resource for characterizing dozens of new forms of DNA damage, defining mechanisms of their formation and repair, and developing them as biomarkers of aging and disease., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2023
- Full Text
- View/download PDF
33. New Cardiotoxicity Risk Assessment Guidelines: Searching for Validation.
- Author
-
Rashed ER and Margulies KB
- Abstract
Competing Interests: Dr Margulies has received sponsored research support from Amgen, Inc and Lexicon Pharmaceuticals; and is a scientific advisory board member for Bristol Myers Squibb. Dr Rashed has reported that she has no relationships relevant to the contents of this paper to disclose.
- Published
- 2023
- Full Text
- View/download PDF
34. Early Increases in Blood Pressure and Major Adverse Cardiovascular Events in Patients With Renal Cell Carcinoma and Thyroid Cancer Treated With VEGFR TKIs.
- Author
-
Narayan V, Liu T, Song Y, Mitchell J, Sicks J, Gareen I, Sun L, Denduluri S, Fisher C, Manikowski J, Wojtowicz M, Vadakara J, Haas N, Margulies KB, and Ky B
- Subjects
- Humans, Blood Pressure, Retrospective Studies, Protein Kinase Inhibitors adverse effects, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell epidemiology, Kidney Neoplasms drug therapy, Kidney Neoplasms epidemiology, Thyroid Neoplasms drug therapy, Thyroid Neoplasms epidemiology, Hypertension chemically induced, Hypertension epidemiology, Hypertension drug therapy
- Abstract
Background: Although VEGFR tyrosine kinase inhibitors (TKIs) are a preferred systemic treatment approach for patients with advanced renal cell carcinoma (RCC) and thyroid carcinoma (TC), treatment-related cardiovascular (CV) toxicity is an important contributor to morbidity. However, the clinical risk assessment and impact of CV toxicities, including early significant hypertension, among real-world advanced cancer populations receiving VEGFR TKI therapies remain understudied., Methods: In a multicenter, retrospective cohort study across 3 large and diverse US health systems, we characterized baseline hypertension and CV comorbidity in patients with RCC and those with TC who are newly initiating VEGFR TKI therapy. We also evaluated baseline patient-, treatment-, and disease-related factors associated with the risk for treatment-related early hypertension (within 6 weeks of TKI initiation) and major adverse CV events (MACE), accounting for the competing risk of death in an advanced cancer population, after VEGFR TKI initiation., Results: Between 2008 and 2020, 987 patients (80.3% with RCC, 19.7% with TC) initiated VEGFR TKI therapy. The baseline prevalence of hypertension was high (61.5% and 53.6% in patients with RCC and TC, respectively). Adverse CV events, including heart failure and cerebrovascular accident, were common (occurring in 14.9% of patients) and frequently occurred early (46.3% occurred within 1 year of VEGFR TKI initiation). Baseline hypertension and Black race were the primary clinical factors associated with increased acute hypertensive risk within 6 weeks of VEGFR TKI initiation. However, early significant "on-treatment" hypertension was not associated with MACE., Conclusions: These multicenter, real-world findings indicate that hypertensive and CV morbidities are highly prevalent among patients initiating VEGFR TKI therapies, and baseline hypertension and Black race represent the primary clinical factors associated with VEGFR TKI-related early significant hypertension. However, early on-treatment hypertension was not associated with MACE, and cancer-specific CV risk algorithms may be warranted for patients initiating VEGFR TKIs.
- Published
- 2023
- Full Text
- View/download PDF
35. Adult human cardiomyocyte mechanics in osteogenesis imperfecta.
- Author
-
Lee BW, Caporizzo MA, Chen CY, Bedi KC Jr, Peyster EG, Prosser BL, Margulies KB, and Vite A
- Subjects
- Humans, Adult, Myocytes, Cardiac metabolism, Collagen metabolism, Collagen Type I metabolism, Extracellular Matrix metabolism, Osteogenesis, Osteogenesis Imperfecta metabolism, Osteogenesis Imperfecta pathology
- Abstract
Osteogenesis imperfecta (OI) is an extracellular matrix disorder characterized by defects in collagen-1 transport or synthesis, resulting in bone abnormalities. Although reduced collagen in OI hearts has been associated with reduced myocardial stiffness and left ventricular remodeling, its impact on cardiomyocyte (CM) function has not been studied. Here, we explore the tissue-level and CM-level properties of a heart from a deceased organ donor with OI type I. Proteomics and histology confirmed strikingly low expression of collagen 1. Trabecular stretch confirmed low stiffness on the tissue level. However, CMs retained normal viscoelastic properties as revealed by nanoindentation. Interestingly, OI CMs were hypercontractile relative to nonfailing controls after 24 h of culture. In response to 48 h of culture on surfaces with physiological (10 kPa) and pathological (50 kPa) stiffness, OI CMs demonstrated a greater reduction in contractility than nonfailing CMs, suggesting that OI CMs may have an impaired stress response. Levels of detyrosinated α-tubulin, known to be responsive to extracellular stiffness, were reduced in OI CMs. Together these data confirm multiple CM-level adaptations to low stiffness that extend our understanding of OI in the heart and how CMs respond to extracellular stiffness. NEW & NOTEWORTHY In a rare donation of a heart from an individual with osteogenesis imperfecta (OI), we explored cardiomyocyte (CM) adaptations to low stiffness. This represents the first assessment of cardiomyocyte mechanics in OI. The data reveal the hypercontractility of OI CMs with rapid rundown when exposed to acute stiffness challenges, extending our understanding of OI. These data demonstrate that the impact of OI on myocardial mechanics includes cardiomyocyte adaptations beyond known direct effects on the extracellular matrix.
- Published
- 2023
- Full Text
- View/download PDF
36. Functional Impact of Alternative Metabolic Substrates in Failing Human Cardiomyocytes.
- Author
-
Vite A, Matsuura TR, Bedi KC, Flam EL, Arany Z, Kelly DP, and Margulies KB
- Abstract
Recent studies suggest that metabolic dysregulation in patients with heart failure might contribute to myocardial contractile dysfunction. To understand the correlation between function and energy metabolism, we studied the impact of different fuel substrates on human nonfailing or failing cardiomyocytes. Consistent with the concept of metabolic flexibility, nonfailing myocytes exhibited excellent contractility in all fuels provided. However, impaired contractility was observed in failing myocytes when carbohydrates alone were used but was improved when additional substrates were added. This study demonstrates the functional significance of fuel utilization shifts in failing human cardiomyocytes., Competing Interests: This project was funded by the National Institutes of Health (NIH) (R01 HL128349 and R01 HL151345, to Dr Kelly) and the Gund Family Fund at the University of Pennsylvania (to Dr Margulies). The procurement of human heart tissue was enabled by grants from NIH (R01 HL149891, to Dr Margulies). The authors have reported that they have no relationships relevant to the contents of this paper to disclose., (© 2024 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
37. Biomechanical Impact of Pathogenic MYBPC3 Truncation Variant Revealed by Dynamically Tuning In Vitro Afterload.
- Author
-
Ramachandran A, Livingston CE, Vite A, Corbin EA, Bennett AI, Turner KT, Lee BW, Lam CK, Wu JC, and Margulies KB
- Subjects
- Humans, Mutation, Heart, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic metabolism, Pluripotent Stem Cells metabolism
- Abstract
Engineered cardiac microtissues were fabricated using pluripotent stem cells with a hypertrophic cardiomyopathy associated c. 2827 C>T; p.R943x truncation variant in myosin binding protein C (MYBPC3
+/- ). Microtissues were mounted on iron-incorporated cantilevers, allowing manipulations of cantilever stiffness using magnets, enabling examination of how in vitro afterload affects contractility. MYPBC3+/- microtissues developed augmented force, work, and power when cultured with increased in vitro afterload when compared with isogenic controls in which the MYBPC3 mutation had been corrected (MYPBC3+/+ (ed)), but weaker contractility when cultured with lower in vitro afterload. After initial tissue maturation, MYPBC3+/- CMTs exhibited increased force, work, and power in response to both acute and sustained increases of in vitro afterload. Together, these studies demonstrate that extrinsic biomechanical challenges potentiate genetically-driven intrinsic increases in contractility that may contribute to clinical disease progression in patients with HCM due to hypercontractile MYBPC3 variants., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2023
- Full Text
- View/download PDF
38. Tissue Rings PLOD Out a Second Hit in Becker Muscular Dystrophy.
- Author
-
Lee BW and Margulies KB
- Abstract
Competing Interests: The authors have reported that they have no relationships relevant to the contents of this paper to disclose.
- Published
- 2023
- Full Text
- View/download PDF
39. Right Ventricular Sarcomere Contractile Depression and the Role of Thick Filament Activation in Human Heart Failure With Pulmonary Hypertension.
- Author
-
Jani V, Aslam MI, Fenwick AJ, Ma W, Gong H, Milburn G, Nissen D, Cubero Salazar IM, Hanselman O, Mukherjee M, Halushka MK, Margulies KB, Campbell KS, Irving TC, Kass DA, and Hsu S
- Subjects
- Humans, Sarcomeres, Calcium, Depression, Stroke Volume, Myocytes, Cardiac, Ventricular Function, Right physiology, Hypertension, Pulmonary, Heart Failure, Ventricular Dysfunction, Right
- Abstract
Background: Right ventricular (RV) contractile dysfunction commonly occurs and worsens outcomes in patients with heart failure with reduced ejection fraction and pulmonary hypertension (HFrEF-PH). However, such dysfunction often goes undetected by standard clinical RV indices, raising concerns that they may not reflect aspects of underlying myocyte dysfunction. We thus sought to characterize RV myocyte contractile depression in HFrEF-PH, identify those components reflected by clinical RV indices, and uncover underlying biophysical mechanisms., Methods: Resting, calcium-, and load-dependent mechanics were prospectively studied in permeabilized RV cardiomyocytes isolated from explanted hearts from 23 patients with HFrEF-PH undergoing cardiac transplantation and 9 organ donor controls., Results: Unsupervised machine learning using myocyte mechanical data with the highest variance yielded 2 HFrEF-PH subgroups that in turn mapped to patients with decompensated or compensated clinical RV function. This correspondence was driven by reduced calcium-activated isometric tension in decompensated clinical RV function, whereas surprisingly, many other major myocyte contractile measures including peak power and myocyte active stiffness were similarly depressed in both groups. Similar results were obtained when subgroups were first defined by clinical indices, and then myocyte mechanical properties in each group compared. To test the role of thick filament defects, myofibrillar structure was assessed by x-ray diffraction of muscle fibers. This revealed more myosin heads associated with the thick filament backbone in decompensated clinical RV function, but not compensated clinical RV function, as compared with controls. This corresponded to reduced myosin ATP turnover in decompensated clinical RV function myocytes, indicating less myosin in a crossbridge-ready disordered-relaxed (DRX) state. Altering DRX proportion (%DRX) affected peak calcium-activated tension in the patient groups differently, depending on their basal %DRX, highlighting potential roles for precision-guided therapeutics. Last, increasing myocyte preload (sarcomere length) increased %DRX 1.5-fold in controls but only 1.2-fold in both HFrEF-PH groups, revealing a novel mechanism for reduced myocyte active stiffness and by extension Frank-Starling reserve in human heart failure., Conclusions: Although there are many RV myocyte contractile deficits in HFrEF-PH, commonly used clinical indices only detect reduced isometric calcium-stimulated force, which is related to deficits in basal and recruitable %DRX myosin. Our results support use of therapies to increase %DRX and enhance length-dependent recruitment of DRX myosin heads in such patients., Competing Interests: Disclosures T.C.I. provides consulting services to Edgewise Therapeutics Inc and receives collaborative research funding from Bristol Myers Squibb Inc, but such work is unrelated to the content of this article. The other authors report no conflicts.
- Published
- 2023
- Full Text
- View/download PDF
40. Perinuclear damage from nuclear envelope deterioration elicits stress responses that contribute to LMNA cardiomyopathy.
- Author
-
Sikder K, Phillips E, Zhong Z, Wang N, Saunders J, Mothy D, Kossenkov A, Schneider T, Nichtova Z, Csordas G, Margulies KB, and Choi JC
- Abstract
Mutations in the LMNA gene encoding nuclear lamins A/C cause a diverse array of tissue-selective diseases, with the heart being the most commonly affected organ. Despite progress in understanding the molecular perturbations emanating from LMNA mutations, an integrative understanding of the pathogenesis leading to cardiac dysfunction remains elusive. Using a novel cell-type specific Lmna deletion mouse model capable of translatome profiling, we found that cardiomyocyte-specific Lmna deletion in adult mice led to rapid cardiomyopathy with pathological remodeling. Prior to the onset of cardiac dysfunction, lamin A/C-depleted cardiomyocytes displayed nuclear envelope deterioration, golgi dilation/fragmentation, and CREB3-mediated golgi stress activation. Translatome profiling identified upregulation of Med25, a transcriptional co-factor that can selectively dampen UPR axes. Autophagy is disrupted in the hearts of these mice, which can be recapitulated by disrupting the golgi or inducing nuclear damage by increased matrix stiffness. Systemic administration of pharmacological modulators of autophagy or ER stress significantly improved the cardiac function. These studies support a hypothesis wherein stress responses emanating from the perinuclear space contribute to the development of LMNA cardiomyopathy., Teaser: Interplay of stress responses underlying the development of LMNA cardiomyopathy.
- Published
- 2023
- Full Text
- View/download PDF
41. Sustained alternate-day fasting potentiates doxorubicin cardiotoxicity.
- Author
-
Ozcan M, Guo Z, Valenzuela Ripoll C, Diab A, Picataggi A, Rawnsley D, Lotfinaghsh A, Bergom C, Szymanski J, Hwang D, Asnani A, Kosiborod M, Zheng J, Hayashi RJ, Woodard PK, Kovacs A, Margulies KB, Schilling J, Razani B, Diwan A, and Javaheri A
- Subjects
- Mice, Humans, Animals, Doxorubicin toxicity, Autophagy, Myocytes, Cardiac metabolism, Fasting, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Lysosomes metabolism, Cardiotoxicity metabolism, Heart Failure chemically induced, Heart Failure metabolism
- Abstract
Fasting strategies are under active clinical investigation in patients receiving chemotherapy. Prior murine studies suggest that alternate-day fasting may attenuate doxorubicin cardiotoxicity and stimulate nuclear translocation of transcription factor EB (TFEB), a master regulator of autophagy and lysosomal biogenesis. In this study, human heart tissue from patients with doxorubicin-induced heart failure demonstrated increased nuclear TFEB protein. In mice treated with doxorubicin, alternate-day fasting or viral TFEB transduction increased mortality and impaired cardiac function. Mice randomized to alternate-day fasting plus doxorubicin exhibited increased TFEB nuclear translocation in the myocardium. When combined with doxorubicin, cardiomyocyte-specific TFEB overexpression provoked cardiac remodeling, while systemic TFEB overexpression increased growth differentiation factor 15 (GDF15) and caused heart failure and death. Cardiomyocyte TFEB knockout attenuated doxorubicin cardiotoxicity, while recombinant GDF15 was sufficient to cause cardiac atrophy. Our studies identify that both sustained alternate-day fasting and a TFEB/GDF15 pathway exacerbate doxorubicin cardiotoxicity., Competing Interests: Declaration of interests A.J. has a pending patent for fusion protein nanodiscs for the treatment of heart failure and eye disease, is a member of the scientific advisory board of Mobius Scientific, and receives research funding from AstraZeneca, unrelated to the studies in this manuscript. M.K. receives consulting fees/honoraria from AstraZeneca, Amgen, Sanofi-Aventis, Boehringer Ingelheim, Glytec, Merck, Janssen Pharmaceuticals, Novartis, Applied Therapeutics, Bayer Healthcare Pharmaceuticals, Eli Lilly and Company, and Vifor Pharma and research grants from AstraZeneca and Boehringer Ingelheim. A. Diwan reports consulting for clinical trials with Clario (previously ERT/Biomedical systems) and serves on the scientific advisory board for Dewpoint Therapeutics, which are not relevant to the current study., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
42. The evolving use of biomarkers in heart transplantation: Consensus of an expert panel.
- Author
-
Kobashigawa J, Hall S, Shah P, Fine B, Halloran P, Jackson AM, Khush KK, Margulies KB, Sani MM, Patel JK, Patel N, and Peyster E
- Subjects
- Humans, Quality of Life, Biomarkers, Graft Rejection diagnosis, Graft Rejection etiology, Kidney Transplantation, Heart Transplantation adverse effects
- Abstract
In heart transplantation, the use of biomarkers to detect the risk of rejection has been evolving. In this setting, it is becoming less clear as to what is the most reliable test or combination of tests to detect rejection and assess the state of the alloimmune response. Therefore, a virtual expert panel was organized in heart and kidney transplantation to evaluate emerging diagnostics and how they may be best utilized to monitor and manage transplant patients. This manuscript covers the heart content of the conference and is a work product of the American Society of Transplantation's Thoracic and Critical Care Community of Practice. This paper reviews currently available and emerging diagnostic assays and defines the unmet needs for biomarkers in heart transplantation. Highlights of the in-depth discussions among conference participants that led to development of consensus statements are included. This conference should serve as a platform to further build consensus within the heart transplant community regarding the optimal framework to implement biomarkers into management protocols and to improve biomarker development, validation and clinical utility. Ultimately, these biomarkers and novel diagnostics should improve outcomes and optimize quality of life for our transplant patients., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
43. Myocardial Metabolomics of Human Heart Failure With Preserved Ejection Fraction.
- Author
-
Hahn VS, Petucci C, Kim MS, Bedi KC Jr, Wang H, Mishra S, Koleini N, Yoo EJ, Margulies KB, Arany Z, Kelly DP, Kass DA, and Sharma K
- Subjects
- Humans, Stroke Volume, Myocardium metabolism, Obesity pathology, Fatty Acids, Heart Failure metabolism, Diabetes Mellitus pathology
- Abstract
Background: The human heart primarily metabolizes fatty acids, and this decreases as alternative fuel use rises in heart failure with reduced ejection fraction (HFrEF). Patients with severe obesity and diabetes are thought to have increased myocardial fatty acid metabolism, but whether this is found in those who also have heart failure with preserved ejection fraction (HFpEF) is unknown., Methods: Plasma and endomyocardial biopsies were obtained from HFpEF (n=38), HFrEF (n=30), and nonfailing donor controls (n=20). Quantitative targeted metabolomics measured organic acids, amino acids, and acylcarnitines in myocardium (72 metabolites) and plasma (69 metabolites). The results were integrated with reported RNA sequencing data. Metabolomics were analyzed using agnostic clustering tools, Kruskal-Wallis test with Dunn test, and machine learning., Results: Agnostic clustering of myocardial but not plasma metabolites separated disease groups. Despite more obesity and diabetes in HFpEF versus HFrEF (body mass index, 39.8 kg/m
2 versus 26.1 kg/m2 ; diabetes, 70% versus 30%; both P <0.0001), medium- and long-chain acylcarnitines (mostly metabolites of fatty acid oxidation) were markedly lower in myocardium from both heart failure groups versus control. In contrast, plasma levels were no different or higher than control. Gene expression linked to fatty acid metabolism was generally lower in HFpEF versus control. Myocardial pyruvate was higher in HFpEF whereas the tricarboxylic acid cycle intermediates succinate and fumarate were lower, as were several genes controlling glucose metabolism. Non-branched-chain and branched-chain amino acids (BCAA) were highest in HFpEF myocardium, yet downstream BCAA metabolites and genes controlling BCAA metabolism were lower. Ketone levels were higher in myocardium and plasma of patients with HFrEF but not HFpEF. HFpEF metabolomic-derived subgroups were differentiated by only a few differences in BCAA metabolites., Conclusions: Despite marked obesity and diabetes, HFpEF myocardium exhibited lower fatty acid metabolites compared with HFrEF. Ketones and metabolites of the tricarboxylic acid cycle and BCAA were also lower in HFpEF, suggesting insufficient use of alternative fuels. These differences were not detectable in plasma and challenge conventional views of myocardial fuel use in HFpEF with marked diabetes and obesity and suggest substantial fuel inflexibility in this syndrome.- Published
- 2023
- Full Text
- View/download PDF
44. Mitochondrial dysfunction in human hypertrophic cardiomyopathy is linked to cardiomyocyte architecture disruption and corrected by improving NADH-driven mitochondrial respiration.
- Author
-
Nollet EE, Duursma I, Rozenbaum A, Eggelbusch M, Wüst RCI, Schoonvelde SAC, Michels M, Jansen M, van der Wel NN, Bedi KC, Margulies KB, Nirschl J, Kuster DWD, and van der Velden J
- Subjects
- Humans, NAD genetics, Mutation, Mitochondria, Heart pathology, Respiration, Myocytes, Cardiac pathology, Cardiomyopathy, Hypertrophic genetics
- Abstract
Aims: Genetic hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomere protein-encoding genes (i.e. genotype-positive HCM). In an increasing number of patients, HCM occurs in the absence of a mutation (i.e. genotype-negative HCM). Mitochondrial dysfunction is thought to be a key driver of pathological remodelling in HCM. Reports of mitochondrial respiratory function and specific disease-modifying treatment options in patients with HCM are scarce., Methods and Results: Respirometry was performed on septal myectomy tissue from patients with HCM (n = 59) to evaluate oxidative phosphorylation and fatty acid oxidation. Mitochondrial dysfunction was most notably reflected by impaired NADH-linked respiration. In genotype-negative patients, but not genotype-positive patients, NADH-linked respiration was markedly depressed in patients with an indexed septal thickness ≥10 compared with <10. Mitochondrial dysfunction was not explained by reduced abundance or fragmentation of mitochondria, as evaluated by transmission electron microscopy. Rather, improper organization of mitochondria relative to myofibrils (expressed as a percentage of disorganized mitochondria) was strongly associated with mitochondrial dysfunction. Pre-incubation with the cardiolipin-stabilizing drug elamipretide and raising mitochondrial NAD+ levels both boosted NADH-linked respiration., Conclusion: Mitochondrial dysfunction is explained by cardiomyocyte architecture disruption and is linked to septal hypertrophy in genotype-negative HCM. Despite severe myocardial remodelling mitochondria were responsive to treatments aimed at restoring respiratory function, eliciting the mitochondria as a drug target to prevent and ameliorate cardiac disease in HCM. Mitochondria-targeting therapy may particularly benefit genotype-negative patients with HCM, given the tight link between mitochondrial impairment and septal thickening in this subpopulation., (© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2023
- Full Text
- View/download PDF
45. Med25 Limits Master Regulators That Govern Adipogenesis.
- Author
-
Saunders J, Sikder K, Phillips E, Ishwar A, Mothy D, Margulies KB, and Choi JC
- Subjects
- Animals, Mice, 3T3-L1 Cells, Cell Differentiation, Lipids pharmacology, Mediator Complex genetics, Mediator Complex metabolism, PPAR gamma metabolism, Transcription Factors metabolism, Adipogenesis genetics, Lamin Type A genetics, Lamin Type A metabolism
- Abstract
Mediator 25 (Med25) is a member of the mediator complex that relays signals from transcription factors to the RNA polymerase II machinery. Multiple transcription factors, particularly those involved in lipid metabolism, utilize the mediator complex, but how Med25 is involved in this context is unclear. We previously identified Med25 in a translatome screen of adult cardiomyocytes (CMs) in a novel cell type-specific model of LMNA cardiomyopathy. In this study, we show that Med25 upregulation is coincident with myocardial lipid accumulation. To ascertain the role of Med25 in lipid accumulation, we utilized iPSC-derived and neonatal CMs to recapitulate the in vivo phenotype by depleting lamins A and C (lamin A/C) in vitro. Although lamin A/C depletion elicits lipid accumulation, this effect appears to be mediated by divergent mechanisms dependent on the CM developmental state. To directly investigate Med25 in lipid accumulation, we induced adipogenesis in Med25 -silenced 3T3-L1 preadipocytes and detected enhanced lipid accumulation. Assessment of pertinent mediators driving adipogenesis revealed that C/EBPα and PPARγ are super-induced by Med25 silencing. Our results indicate that Med25 limits adipogenic potential by suppressing the levels of master regulators that govern adipogenesis. Furthermore, we caution the use of early-developmental-stage cardiomyocytes to model adult-stage cells, particularly for dissecting metabolic perturbations emanating from LMNA mutations.
- Published
- 2023
- Full Text
- View/download PDF
46. Single-nucleus RNA sequencing in ischemic cardiomyopathy reveals common transcriptional profile underlying end-stage heart failure.
- Author
-
Simonson B, Chaffin M, Hill MC, Atwa O, Guedira Y, Bhasin H, Hall AW, Hayat S, Baumgart S, Bedi KC Jr, Margulies KB, Klattenhoff CA, and Ellinor PT
- Subjects
- Humans, Endothelial Cells metabolism, Sequence Analysis, RNA, Myocardial Ischemia genetics, Myocardial Ischemia metabolism, Heart Failure genetics, Heart Failure metabolism, Cardiomyopathy, Dilated, Cardiomyopathies genetics
- Abstract
Ischemic cardiomyopathy (ICM) is the leading cause of heart failure worldwide, yet the cellular and molecular signature of this disease is largely unclear. Using single-nucleus RNA sequencing (snRNA-seq) and integrated computational analyses, we profile the transcriptomes of over 99,000 human cardiac nuclei from the non-infarct region of the left ventricle of 7 ICM transplant recipients and 8 non-failing (NF) controls. We find the cellular composition of the ischemic heart is significantly altered, with decreased cardiomyocytes and increased proportions of lymphatic, angiogenic, and arterial endothelial cells in patients with ICM. We show that there is increased LAMININ signaling from endothelial cells to other cell types in ICM compared with NF. Finally, we find that the transcriptional changes that occur in ICM are similar to those in hypertrophic and dilated cardiomyopathies and that the mining of these combined datasets can identify druggable genes that could be used to target end-stage heart failure., Competing Interests: Declaration of interests C.A.K. is an employee of Bayer US LLC (a subsidiary of Bayer AG) and may own stock in Bayer. S.H., S.B., and C.A.K. were full-time employees of Bayer when this work was performed. P.T.E. has received sponsored research support from Bayer AG, IBM Health, Bristol Myers Squibb, and Pfizer; he has also served on advisory boards or consulted for Bayer AG, MyoKardia, and Novartis. K.B.M. has research grant funding from Amgen, USA and has also served on advisory boards for MyoKardia, Bristol-Myers Squibb, and Pfizer., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
47. Apolipoprotein M Attenuates Anthracycline Cardiotoxicity and Lysosomal Injury.
- Author
-
Guo Z, Valenzuela Ripoll C, Picataggi A, Rawnsley DR, Ozcan M, Chirinos JA, Chendamarai E, Girardi A, Riehl T, Evie H, Diab A, Kovacs A, Hyrc K, Ma X, Asnani A, Shewale SV, Scherrer-Crosbie M, Cowart LA, Parks JS, Zhao L, Gordon D, Ramirez-Valle F, Margulies KB, Cappola TP, Desai AA, Pedersen LN, Bergom C, Stitziel NO, Rettig MP, DiPersio JF, Hajny S, Christoffersen C, Diwan A, and Javaheri A
- Abstract
Apolipoprotein M (ApoM) binds sphingosine-1-phosphate (S1P) and is inversely associated with mortality in human heart failure (HF). Here, we show that anthracyclines such as doxorubicin (Dox) reduce circulating ApoM in mice and humans, that ApoM is inversely associated with mortality in patients with anthracycline-induced heart failure, and ApoM heterozygosity in mice increases Dox-induced mortality. In the setting of Dox stress, our studies suggest ApoM can help sustain myocardial autophagic flux in a post-transcriptional manner, attenuate Dox cardiotoxicity, and prevent lysosomal injury., Competing Interests: Dr Javaheri was supported by R01HL155344 and K08HL138262 from the National Heart, Lung, and Blood Institute and by the Diabetes Research Center at Washington University in St Louis of the National Institutes of Health (NIH) under award number P30DK020579, as well as NIH grant P30DK056341 (Nutrition Obesity Research Center), and by the Children’s Discovery Institute of Washington University (MC-FR-2020-919) and St Louis Children’s Hospital. Dr Guo was supported by an American Heart Association Postdoctoral Fellowship (898679). Dr Diwan was supported by grants from the Department of Veterans Affairs (I01BX004235) and the NIH (HL107594, HL43431, and NS094692). Dr Scherrer-Crosbie is supported by R01HL130539 and R01HL131613. Dr Rawnsley was supported by training grant support from the NIH (T32007081). Dr Desai was supported by R01HL136603. Dr Bergom was supported by R01HL147884. Research reported in this publication was also supported by the National Cancer Institute of the NIH under award numbers R50CA211466 (Dr Rettig), R35CA210084 (Dr DiPersio), P01CA101937 (Dr DiPersio), and R01HL119962 (Dr Parks). Human heart tissue procurement was supported by the National Heart, Lung, and Blood Institute via R01HL105993 (Drs Margulies and Cappola). Drs Christoffersen and Hajny were supported by the Novo Nordisk Foundation (0053008 and NNF13OC0003898). Dr Stitziel was supported in part by R01HL131961, R01HL159171, P01HL151328, and UM1HG008853 and by the Foundation for Barnes-Jewish Hospital. We acknowledge support from the NIH Shared Instrumentation Grant (S10RR027552) for support through the Hope Center Neuroimaging Core, the Molecular Microbiology Imaging Facility, and the Advanced Imaging and Tissue Analysis Core of the Digestive Disease Research Core Center (DDRCC NIH P30DK052574) at Washington University School of Medicine. Dr Stitziel has received consulting fees from Sension Therapeutics and investigator-initiated research funding from Regeneron Pharmaceuticals unrelated to the content of this study. Dr Javaheri has a pending patent for fusion protein nanodiscs for the treatment of heart failure and eye diseases, receives research funding from AstraZeneca, and is on the Scientific Advisory Board of Mobius Scientific. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
48. Noncanonical Form of ERAD Regulates Cardiac Hypertrophy.
- Author
-
Blackwood EA, MacDonnell LF, Thuerauf DJ, Bilal AS, Murray VB, Bedi KC Jr, Margulies KB, and Glembotski CC
- Subjects
- Animals, Humans, Mice, Endoplasmic Reticulum metabolism, Myocytes, Cardiac metabolism, Unfolded Protein Response physiology, Cardiomegaly metabolism, Endoplasmic Reticulum-Associated Degradation physiology
- Abstract
Background: Cardiac hypertrophy increases demands on protein folding, which causes an accumulation of misfolded proteins in the endoplasmic reticulum (ER). These misfolded proteins can be removed by the adaptive retrotranslocation, polyubiquitylation, and a proteasome-mediated degradation process, ER-associated degradation (ERAD), which, as a biological process and rate, has not been studied in vivo. To investigate a role for ERAD in a pathophysiological model, we examined the function of the functional initiator of ERAD, valosin-containing protein-interacting membrane protein (VIMP), positing that VIMP would be adaptive in pathological cardiac hypertrophy in mice., Methods: We developed a new method involving cardiac myocyte-specific adeno-associated virus serovar 9-mediated expression of the canonical ERAD substrate, TCRα, to measure the rate of ERAD, ie, ERAD flux, in the heart in vivo. Adeno-associated virus serovar 9 was also used to either knock down or overexpress VIMP in the heart. Then mice were subjected to transverse aortic constriction to induce pressure overload-induced cardiac hypertrophy., Results: ERAD flux was slowed in both human heart failure and mice after transverse aortic constriction. Surprisingly, although VIMP adaptively contributes to ERAD in model cell lines, in the heart, VIMP knockdown increased ERAD and ameliorated transverse aortic constriction-induced cardiac hypertrophy. Coordinately, VIMP overexpression exacerbated cardiac hypertrophy, which was dependent on VIMP engaging in ERAD. Mechanistically, we found that the cytosolic protein kinase SGK1 (serum/glucocorticoid regulated kinase 1) is a major driver of pathological cardiac hypertrophy in mice subjected to transverse aortic constriction, and that VIMP knockdown decreased the levels of SGK1, which subsequently decreased cardiac pathology. We went on to show that although it is not an ER protein, and resides outside of the ER, SGK1 is degraded by ERAD in a noncanonical process we call ERAD-Out. Despite never having been in the ER, SGK1 is recognized as an ERAD substrate by the ERAD component DERLIN1, and uniquely in cardiac myocytes, VIMP displaces DERLIN1 from initiating ERAD, which decreased SGK1 degradation and promoted cardiac hypertrophy., Conclusions: ERAD-Out is a new preferentially favored noncanonical form of ERAD that mediates the degradation of SGK1 in cardiac myocytes, and in so doing is therefore an important determinant of how the heart responds to pathological stimuli, such as pressure overload.
- Published
- 2023
- Full Text
- View/download PDF
49. Comprehensive nutrient consumption estimation and metabolic profiling during ketogenic diet and relationship with myocardial glucose uptake on FDG-PET.
- Author
-
Selvaraj S, Seidelmann SB, Soni M, Bhattaru A, Margulies KB, Shah SH, Dugyala S, Qian C, Pryma DA, Arany Z, Kelly DP, Chirinos JA, and Bravo PE
- Subjects
- Humans, Female, Adult, Male, Positron-Emission Tomography methods, Nutrients, Glucose, Radiopharmaceuticals, Fluorodeoxyglucose F18, Diet, Ketogenic
- Abstract
Aims: The ketogenic diet (KD) is standard-of-care to achieve myocardial glucose suppression (MGS) for assessing inflammation using fluorine-18 fluorodeoxyglucose-positron emission tomography (FDG-PET). As KD protocols remain highly variable between centres (including estimation of nutrient intake by dietary logs for adequacy of dietary preparation), we aimed to assess the predictive utility of nutrient intake in achieving MGS., Methods and Results: Nineteen healthy participants underwent short-term KD, with FDG-PET performed after 1 and 3 days of KD (goal carbohydrate intake <20 g/day). Nutrient consumption was estimated from dietary logs using nutrition research software. The area under receiver operating characteristics (AUROC) of macronutrients (carbohydrate, fat, and protein intake) for predicting MGS was analysed. The association between 133 nutrients and 4 biomarkers [beta-hydroxybutyrate (BHB), non-esterified fatty acids, insulin, and glucagon] with myocardial glucose uptake was assessed using mixed effects regression with false discovery rate (FDR) correction. Median (25th-75th percentile) age was 29 (25-34) years, 47% were women, and 42% were non-white. Median (25th-75th percentile) carbohydrate intake (g) was 18.7 (13.1-30.7), 16.9 (10.4-28.7), and 21.1 (16.6-29.0) on Days 1-3. No macronutrient intake (carbohydrate, fat, or protein) predicted MGS (c-statistic 0.45, 0.53, 0.47, respectively). Of 133 nutrients and 4 biomarkers, only BHB was associated with myocardial glucose uptake after FDR correction (corrected P-value 0.003)., Conclusions: During highly supervised, short-term KD, approximately half of patients meet strict carbohydrate goals. Yet, in healthy volunteers, dietary review does not provide reassurance for adequacy of myocardial preparation since no clear thresholds for carbohydrate or fat intake reliably predict MGS., Competing Interests: Conflict of interest: S.S. receives research support from the Doris Duke Charitable Foundation (Physician Scientist Fellowship Award 2020061), the Measey Foundation, Institute for Translational Medicine and Therapeutics (Junior Investigator Preliminary/Feasibility Grant Program award and Translational Bio-Imaging Center award), and the American Society of Nuclear Cardiology (Institute for the Advancement of Nuclear Cardiology award). D.A.P. has received research grants from Siemens AG, 511 Pharma, Progenics Pharmaceuticals, Inc.; served as a research consultant for 511 Pharma, Progenics Pharmaceuticals, Inc., Actinium Pharmaceuticals, Inc., Ipsen; received clinical trial funding from Nordic Nanovector ASA. J.A.C. received University of Pennsylvania research grants from National Institutes of Health, American College of Radiology Network, American Heart Association, Fukuda Denshi, Bristol-Myers Squibb, Abbott, and Microsoft (not related to the present work); he has received payments for editorial roles from the American Heart Association and the American College of Cardiology; he has recently consulted for Bayer, Sanifit, Fukuda-Denshi, Bristol-Myers Squibb, JNJ, Edwards Life Sciences, Merck, Bionest, and the Galway-Mayo Institute of Technology (not related to the present work). The other authors report no conflict of interest., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2022. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
50. Genome-wide association and multi-trait analyses characterize the common genetic architecture of heart failure.
- Author
-
Levin MG, Tsao NL, Singhal P, Liu C, Vy HMT, Paranjpe I, Backman JD, Bellomo TR, Bone WP, Biddinger KJ, Hui Q, Dikilitas O, Satterfield BA, Yang Y, Morley MP, Bradford Y, Burke M, Reza N, Charest B, Judy RL, Puckelwartz MJ, Hakonarson H, Khan A, Kottyan LC, Kullo I, Luo Y, McNally EM, Rasmussen-Torvik LJ, Day SM, Do R, Phillips LS, Ellinor PT, Nadkarni GN, Ritchie MD, Arany Z, Cappola TP, Margulies KB, Aragam KG, Haggerty CM, Joseph J, Sun YV, Voight BF, and Damrauer SM
- Subjects
- Humans, Phenotype, Heart, Gene Expression Profiling, Polymorphism, Single Nucleotide, Genetic Predisposition to Disease, Genome-Wide Association Study methods, Heart Failure genetics
- Abstract
Heart failure is a leading cause of cardiovascular morbidity and mortality. However, the contribution of common genetic variation to heart failure risk has not been fully elucidated, particularly in comparison to other common cardiometabolic traits. We report a multi-ancestry genome-wide association study meta-analysis of all-cause heart failure including up to 115,150 cases and 1,550,331 controls of diverse genetic ancestry, identifying 47 risk loci. We also perform multivariate genome-wide association studies that integrate heart failure with related cardiac magnetic resonance imaging endophenotypes, identifying 61 risk loci. Gene-prioritization analyses including colocalization and transcriptome-wide association studies identify known and previously unreported candidate cardiomyopathy genes and cellular processes, which we validate in gene-expression profiling of failing and healthy human hearts. Colocalization, gene expression profiling, and Mendelian randomization provide convergent evidence for the roles of BCKDHA and circulating branch-chain amino acids in heart failure and cardiac structure. Finally, proteome-wide Mendelian randomization identifies 9 circulating proteins associated with heart failure or quantitative imaging traits. These analyses highlight similarities and differences among heart failure and associated cardiovascular imaging endophenotypes, implicate common genetic variation in the pathogenesis of heart failure, and identify circulating proteins that may represent cardiomyopathy treatment targets., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.