1
|
Shehadeh LA, Robleto E, Lopaschuk GD. Cardiac energy substrate utilization in heart failure with preserved ejection fraction: reconciling conflicting evidence on fatty acid and glucose metabolism. Am J Physiol Heart Circ Physiol 2025; 328:H1267-H1295. [PMID: 40251758 DOI: 10.1152/ajpheart.00121.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/10/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by complex metabolic derangements, yet considerable controversy exists regarding the role, and specifically the direction, of fatty acid oxidation (FAO) in disease progression. Through a systematic review with narrative synthesis of 44 studies identified from MEDLINE, Embase, and Web of Science databases, we critically examine the seemingly contradictory evidence regarding cardiac FAO in HFpEF. Our systematic analysis of experimental approaches reveals that many apparent contradictions can be resolved by considering differences in methodological approaches, interpretation of indirect metabolic markers, and the dynamic nature of metabolic adaptation in disease progression. Direct measurements consistently demonstrate that FAO remains active or increased in HFpEF hearts, whereas glucose oxidation becomes impaired, challenging previous assumptions based on indirect metabolic assessments. Methodological differences, particularly between studies using isolated mitochondria versus intact hearts and indirect versus direct substrate utilization measurements, can explain many apparent contradictions in the literature. Clinical and experimental evidence supports that FAO is maintained or elevated in HFpEF, with primary defects occurring in glucose oxidation and mitochondrial quality control. These findings suggest that successful therapeutic strategies for HFpEF should prioritize restoring metabolic flexibility and optimizing substrate utilization patterns rather than simply modulating FAO pathways. Our synthesis of the literature provides a comprehensive framework for understanding cardiac energy metabolism in HFpEF and identifies critical areas for future investigation.NEW & NOTEWORTHY Direct measurements reveal fatty acid oxidation remains active or increased in HFpEF hearts, whereas glucose oxidation becomes impaired, challenging previous assumptions. Apparent contradictions in HFpEF metabolism literature arise from methodological differences-studies using isolated mitochondria versus intact hearts. Evidence demonstrates fatty acid oxidation is maintained in HFpEF, with defects primarily in glucose oxidation. Successful therapeutic strategies should prioritize restoring metabolic flexibility rather than simply modulating fatty acid oxidation pathways.
Collapse
Affiliation(s)
- Lina A Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States
- Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States
- Miami Veterans' Affairs Medical Center, Miami, Florida, United States
| | - Emely Robleto
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States
- Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
2
|
Sun Q, Wagg CS, Wong N, Wei K, Ketema EB, Zhang L, Fang L, Seubert JM, Lopaschuk GD. Alterations of myocardial ketone metabolism in heart failure with preserved ejection fraction (HFpEF). ESC Heart Fail 2025. [PMID: 40420397 DOI: 10.1002/ehf2.15319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/04/2025] [Accepted: 04/16/2025] [Indexed: 05/28/2025] Open
Abstract
INTRODUCTION Cardiac energy metabolism is disrupted in heart failure with preserved ejection fraction (HFpEF), as characterized by a switch from glucose oxidation towards fatty acid oxidation. However, although oxidation of ketones is an important source of ATP it remains unclear how the heart oxidizes ketones in HFpEF. It is also unclear whether elevating ketone supply to the heart can improve cardiac energetics and/or provide functional benefit for the hearts in HFpEF. AIMS The present study investigated the effects of increasing ketone supply to the heart via ketone supplementation or SGLT2 inhibitor treatment in a mouse model of HFpEF. METHODS HFpEF was induced in 13-month-old C57BL/6N female mice with 60% high-fat diet and L-NAME (0.5 g/L/day in the drinking water) for 6 weeks. In parallel, two other groups of mice were maintained on the HFpEF protocol while also receiving either a ketone ester supplement (1-3 butanediol 1 g/kg/day) or SGLT2 inhibitor (empagliflozin 10 mg/kg/day) for 6 weeks. Control mice were fed with regular low-fat diet and regular drinking water. Hearts of the mice were excised and perfused in the isolated working mode aerobically with 5-mM glucose, 0.8-mM palmitate, 100-μU/mL insulin, with either low (0.6 mM) or high (1 mM) levels of β-hydroxybutyrate. Metabolic rates of the hearts were measured with radiolabelled [U-14C] glucose, [9,10-3H] palmitate and [3-14C] β-hydroxybutyrate. RESULTS In HFpEF mouse hearts, glucose oxidation was significantly decreased with a parallel increase in fatty acid oxidation. Increasing β-hydroxybutyrate levels from 0.6 to 1 mM in the perfusate resulted in a rise in ketone oxidation rates in control hearts (from 861 ± 63 to 1377 ± 94 nmol g dry wt-1 min-1), which was muted in HFpEF hearts (from 737 ± 68 to 897 ± 134 nmol g dry wt-1 min-1). Following ketone ester supplement or SGLT2 inhibitor treatment, HFpEF mice presented with restored ketone oxidation rates (from 674 ± 36 to 1181 ± 115 nmol g dry wt-1 min-1 with ketone ester supplement and from 797 ± 121 to 1240 ± 120 nmol g dry wt-1 min-1 with SGLT2i). Yet, this was not associated with improvement in cardiac function. CONCLUSIONS In HFpEF mice, the heart switches from glucose oxidation to fatty acid oxidation, with ketone oxidation being impaired. Increasing ketone supply to the heart via ketone ester supplementation or SGLT2 inhibitor treatment increases myocardial ketone oxidation rates but was not associated with functional improvements. Unlike HFrEF, ketone supplementation strategies may be less effective in HFpEF due to an impairment of myocardial ketone oxidation in HFpEF.
Collapse
Affiliation(s)
- Qiuyu Sun
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Cory S Wagg
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Nathan Wong
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Kaleigh Wei
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ezra B Ketema
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Liyan Zhang
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Liye Fang
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
Capone F, Vacca A, Bidault G, Sarver D, Kaminska D, Strocchi S, Vidal-Puig A, Greco CM, Lusis AJ, Schiattarella GG. Decoding the Liver-Heart Axis in Cardiometabolic Diseases. Circ Res 2025; 136:1335-1362. [PMID: 40403112 DOI: 10.1161/circresaha.125.325492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
The liver and heart are closely interconnected organs, and their bidirectional interaction plays a central role in cardiometabolic disease. In this review, we summarize current evidence linking liver dysfunction-particularly metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, and cirrhosis-with an increased risk of heart failure and other cardiovascular diseases. We discuss how these liver conditions contribute to cardiac remodeling, systemic inflammation, and hemodynamic stress and how cardiac dysfunction in turn impairs liver perfusion and promotes hepatic injury. Particular attention is given to the molecular mediators of liver-heart communication, including hepatokines and cardiokines, as well as the emerging role of advanced research methodologies, including omics integration, proximity labeling, and organ-on-chip platforms, that are redefining our understanding of interorgan cross talk. By integrating mechanistic insights with translational tools, this review aims to support the development of multiorgan therapeutic strategies for cardiometabolic disease.
Collapse
Affiliation(s)
- Federico Capone
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Department of Medicine, Unit of Internal Medicine III, Padua University Hospital, University of Padua, Padova, Italy (F.C.)
- Department of Biomedical Sciences, University of Padova, Italy (F.C.)
| | - Antonio Vacca
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Clinica Medica, Department of Medicine, University of Udine, Italy (A.V.)
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, United Kingdom (G.B., A.V.-P.)
| | - Dylan Sarver
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
- Department of Microbiology, Immunology and Molecular Genetics (D.S., A.J.L.), University of California, Los Angeles
- Department of Human Genetics (D.S., A.J.L.), University of California, Los Angeles
| | - Dorota Kaminska
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
| | - Stefano Strocchi
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (S.S., G.G.S.)
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, United Kingdom (G.B., A.V.-P.)
- Centro de Investigacion Principe Felipe, Valencia, Spain (A.V.-P.)
| | - Carolina M Greco
- Department of Biomedical Sciences, Humanitas University, Milan, Italy (C.M.G.)
- IRCCS Humanitas Research Hospital, Milan, Italy (C.M.G.)
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
- Department of Microbiology, Immunology and Molecular Genetics (D.S., A.J.L.), University of California, Los Angeles
- Department of Human Genetics (D.S., A.J.L.), University of California, Los Angeles
| | - Gabriele G Schiattarella
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (S.S., G.G.S.)
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany (G.G.S.)
- Friede Springer Cardiovascular Prevention Center at Charité-Universitätsmedizin Berlin, Germany (G.G.S.)
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (G.G.S.)
| |
Collapse
|
4
|
Pepin ME, Konrad PJM, Nazir S, Bazgir F, Maack C, Nickel A, Gorman J, Hohl M, Schreiter F, Dewenter M, de Britto Chaves Filho A, Schulze A, Karlstaedt A, Frey N, Seidman C, Seidman J, Backs J. Mitochondrial NNT Promotes Diastolic Dysfunction in Cardiometabolic HFpEF. Circ Res 2025. [PMID: 40340422 DOI: 10.1161/circresaha.125.326154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Clinical management of heart failure with preserved ejection fraction (HFpEF) is hindered by a lack of disease-modifying therapies capable of altering its distinct pathophysiology. Despite the widespread implementation of a 2-hit model of cardiometabolic HFpEF to inform precision therapy, which utilizes ad libitum high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester, we observe that C57BL6/J mice exhibit less cardiac diastolic dysfunction in response to high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester. METHODS Genetic strain-specific single-nucleus transcriptomic analysis identified disease-relevant genes that enrich oxidative metabolic pathways within cardiomyocytes. Because C57BL/6J mice are known to harbor a loss-of-function mutation affecting the inner mitochondrial membrane protein Nnt (nicotinamide nucleotide transhydrogenase), we used an isogenic model of Nnt loss-of-function to determine whether intact NNT is necessary for the pathological cardiac manifestations of high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester. Twelve-week-old mice cross-bred to isolate wild-type (Nnt+/+) or loss-of-function (Nnt-/-) Nnt in the C57BL/6N background were challenged with high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester for 9 weeks (n=6-10). RESULTS Nnt+/+ mice exhibited impaired ventricular diastolic relaxation and pathological remodeling, as assessed via E/e' (42.8 versus 21.5, P=1.2×10-10), E/A (2.3 versus 1.4, P=4.1×10-2), diastolic stiffness (0.09 versus 0.04 mm Hg/μL, P=5.1×10-3), and myocardial fibrosis (P=2.3×10-2). Liquid chromatography and mass spectroscopy exposed a 40.0% reduction in NAD+ (P=8.4×10-3) and a 38.8% reduction in glutathione:GSSG (P=2.6×10-2) among Nnt+/+ mice after high-fat diet and 0.5% N(ω)-nitro-L-arginine methyl ester feeding. Using single-nucleus ligand-receptor analysis, we implicate Fgf1 (fibroblast growth factor 1) as a putative NNT-dependent mediator of cardiomyocyte-to-fibroblast signaling of myocardial fibrosis. CONCLUSIONS Together, these findings underscore the pivotal role of mitochondrial dysfunction in HFpEF pathogenesis, implicating both NNT and Fgf1 as novel therapeutic targets.
Collapse
Affiliation(s)
- Mark E Pepin
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
- Broad Institute of Harvard and MIT, Boston, MA (M.E.P.)
- Division of Cardiovascular Medicine, Stanford University Hospital, CA (M.E.P.)
| | - Philipp J M Konrad
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine III, Heidelberg University Hospital, Germany. (P.J.M.K., N.F.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Sumra Nazir
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Farhad Bazgir
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Germany (C.M., A.N.)
- Medical Clinic I, University Clinic Würzburg, Germany (C.M.)
| | - Alexander Nickel
- Comprehensive Heart Failure Center, University Clinic Würzburg, Germany (C.M., A.N.)
| | - Joshua Gorman
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
| | - Mathias Hohl
- Department of Internal Medicine III, Saarland University Hospital and Saarland University, Homburg/Saar, Germany (M.H.)
| | - Friederike Schreiter
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Matthias Dewenter
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Germany and Heidelberg University, Germany (M.D., J.B.)
| | | | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg (A.d.B.C.F., A.S.)
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (A.K.)
| | - Norbert Frey
- Department of Internal Medicine III, Heidelberg University Hospital, Germany. (P.J.M.K., N.F.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
| | - Christine Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
| | - Jonathan Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (M.E.P., J.G., C.S., J.S.)
| | - Johannes Backs
- Medical Faculty Heidelberg, Institute of Experimental Cardiology, Heidelberg University, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Heidelberg University, Germany. (J.B.)
- Department of Internal Medicine VIII, Heidelberg University Hospital, Germany. (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., J.B.)
- German Center for Cardiovascular Research (DZHK) (M.E.P., P.J.M.K., S.N., F.B., F.S., M.D., N.F., J.B.)
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL), Germany and Heidelberg University, Germany (M.D., J.B.)
| |
Collapse
|
5
|
Koay YC, McIntosh B, Ng YH, Cao Y, Wang XS, Han Y, Tomita S, Bai AY, Hunter B, Misra A, Loughrey CM, Bannon PG, Lal S, Lusis AJ, Kaye DM, Larance M, O’Sullivan JF. The Heart Has Intrinsic Ketogenic Capacity that Mediates NAD + Therapy in HFpEF. Circ Res 2025; 136:1113-1130. [PMID: 40211954 PMCID: PMC12063684 DOI: 10.1161/circresaha.124.325550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) has overtaken heart failure with reduced ejection fraction as the leading type of heart failure globally and is marked by high morbidity and mortality rates, yet with only a single approved pharmacotherapy: SGLT2i (sodium-glucose co-transporter 2 inhibitor). A prevailing theory for the mechanism underlying SGLT2i is nutrient deprivation signaling, of which ketogenesis is a hallmark. However, it is unclear whether the canonical ketogenic enzyme, HMGCS2 (3-hydroxy-3-methylglutaryl-coenzyme A synthase 2), plays any cardiac role in HFpEF pathogenesis or therapeutic response. METHODS We used human myocardium, human HFpEF and heart failure with reduced ejection fraction transcardiac blood sampling, an established murine model of HFpEF, ex vivo Langendorff perfusion, stable isotope tracing in isolated cardiomyocytes, targeted metabolomics, proteomics, lipidomics, and a novel cardiomyocyte-specific conditional HMGCS2-deficient model that we generated. RESULTS We demonstrate, for the first time, the intrinsic capacity of the human heart to produce ketones via HMGCS2. We found that increased acetylation of HMGCS2 led to a decrease in the enzyme's specific activity. However, this was overcome by an increase in the steady-state levels of protein. Oxidized form of nicotinamide adenine dinucleotide repletion restored HMGCS2 function via deacetylation, increased fatty acid oxidation, and rescued cardiac function in HFpEF. Critically, using a conditional, cardiomyocyte-specific HMGCS2 knockdown murine model, we revealed that the oxidized form of nicotinamide adenine dinucleotide is unable to rescue HFpEF in the absence of cardiomyocyte HMGCS2. CONCLUSIONS The canonical ketogenic enzyme, HMGCS2, mediates the therapeutic effects of the oxidized form of nicotinamide adenine dinucleotide repletion in HFpEF by restoring normal lipid metabolism and mitochondrial function.
Collapse
Affiliation(s)
- Yen Chin Koay
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Bailey McIntosh
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Yann Huey Ng
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Yang Cao
- Division of Life Sciences and Medicine, Department of Cardiology, The First Affiliated Hospital of USTC (Y.C.), University of Science and Technology of China (USTC), Hefei
- Division of Life Sciences and Medicine, School of Basic Medical Sciences (Y.C.), University of Science and Technology of China (USTC), Hefei
| | - Xiao Suo Wang
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Yanchuang Han
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Saki Tomita
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Angela Yu Bai
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - Benjamin Hunter
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory (B.H., S.L.), The University of Sydney, New South Wales, Australia
| | - Ashish Misra
- Heart Research Institute (A.M.), The University of Sydney, New South Wales, Australia
| | - Christopher M. Loughrey
- School of Cardiovascular and Metabolic Health and School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom (C.M.L.)
| | - Paul G. Bannon
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Department of Cardiothoracic Surgery (P.G.B., J.F.O.), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, New South Wales, Australia (P.G.B., S.L., J.F.O.)
| | - Sean Lal
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory (B.H., S.L.), The University of Sydney, New South Wales, Australia
- Department of Cardiology (S.L., J.F.O.), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, New South Wales, Australia (P.G.B., S.L., J.F.O.)
| | - Aldons J. Lusis
- Department of Medicine, Microbiology and Human Genetics, University of California, Los Angeles (A.J.L.)
| | - David M. Kaye
- Department of Cardiology, Alfred Hospital, Melbourne, Victoria, Australia (D.M.K.)
- Heart Failure Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (D.M.K.)
- Faculty of Medicine, Nursing, and Health Sciences, Central Clinical School, Monash University, Melbourne, Victoria, Australia (D.M.K.)
| | - Mark Larance
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
| | - John F. O’Sullivan
- Faculty of Medicine and Health, School of Medical Sciences (Y.C.K., B.M., Y.H.N., X.W., Y.H., S.T., A.Y.B., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Cardiometabolic Medicine (Y.C.K., B.M., Y.H.N., X.W., Y.H., P.G.B., S.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Charles Perkins Centre (Y.C.K., B.M., Y.H.N., X.W., Y.H., B.H., P.G.B., S.L., M.L., J.F.O.), The University of Sydney, New South Wales, Australia
- Department of Cardiothoracic Surgery (P.G.B., J.F.O.), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Department of Cardiology (S.L., J.F.O.), Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, New South Wales, Australia (P.G.B., S.L., J.F.O.)
- Faculty of Medicine, Technische Universität Dresden, Germany (J.F.O.)
| |
Collapse
|
6
|
Savarese G, Schiattarella GG, Lindberg F, Anker MS, Bayes-Genis A, Bäck M, Braunschweig F, Bucciarelli-Ducci C, Butler J, Cannata A, Capone F, Chioncel O, D'Elia E, González A, Filippatos G, Girerd N, Hulot JS, Lam CSP, Lund LH, Maack C, Moura B, Petrie MC, Piepoli M, Shehab A, Yilmaz MB, Seferovic P, Tocchetti CG, Rosano GMC, Metra M. Heart failure and obesity: Translational approaches and therapeutic perspectives. A scientific statement of the Heart Failure Association of the ESC. Eur J Heart Fail 2025. [PMID: 40328668 DOI: 10.1002/ejhf.3676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Obesity and heart failure (HF) represent two growing pandemics. In the general population, obesity affects one in eight adults and is linked with an increased risk for HF. Obesity is even more common in patients with HF, where it complicates the diagnosis of HF and is linked with worse symptoms and impaired exercise capacity. Over the past few years, new evidence on the mechanisms linking obesity with HF has been reported, particularly in relation to HF with preserved ejection fraction. Novel therapies inducing weight loss appear to have favourable effects on health status and cardiovascular risk. Against the backdrop of this rapidly evolving evidence landscape, HF clinicians are increasingly required to tailor their preventive, diagnostic, and therapeutic approaches to HF in the presence of obesity. This scientific statement by the Heart Failure Association of the European Society of Cardiology provides an up-to-date summary on obesity in HF, covering key areas such as epidemiology, translational aspects, diagnostic challenges, therapeutic approaches, and trial design.
Collapse
Affiliation(s)
- Gianluigi Savarese
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Felix Lindberg
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Markus S Anker
- Department of Cardiology CBF German Heart Center Charité, DZHK, BCRT, University Medicine Berlin FU and HU, Berlin, Germany
| | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari Germasn Trias I Pujol, CIBERCV, Badalona, Spain
| | - Magnus Bäck
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Chiara Bucciarelli-Ducci
- Royal Brompton and Harefield Hospitals, Guys' and St Thomas NHS Trust, London, UK
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College University, London, UK
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- University of Mississippi, Jackson, MS, USA
| | - Antonio Cannata
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine, Faculty of Life Science, King's College London, London, UK
- Cardiology Department, King's College Hospital NHS Foundation Trust, London, UK
| | - Federico Capone
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Unit of Internal Medicine III, Department of Medicine (DIMED), Padua University Hospital, University of Padua, Padova, Italy
- Department of Biomedical Sciences, University of Padua, Padova, Italy
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases 'Prof. C.C. Iliescu', University of Medicine Carol Davila, Bucharest, Romania
| | - Emilia D'Elia
- Cardiovascular Department, Papa Giovanni XXIII Hospital, Bergamo, Italy
- School of Medicine and Surgery, University Milano-Bicocca, Milan, Italy
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology and Cardiac Surgery, Clínica Universidad de Navarra and IdiSNA, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Gerasimos Filippatos
- Department of Cardiology, University Hospital Attikon, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Nicolas Girerd
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - Jean-Sébastien Hulot
- Université Paris Cité, INSERM, PARCC, Paris, France
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Carolyn S P Lam
- National Heart Centre Singapore & Duke-National University of Singapore, Singapore
| | - Lars H Lund
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
- Medical Clinic 1, University Clinic Würzburg, Würzburg, Germany
| | - Brenda Moura
- Department of Cardiology, Armed Forces Hospital, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Mark C Petrie
- School of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Massimo Piepoli
- Clinical Cardiology, IRCCS Policlinico San Donato, Milan, Italy
- Department of Preventive Cardiology, University of Wroclaw, Wroclaw, Poland
| | - Abdullah Shehab
- Department of Cardiology, Royal Burjeel Hospital, UAE University, Al Ain, UAE
| | - Mehmet B Yilmaz
- Department of Cardiology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Peter Seferovic
- Faculty of Medicine, University of Belgrade, and Serbian Academy of Sciences and Arts, Belgrade, Serbia
- University of Belgrade, Belgrade, Serbia
| | - Carlo G Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences (DISMET), Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Giuseppe M C Rosano
- Department of Human Sciences and Promotion of Quality of Life, San Raffaele Open University of Rome, Rome, Italy
- Cardiology, San Raffaele Cassino Hospital, Cassino, Italy
| | - Marco Metra
- Cardiology and Cardiac Catheterization Laboratory, Cardio-Thoracic Department, Civil Hospitals, Brescia, Italy
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| |
Collapse
|
7
|
Zhou R, Wang Y, Liu S, Su Y, Liu Z, Yang B, Li X, Zhao J, Xu J, Liu Q, Song F. Brevilin A, a novel BNIP3 inhibitor suppresses osteoclastogenesis and prevents ovariectomy-induced bone loss via impairing mitophagy and mitochondrial metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156774. [PMID: 40408943 DOI: 10.1016/j.phymed.2025.156774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/29/2025] [Accepted: 04/13/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND The mitochondrial dysfunction and overactive osteoclasts is involved in the progress of osteoporosis. Brevilin A (BA), a sesquiterpene lactone, is a compound extracted and purified from Centipeda minima. It exhibits a range of pharmacological activities, such as anti-inflammatory and antioxidant effects. However, its specific impact on osteoporosis remains unclear. The present study is designed to explore BA as a novel osteoclast inhibitor for the treatment of osteoporosis as well as its molecular mechanisms of action via BNIP3-mediated mitophagy. METHODS The cytotoxicity of BA in vitro was evaluated using the CCK8 assay, while tartrate-resistant acid phosphatase (TRAcP) staining and bone resorption assays were conducted to examine its effects on osteoclastogenesis and osteoclast function. To elucidate the molecular mechanisms by which BA targets BNIP3 in osteoclasts, RNA-seq, molecular docking analysis, Surface plasmon resonance (SPR), qPCR, western blot, mitochondrial oxygen consumption rate (OCR), transmission electron microscopy (TEM), Single cell sequencing and immunofluorescence staining were employed. In addition, a specific BNIP3 agonist IOX5, was used to revalidate the inhibitory effect of BA on BNIP3. To investigate the effects and protective role of BA in modulating BNIP3 on bone loss in osteoporotic mice induced by ovariectomy (OVX), we employed in vivo micro-CT scanning and histological immunostaining techniques. RESULTS Our study demonstrated that BA inhibited RANKL-induced osteoclastogenesis in a concentration-dependent manner without any cell cytotoxicity. Further, BA abrogated MAPK-related proteins and intracellular and mitochondrial ROS level, subsequently inhibiting NFATc1 activity. RNA-seq analysis revealed that the molecular mechanism by which BA inhibited osteoclasts is closely related to mitophagy and mitochondrial function. Here, we found that BA suppressed oxygen consumption rate and mitochondrial oxidative phosphorylation during osteoclastogenesis. This compound abolished expression of ATG5, SIRT3, Beclin1 and LC3B. RANKL-induced mitophagy associated protein (PINK1 and Parkin) were also suppressed by BA. BA interacted with BNIP3 and IOX5 treatment further verified the targeted inhibition effect of BA on BNIP3. In addition, we found that BNIP3 deficient inhibited osteoclast differentiation related with mitophagy and mitochondrial function. In vivo experiments confirmed that BA significantly prevent OVX-induced bone loss associated with BNIP3-mediated mitophagy. CONCLUSIONS Our study reveals for the first time that BA acts as a novel inhibitor of BNIP3, which ameliorates osteoclast activity and OVX-induced osteoporosis via limiting mitophagy and mitochondrial energy production, suggesting that it could be a novel therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Rui Zhou
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Life Sciences Institute, Guangxi Medical University, Nanning 530021, China
| | - Yiyuan Wang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Life Sciences Institute, Guangxi Medical University, Nanning 530021, China
| | - Siyi Liu
- Life Sciences Institute, Guangxi Medical University, Nanning 530021, China
| | - Yuangang Su
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhijuan Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Baihui Yang
- The First Clinical Medical College, Guangxi Medical University, Nanning 530021, China
| | - Xiangde Li
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jinmin Zhao
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jiake Xu
- School of Biomedical Sciences, the University of Western Australia, Perth, Australia; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518000, China; Shenzhen University of Advanced Technology, Shenzhen 518000, China.
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Fangming Song
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Life Sciences Institute, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
8
|
Panwar A, Malik SO, Adib M, Lopaschuk GD. Cardiac energy metabolism in diabetes: emerging therapeutic targets and clinical implications. Am J Physiol Heart Circ Physiol 2025; 328:H1089-H1112. [PMID: 40192025 DOI: 10.1152/ajpheart.00615.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/15/2024] [Accepted: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Patients with diabetes are at an increased risk for developing diabetic cardiomyopathy and other cardiovascular complications. Alterations in cardiac energy metabolism in patients with diabetes, including an increase in mitochondrial fatty acid oxidation and a decrease in glucose oxidation, are important contributing factors to this increase in cardiovascular disease. A switch from glucose oxidation to fatty acid oxidation not only decreases cardiac efficiency due to increased oxygen consumption but it can also increase reactive oxygen species production, increase lipotoxicity, and redirect glucose into other metabolic pathways that, combined, can lead to heart dysfunction. Currently, there is a lack of therapeutics available to treat diabetes-induced heart failure that specifically target cardiac energy metabolism. However, it is becoming apparent that part of the benefit of existing agents such as GLP-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors may be related to their effects on cardiac energy metabolism. In addition, direct approaches aimed at inhibiting cardiac fatty acid oxidation or increasing glucose oxidation hold future promise as potential therapeutic approaches to treat diabetes-induced cardiovascular disease.
Collapse
Affiliation(s)
- Archee Panwar
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Sufyan O Malik
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Muhtasim Adib
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
9
|
Darwesh AM, Fang L, Altamimi TR, Jamieson KL, Bassiouni W, Valencia R, Huang A, Wang F, Zhang H, Ahmed M, Gopal K, Zhang Y, Michelakis ED, Ussher JR, Edin ML, Zeldin DC, Barakat K, Oudit GY, Kassiri Z, Lopaschuk GD, Seubert JM. Cardioprotective effect of 19,20-epoxydocosapentaenoic acid (19,20-EDP) in ischaemic injury involves direct activation of mitochondrial sirtuin 3. Cardiovasc Res 2025; 121:267-282. [PMID: 39658136 PMCID: PMC12012443 DOI: 10.1093/cvr/cvae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/12/2024] [Accepted: 10/17/2024] [Indexed: 12/12/2024] Open
Abstract
AIMS Although current clinical therapies following myocardial infarction (MI) have improved patient outcomes, morbidity, and mortality rates, secondary to ischaemic and ischaemia reperfusion (IR) injury remains high. Maintaining mitochondrial quality is essential to limit myocardial damage following cardiac ischaemia and IR injury. The mitochondrial deacetylase sirtuin 3 (SIRT3) plays a pivotal role in regulating mitochondrial function and cardiac energy metabolism. In the current study, we hypothesize that 19,20-epoxydocosapentaenoic acid (19,20-EDP) attenuates cardiac IR injury via stimulating mitochondrial SIRT3. METHODS AND RESULTS Ex vivo models of isolated heart perfusions were performed in C57BL/6 mice to assess the effect of 19,20-EDP on cardiac function and energy metabolism following IR injury. In vivo permanent occlusion of the left anterior descending coronary artery was performed to induce MI; mice were administered 19,20-EDP with or without the SIRT3 selective inhibitor 3-TYP. Mitochondrial SIRT3 targets and respiration were assessed in human left ventricular tissues obtained from individuals with ischaemic heart disease (IHD) and compared to non-failing controls (NFCs). Binding affinity of 19,20-EDP to human SIRT3 was assessed using molecular modelling and fluorescence thermal shift assay. Results demonstrated that hearts treated with 19,20-EDP had improved post-ischaemic cardiac function, better glucose oxidation rates, and enhanced cardiac efficiency. The cardioprotective effects were associated with enhanced mitochondrial SIRT3 activity. Interestingly, treatment with 19,20-EDP markedly improved mitochondrial respiration and SIRT3 activity in human left ventricle (LV) fibres with IHD compared to NFC. Moreover, 19,20-EDP was found to bind to the human SIRT3 protein enhancing the NAD+-complex stabilization leading to improved SIRT3 activity. Importantly, the beneficial effects of 19,20-EDP were abolished by SIRT3 inhibition or using the S149A mutant SIRT3. CONCLUSION These data demonstrate that 19,20-EDP-mediated cardioprotective mechanisms against ischaemia and IR injury involve mitochondrial SIRT3, resulting in improved cardiac efficiency.
Collapse
Affiliation(s)
- Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2-35 Medical Sciences Building, Edmonton, AB, Canada T6G 2H1
| | - Liye Fang
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, 7-55 Medical Sciences Building, Edmonton, AB, Canada T6G 2H7
| | - Tariq R Altamimi
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - K Lockhart Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2-35 Medical Sciences Building, Edmonton, AB, Canada T6G 2H1
| | - Wesam Bassiouni
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, 7-55 Medical Sciences Building, Edmonton, AB, Canada T6G 2H7
| | - Robert Valencia
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, 7-55 Medical Sciences Building, Edmonton, AB, Canada T6G 2H7
| | - Andy Huang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2-35 Medical Sciences Building, Edmonton, AB, Canada T6G 2H1
| | - Faqi Wang
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Hao Zhang
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Marawan Ahmed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2-35 Medical Sciences Building, Edmonton, AB, Canada T6G 2H1
- Quantitative Solutions, API, Edmonton, AB, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2-35 Medical Sciences Building, Edmonton, AB, Canada T6G 2H1
| | - Yongneng Zhang
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Evangelos D Michelakis
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2-35 Medical Sciences Building, Edmonton, AB, Canada T6G 2H1
| | - Matthew L Edin
- National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC, USA
| | - Darryl C Zeldin
- National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC, USA
| | - Khaled Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2-35 Medical Sciences Building, Edmonton, AB, Canada T6G 2H1
- Li Ka Shing Applied Virology Institute, University of Alberta, Edmonton, AB, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Gary D Lopaschuk
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 2-35 Medical Sciences Building, Edmonton, AB, Canada T6G 2H1
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, 7-55 Medical Sciences Building, Edmonton, AB, Canada T6G 2H7
| |
Collapse
|
10
|
Qin C, Qin Y, Zhou S. Methylations in dilated cardiomyopathy and heart failure. Front Cardiovasc Med 2025; 12:1559550. [PMID: 40290189 PMCID: PMC12021892 DOI: 10.3389/fcvm.2025.1559550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Dilated cardiomyopathy (DCM) is characterized by impaired expansion or contraction of the left or both ventricles in the absence of abnormal load conditions (such as primary valve disease) or severe coronary artery disease that can lead to ventricular remodeling. Genetic mutations, infections, inflammation, autoimmune diseases, exposure to toxins, and endocrine or neuromuscular factors have all been implicated in the causation of DCM. Cardiomyopathy, particularly DCM, often has genetic underpinnings, with established or suspected genetic origins. Up to 40% of DCM cases involve probable or confirmed genetic variations. The significance of RNA modification in the pathogenesis of hypertension, cardiac hypertrophy, and atherosclerosis is well-established. Of late, RNA methylation has garnered attention for its involvement in DCM. This review examines the biological mechanisms and effects of RNA methylation in DCM and heart failure.
Collapse
Affiliation(s)
- Cong Qin
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Yansong Qin
- Undergraduate School, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shanshan Zhou
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Li H, Xiao F, Zhou C, Zhu T, Wang S. Metabolic Adaptations and Therapies in Cardiac Hypoxia: Mechanisms and Clinical Implications/ Potential Strategies. JACC Basic Transl Sci 2025:S2452-302X(24)00458-3. [PMID: 40265246 DOI: 10.1016/j.jacbts.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 04/24/2025]
Abstract
Cardiac hypoxia triggers a cascade of responses and functional changes in myocardial and non-myocardial cells, profoundly affecting cellular metabolism, oxygen-sensing mechanisms, and immune responses. Myocardial cells, being the primary cell type in cardiac tissue, undergo significant alterations in energy metabolism, including glycolysis, fatty acid metabolism, ketone body utilization, and branched-chain amino acid metabolism, to maintain cardiac function under hypoxic conditions. Non-myocardial cells, such as fibroblasts, endothelial cells, and immune cells, although fewer in number, play crucial roles in regulating cardiac homeostasis, maintaining structural integrity, and responding to injury. This review discusses the metabolic reprogramming of immune cells, particularly macrophages, during ischemia-reperfusion injury and explores various therapeutic strategies that modulate these metabolic pathways to protect the heart during hypoxia. Understanding these interactions provides valuable insights and potential therapeutic targets for heart disease treatment.
Collapse
Affiliation(s)
- Huili Li
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Emergency Department, The State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Fei Xiao
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chenghui Zhou
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012, West China Hospital, Sichuan University, Chengdu, China.
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Linzhi People's Hospital, Linzhi, Tibet, China.
| |
Collapse
|
12
|
Li Y, Lin Z, Li Y. Visceral obesity and HFpEF: targets and therapeutic opportunities. Trends Pharmacol Sci 2025; 46:337-356. [PMID: 40113531 DOI: 10.1016/j.tips.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
The effectiveness of weight-loss drugs in heart failure (HF) with preserved ejection fraction (HFpEF) highlights the link between obesity (adipose tissue) and HF (the heart). Recent guidelines incorporating the waist:height ratio for diagnosing and treating obesity reflect the growing recognition of the significance of visceral adiposity. However, its unique impact on HFpEF and their complex relationship remain underexplored. With limited treatment options for obesity-related HFpEF, novel disease-modifying treatments are urgently needed. Here, we clarify the relationship between visceral obesity and HFpEF, introducing the concept of the visceral adipose tissue-heart axis to explore its mechanisms and therapeutic potential. We also discuss promising strategies targeting visceral obesity in HFpEF and propose directions for future research.
Collapse
Affiliation(s)
- Yilin Li
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of the Ministry of Education for Cardiovascular Remodeling-Related Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Zhuofeng Lin
- The Innovation Center of Cardiometabolic Disease, Guangdong Medical University, Dongguan 523808, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of the Ministry of Education for Cardiovascular Remodeling-Related Diseases, Beijing Collaborative Innovative Research Center for Cardiovascular Diseases, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China.
| |
Collapse
|
13
|
Cui X, Spanos M, Zhao C, Wan W, Cui C, Wang L, Xiao J. Mitochondrial Dysfunction in HFpEF: Potential Interventions Through Exercise. J Cardiovasc Transl Res 2025; 18:442-456. [PMID: 39863753 DOI: 10.1007/s12265-025-10591-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
HFpEF is a prevalent and complex type of heart failure. The concurrent presence of conditions such as obesity, hypertension, hyperglycemia, and hyperlipidemia significantly increase the risk of developing HFpEF. Mitochondria, often referred to as the powerhouses of the cell, are crucial in maintaining cellular functions, including ATP production, intracellular Ca2+ regulation, reactive oxygen species generation and clearance, and the regulation of apoptosis. Exercise plays a vital role in preserving mitochondrial homeostasis, thereby protecting the cardiovascular system from acute stress, and is a fundamental component in maintaining cardiovascular health. In this study, we review the mitochondrial dysfunction underlying the development and progression of HFpEF. Given the pivotal role of exercise in modulating cardiovascular diseases, we particularly focus on exercise as a potential therapeutic strategy for improving mitochondrial function. Graphical abstract Note: This picture was created with BioRender.com.
Collapse
Affiliation(s)
- Xinxin Cui
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Albert Einstein College of Medicine, Department of Internal Medicine, NCB, Bronx, NY, USA
| | - Cuimei Zhao
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wensi Wan
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China
| | - Caiyue Cui
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China
| | - Lijun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China.
| |
Collapse
|
14
|
Hathorn B, Haykowsky MJ, Almandoz J, Pandey A, Sarma S, Hearon CM, Babb TG, Balmain BN, Fu Q, Zaha VG, Levine BD, Nelson MD. Insights Into the Role of Obesity in Heart Failure With Preserved Ejection Fraction Pathophysiology and Management. Can J Cardiol 2025:S0828-282X(25)00199-0. [PMID: 40122162 DOI: 10.1016/j.cjca.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025] Open
Abstract
Heart failure (HF) is a significant global health issue, categorized by left ventricular ejection fraction, being either reduced (HFrEF < 0.40) or preserved (HFpEF > 0.50), or in the middle of this range. Although the overall incidence of HF remains stable, HFpEF cases are increasing, representing about 50% of all HF cases. Outcomes for HFpEF are similar to those for HFrEF, leading to substantial health-care resource use. Despite extensive research over the past 2 decades, the prognosis and mortality rates for HFpEF remain high. A key feature of HFpEF is exercise intolerance, characterized by severe exertional dyspnea and fatigue, which significantly impacts quality of life. The underlying mechanisms of exercise intolerance are not fully understood due to the complex pathophysiology and multisystem involvement. Obesity is a common comorbidity in HFpEF, especially in North America, leading to worsening symptoms, hemodynamics, and mortality rates. Increased adiposity leads to inflammation, hypertension, dyslipidemia, and insulin resistance, and impairing cardiac, vascular, pulmonary, and skeletal muscle function. Therefore, managing obesity is crucial in treating HFpEF. In this review we explore the pathophysiologic mechanisms of HFpEF, emphasizing obesity's role, and we discuss current management strategies while identifying areas needing further research.
Collapse
Affiliation(s)
- Brandon Hathorn
- Applied Physiology and Advanced Imaging Laboratory, University of Texas at Arlington, Arlington, Texas, USA
| | - Mark J Haykowsky
- College of Health Sciences, Faculty of Nursing, University of Alberta, Edmonton, Alberta, Canada
| | - Jaime Almandoz
- Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ambarish Pandey
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Satyam Sarma
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA; Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
| | - Christopher M Hearon
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
| | - Tony G Babb
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Bryce N Balmain
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Qi Fu
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA; Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
| | - Vlad G Zaha
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA; Clinical Imaging Research Center, University of Texas at Arlington, Arlington, Texas, USA
| | - Benjamin D Levine
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA; Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
| | - Michael D Nelson
- Applied Physiology and Advanced Imaging Laboratory, University of Texas at Arlington, Arlington, Texas, USA; Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA; Clinical Imaging Research Center, University of Texas at Arlington, Arlington, Texas, USA.
| |
Collapse
|
15
|
Jani VP, Yoo EJ, Binek A, Guo A, Kim JS, Aguilan J, Keykhaei M, Jenkin SR, Sidoli S, Sharma K, Van Eyk JE, Kass DA, Hahn VS. Myocardial Proteome in Human Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2025; 14:e038945. [PMID: 40079330 DOI: 10.1161/jaha.124.038945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/16/2024] [Indexed: 03/15/2025]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) constitutes more than half of all HF but has few effective therapies. Recent human myocardial transcriptomics and metabolomics have identified major differences between HFpEF and controls. How this translates at the protein level is unknown. METHODS AND RESULTS Myocardial tissue from patients with HFpEF and nonfailing donor controls was analyzed by data-dependent acquisition (n=10 HFpEF, n=10 controls) and data-independent acquisition (n=44 HFpEF, n=5 controls) mass spectrometry-based proteomics. Differential protein expression analysis, pathway overrepresentation, weighted coexpression network analysis, and machine learning were integrated with clinical characteristics and previously reported transcriptomics. Principal component analysis (data-dependent acquisition-mass spectrometry) found HFpEF separated into 2 subgroups: one similar to controls and the other disparate. Downregulated proteins in HFpEF versus controls were enriched in mitochondrial transport/organization, translation, and metabolism including oxidative phosphorylation. Proteins upregulated in HFpEF were related to immune activation, reactive oxygen species, and inflammatory response. Ingenuity pathway analysis predicted downregulation of protein translation, mitochondrial function, and glucose and fat metabolism in HFpEF. Expression of oxidative phosphorylation and metabolism genes (higher) versus proteins (lower) was discordant in HFpEF versus controls. Data-independent acquisition-mass spectrometry proteomics also yielded 2 HFpEF subgroups; the one most different from controls had a higher proportion of patients with severe obesity and exhibited lower proteins related to fuel metabolism, oxidative phosphorylation, and protein translation. Three modules of correlated proteins in HFpEF that correlated with left ventricular hypertrophy and right ventricular load related to (1) proteasome; (2) fuel metabolism; and (3) protein translation, oxidative phosphorylation, and sarcomere organization. CONCLUSIONS Integrative proteomics, transcriptomics, and pathway analysis supports a defect in both metabolism and translation in HFpEF. Patients with HFpEF with more distinct proteomic signatures from control more often had severe obesity, supporting therapeutic efforts targeting metabolism and translation, particularly in this subgroup.
Collapse
Affiliation(s)
- Vivek P Jani
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Edwin J Yoo
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Aleksandra Binek
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center Los Angeles CA USA
| | - Alina Guo
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Julie S Kim
- Department of Biochemistry Albert Einstein College of Medicine Bronx NY USA
| | - Jennifer Aguilan
- Department of Pathology Albert Einstein College of Medicine Bronx NY USA
| | - Mohammad Keykhaei
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Sydney R Jenkin
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Simone Sidoli
- Department of Biochemistry Albert Einstein College of Medicine Bronx NY USA
| | - Kavita Sharma
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center Los Angeles CA USA
| | - David A Kass
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
- Department of Pharmacology and Molecular Sciences, and Department of Biomedical Engineering Johns Hopkins University Baltimore MD USA
| | - Virginia S Hahn
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| |
Collapse
|
16
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
17
|
Rai NK, Venugopal H, Rajesh R, Ancha P, Venkatesh S. Mitochondrial complex-1 as a therapeutic target for cardiac diseases. Mol Cell Biochem 2025; 480:869-890. [PMID: 39033212 PMCID: PMC12076218 DOI: 10.1007/s11010-024-05074-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
Mitochondrial dysfunction is critical for the development and progression of cardiovascular diseases (CVDs). Complex-1 (CI) is an essential component of the mitochondrial electron transport chain that participates in oxidative phosphorylation and energy production. CI is the largest multisubunit complex (~ 1 Mda) and comprises 45 protein subunits encoded by seven mt-DNA genes and 38 nuclear genes. These subunits function as the enzyme nicotinamide adenine dinucleotide hydrogen (NADH): ubiquinone oxidoreductase. CI dysregulation has been implicated in various CVDs, including heart failure, ischemic heart disease, pressure overload, hypertrophy, and cardiomyopathy. Several studies demonstrated that impaired CI function contributes to increased oxidative stress, altered calcium homeostasis, and mitochondrial DNA damage in cardiac cells, leading to cardiomyocyte dysfunction and apoptosis. CI dysfunction has been associated with endothelial dysfunction, inflammation, and vascular remodeling, critical processes in developing atherosclerosis and hypertension. Although CI is crucial in physiological and pathological conditions, no potential therapeutics targeting CI are available to treat CVDs. We believe that a lack of understanding of CI's precise mechanisms and contributions to CVDs limits the development of therapeutic strategies. In this review, we comprehensively analyze the role of CI in cardiovascular health and disease to shed light on its potential therapeutic target role in CVDs.
Collapse
Affiliation(s)
- Neeraj Kumar Rai
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, School of Medicine, West Virginia University, Morgantown, 26505, WV, USA
- Nora Eccles Harrison Cardiovascular Research and Training Institute, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Harikrishnan Venugopal
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ritika Rajesh
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, School of Medicine, West Virginia University, Morgantown, 26505, WV, USA
| | - Pranavi Ancha
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, School of Medicine, West Virginia University, Morgantown, 26505, WV, USA
| | - Sundararajan Venkatesh
- Department of Physiology, Pharmacology and Toxicology, School of Medicine, School of Medicine, West Virginia University, Morgantown, 26505, WV, USA.
| |
Collapse
|
18
|
Ketema EB, Lopaschuk GD. The Impact of Obesity on Cardiac Energy Metabolism and Efficiency in Heart Failure With Preserved Ejection Fraction. Can J Cardiol 2025:S0828-282X(25)00099-6. [PMID: 39892611 DOI: 10.1016/j.cjca.2025.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025] Open
Abstract
The incidence and prevalence of heart failure with preserved ejection fraction (HFpEF) continues to rise, and now comprises more than half of all heart failure cases. There are many risk factors for HFpEF, including older age, hypertension, diabetes, dyslipidemia, sedentary behaviour, and obesity. The rising prevalence of obesity in society is a particularly important contributor to HFpEF development and severity. Obesity can adversely affect the heart, including inducing marked alterations in cardiac energy metabolism. This includes obesity-induced impairments in mitochondrial function, and an increase in fatty acid uptake and mitochondrial fatty acid β-oxidation. This increase in myocardial fatty acid metabolism is accompanied by an impaired myocardial insulin signaling and a marked decrease in glucose oxidation. This switch from glucose to fatty acid metabolism decreases cardiac efficiency and can contribute to severity of HFpEF. Increased myocardial fatty acid uptake in obesity is also associated with the accumulation of fatty acids, resulting in cardiac lipotoxicity. Obesity also results in dramatic changes in the release of adipokines, which can negatively impact cardiac function and energy metabolism. Obesity-induced increases in epicardial fat can also increase cardiac insulin resistance and negatively affect cardiac energy metabolism and HFpEF. However, optimizing cardiac energy metabolism in obese subjects may be one approach to preventing and treating HFpEF. This review discusses what is presently known about the effects of obesity on cardiac energy metabolism and insulin signaling in HFpEF. The clinical implications of obesity and energy metabolism on HFpEF are also discussed.
Collapse
Affiliation(s)
- Ezra B Ketema
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada. https://twitter.com/Ketema
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
19
|
Fuerlinger A, Stockner A, Sedej S, Abdellatif M. Caloric restriction and its mimetics in heart failure with preserved ejection fraction: mechanisms and therapeutic potential. Cardiovasc Diabetol 2025; 24:21. [PMID: 39827109 PMCID: PMC11742808 DOI: 10.1186/s12933-024-02566-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025] Open
Abstract
The global increase in human life expectancy, coupled with an unprecedented rise in the prevalence of obesity, has led to a growing clinical and socioeconomic burden of heart failure with preserved ejection fraction (HFpEF). Mechanistically, the molecular and cellular hallmarks of aging are omnipresent in HFpEF and are further exacerbated by obesity and associated metabolic diseases. Conversely, weight loss strategies, particularly caloric restriction, have shown promise in improving health status in patients with HFpEF and are considered the gold standard for promoting longevity and healthspan (disease-free lifetime) in model organisms. In this review, we implicate fundamental mechanisms of aging in driving HFpEF and elucidate how caloric restriction mitigates the disease progression. Furthermore, we discuss the potential for pharmacologically mimicking the beneficial effects of caloric restriction in HFpEF using clinically approved and emerging caloric restriction mimetics. We surmise that these compounds could offer novel therapeutic avenues for HFpEF and alleviate the challenges associated with the implementation of caloric restriction and other lifestyle modifications to reduce the burden of HFpEF at a population level.
Collapse
Affiliation(s)
- Alexander Fuerlinger
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
- BioTechMed-Graz, 8010, Graz, Austria
| | - Alina Stockner
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
- BioTechMed-Graz, 8010, Graz, Austria
- Faculty of Medicine, University of Maribor, 2000, Maribor, Slovenia
| | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria.
- BioTechMed-Graz, 8010, Graz, Austria.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805, Villejuif, France.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, 75006, France.
| |
Collapse
|
20
|
Zampas P, Li Z, Katsouda A, Varela A, Psarras S, Davos CH, Lefer DJ, Papapetropoulos A. Protective role of 3-mercaptopyruvate sulfurtransferase (MPST) in the development of metabolic syndrome and vascular inflammation. Pharmacol Res 2025; 211:107542. [PMID: 39667544 DOI: 10.1016/j.phrs.2024.107542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/19/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
Metabolic syndrome (MetS) is a cluster of metabolic abnormalities that occur concurrently and increase the risk of cardiovascular disease. 3-mercaptopyruvate sulfurtransferase (MPST) is a cysteine-catabolizing enzyme that yields pyruvate and hydrogen sulfide (H2S) and plays a central role in the regulation of energy homeostasis. Herein, we seek to investigate the role of MPST/H2S in MetS and its cardiovascular consequences using a mouse model of the disease. Mice were fed a high-fat diet (HFD) for 15 weeks to induce obesity and hyperglycemia and administrated a nitric oxide synthase inhibitor, during the last 5 weeks to induce hypertension and MetS. This model caused a mild left ventricular (LV) diastolic dysfunction and vascular endothelial dysfunction. Free H2S and sulfane-sulfur levels were decreased in the aorta, but unaltered in the heart. Also, downregulation of MPST and thiosulfate sulfuretransferase (TST) were observed in the aorta. Global deletion of Mpst (Mpst-/-) resulted in increased body weight and greater glucose intolerance in mice with MetS, without affecting their blood pressure, and caused an upregulation of genes involved in immune responses in the vasculature suggestive of T-cell infiltration and activation. Pharmacological restoration of H2S levels ameliorated the comorbidities of MetS; GYY4137 administration reduced body weight and blood pressure, attenuated cardiac fibrosis and improved glucose handling and endothelium-dependent relaxation. In conclusion, this study found that reduced MPST/H2S exacerbates the pathological changes associated with MetS and contributes to vascular inflammation. H2S supplementation emerges as a potential therapeutic approach to treat the abnormalities associated with MetS.
Collapse
Affiliation(s)
- Paraskevas Zampas
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, Greece; Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Zhen Li
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Antonia Katsouda
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, Greece; British Heart Foundation Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, England, UK
| | - Aimilia Varela
- Cardiovascular Laboratory, Biomedical Research Foundation Academy of Athens, Greece
| | - Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Greece
| | - Constantinos H Davos
- Cardiovascular Laboratory, Biomedical Research Foundation Academy of Athens, Greece
| | - David J Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andreas Papapetropoulos
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, Greece; Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
21
|
He H, Huang W, Pan Z, Wang L, Yang Z, Chen Z. Intercellular Mitochondrial transfer: Therapeutic implications for energy metabolism in heart failure. Pharmacol Res 2025; 211:107555. [PMID: 39710083 DOI: 10.1016/j.phrs.2024.107555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Heart failure (HF) remains one of the leading causes of high morbidity and mortality globally. Impaired cardiac energy metabolism plays a critical role in the pathological progression of HF. Various forms of HF exhibit marked differences in energy metabolism, particularly in mitochondrial function and substrate utilization. Recent studies have increasingly highlighted that improving energy metabolism in HF patients as a crucial treatment strategy. Mitochondrial transfer is emerging as a promising and precisely regulated therapeutic strategy for treating metabolic disorders. This paper specifically reviews the characteristics of mitochondrial energy metabolism across different types of HF and explores the modes and mechanisms of mitochondrial transfer between different cell types in the heart, such as cardiomyocytes, fibroblasts, and immune cells. We focused on the therapeutic potential of intercellular mitochondrial transfer in improving energy metabolism disorders in HF. We also discuss the role of signal transduction in mitochondrial transfer, highlighting that mitochondria not only function as energy factories but also play crucial roles in intercellular communication, metabolic regulation, and tissue repair. This study provides new insights into improving energy metabolism in heart failure patients and proposes promising new therapeutic strategies.
Collapse
Affiliation(s)
- Huan He
- State Key Laboratory of Traditional Chinese Medicine Syndrome, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Weiwei Huang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Zigang Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Lingjun Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, PR China
| | - Zhongqi Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, PR China.
| | - Zixin Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, PR China.
| |
Collapse
|
22
|
Mao X, Liu T, Yu S, Wei Y, Zhou C, Kuai X. CEACAM6 facilitates gastric cancer progression through upregulating SLC27A2. Cancer Gene Ther 2025; 32:51-60. [PMID: 39562695 DOI: 10.1038/s41417-024-00846-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 11/21/2024]
Abstract
Gastric cancer (GC) is one of the most lethal cancers. However, the underlying mechanisms are not yet fully understood. Here, we investigated the role of carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) in tumor initiation and progression in GC and proposed therapeutic strategies for CEACAM6-positive patients. In this article, we found that CEACAM6 overexpression promoted GC initiation and progression by overactivating FAO. CEACAM6 promotes SLC27A2 expression, contributing to enhanced fatty acid incorporation. CEACAM6 interacts with both SLC27A2 and USP29, facilitating the deubiquitination of USP29 on SLC27A2. Pharmacological inhibition of SLC27A2 attenuates the tumor-initiating ability of GC. Taken together, CEACAM6 overexpression facilitates GC progression by upregulating fatty acid uptake through SLC27A2, thereby contributing to FAO. Genetic ablation of SLC27A2 is a promising therapeutic strategy for patients with CEACAM6-positive GC.
Collapse
Affiliation(s)
- Xiaqiong Mao
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tongtai Liu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, China
| | - Shunying Yu
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yuqi Wei
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Chunli Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China.
| | - Xiaoyi Kuai
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China.
| |
Collapse
|
23
|
Sun Q, Karwi QG, Wong N, Lopaschuk GD. Advances in myocardial energy metabolism: metabolic remodelling in heart failure and beyond. Cardiovasc Res 2024; 120:1996-2016. [PMID: 39453987 PMCID: PMC11646102 DOI: 10.1093/cvr/cvae231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/28/2024] [Accepted: 07/03/2024] [Indexed: 10/27/2024] Open
Abstract
The very high energy demand of the heart is primarily met by adenosine triphosphate (ATP) production from mitochondrial oxidative phosphorylation, with glycolysis providing a smaller amount of ATP production. This ATP production is markedly altered in heart failure, primarily due to a decrease in mitochondrial oxidative metabolism. Although an increase in glycolytic ATP production partly compensates for the decrease in mitochondrial ATP production, the failing heart faces an energy deficit that contributes to the severity of contractile dysfunction. The relative contribution of the different fuels for mitochondrial ATP production dramatically changes in the failing heart, which depends to a large extent on the type of heart failure. A common metabolic defect in all forms of heart failure [including heart failure with reduced ejection fraction (HFrEF), heart failure with preserved EF (HFpEF), and diabetic cardiomyopathies] is a decrease in mitochondrial oxidation of pyruvate originating from glucose (i.e. glucose oxidation). This decrease in glucose oxidation occurs regardless of whether glycolysis is increased, resulting in an uncoupling of glycolysis from glucose oxidation that can decrease cardiac efficiency. The mitochondrial oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in HFpEF and diabetic cardiomyopathies myocardial fatty acid oxidation increases, while in HFrEF myocardial fatty acid oxidation either decreases or remains unchanged. The oxidation of ketones (which provides the failing heart with an important energy source) also differs depending on the type of heart failure, being increased in HFrEF, and decreased in HFpEF and diabetic cardiomyopathies. The alterations in mitochondrial oxidative metabolism and glycolysis in the failing heart are due to transcriptional changes in key enzymes involved in the metabolic pathways, as well as alterations in redox state, metabolic signalling and post-translational epigenetic changes in energy metabolic enzymes. Of importance, targeting the mitochondrial energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac function and cardiac efficiency in the failing heart.
Collapse
Affiliation(s)
- Qiuyu Sun
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John’s, NL A1B 3V6, Canada
| | - Nathan Wong
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
24
|
Chang JWH, Chen S, Hamilton C, Shanks J, Pachen M, Pauza A, George B, Ramchandra R. Characterization of a novel ovine model of hypertensive heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol 2024; 327:H1490-H1502. [PMID: 39546298 PMCID: PMC11684944 DOI: 10.1152/ajpheart.00548.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
The lack of animal models that accurately represent heart failure with preserved ejection fraction (HFpEF) has been a major barrier to the mechanistic understanding and development of effective therapies for this prevalent and debilitating syndrome characterized by multisystem impairments. Herein, we describe the development and characterization of a novel large animal model of HFpEF in older, female sheep with chronic 2-kidney, 1-clip hypertension. At 6-wk post unilateral renal artery clipping, hypertensive HFpEF sheep had higher mean arterial pressure compared with similarly aged ewes without unilateral renal artery clipping (mean arterial pressure = 112.7 ± 15.9 vs. 76.0 ± 10.1 mmHg, P < 0.0001). The hypertensive HFpEF sheep were characterized by 1) echocardiographic evidence of diastolic dysfunction (lateral e' = 0.11 ± 0.02 vs. 0.14 ± 0.04 m/s, P = 0.011; lateral E/e' = 4.25 ± 0.77 vs. 3.63 ± 0.54, P = 0.028) and concentric left ventricular hypertrophy without overt systolic impairment, 2) elevated directly measured left ventricular end-diastolic pressure (13 ± 5 vs. 0.5 ± 1 mmHg, P = 2.1 × 10-6), and 3) normal directly measured cardiac output. Crucially, these hypertensive HFpEF sheep had impaired exercise capacity as demonstrated by their 1) attenuated cardiac output (P = 0.001), 2) augmented pulmonary capillary wedge pressure (P = 0.026), and 3) attenuated hindlimb blood flow (P = 3.4 × 10-4) responses, during graded treadmill exercise testing. In addition, exercise renal blood flow responses were also altered. Collectively, our data indicates that this novel ovine model of HFpEF may be a useful translational research tool because it exhibits similar and clinically relevant impairments as that of patients with HFpEF.NEW & NOTEWORTHY We show that older, female sheep with chronic 2-kidney, 1-clip hypertension have similar cardiac and noncardiac exercise hemodynamic abnormalities as patients with HFpEF. This clinically relevant, translatable, and novel large animal model of HFpEF may be useful for elucidating mechanisms and developing treatments for this increasingly common syndrome with few clinically impactful therapies.
Collapse
Affiliation(s)
- Joshua W-H Chang
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Siyi Chen
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Charlotte Hamilton
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Julia Shanks
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Mridula Pachen
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Audrys Pauza
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Bindu George
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Rohit Ramchandra
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
25
|
Moraña-Fernández S, Vázquez-Abuín X, Aragón-Herrera A, Anido-Varela L, García-Seara J, Otero-García Ó, Rodríguez-Penas D, Campos-Toimil M, Otero-Santiago M, Rodrigues A, Gonçalves A, Pereira Morais J, Alves IN, Sousa-Mendes C, Falcão-Pires I, González-Juanatey JR, Feijóo-Bandín S, Lago F. Cardiometabolic effects of sacubitril/valsartan in a rat model of heart failure with preserved ejection fraction. Biochem Pharmacol 2024; 230:116571. [PMID: 39424202 DOI: 10.1016/j.bcp.2024.116571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/30/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
The promising results obtained in the PARADIGM-HF trial prompted the approval of sacubitril/valsartan (SAC/VAL) as a first-in-class treatment for heart failure with reduced ejection fraction (HFrEF) patients. The effect of SAC/VAL treatment was also studied in patients with heart failure with preserved ejection fraction (HFpEF) and, although improvements in New York Heart Association (NYHA) class, HF hospitalizations, and cardiovascular deaths were observed, these results were not so promising. However, the demand for HFpEF therapies led to the approval of SAC/VAL as an alternative treatment, although further studies are needed. We aimed to elucidate the effects of a 9-week SAC/VAL treatment in cardiac function and metabolism using a preclinical model of HFpEF, the Zucker Fatty and Spontaneously Hypertensive (ZSF1) rats. We found that SAC/VAL significantly improved diastolic function parameters and modulated respiratory quotient during exercise. Ex-vivo studies showed that SAC/VAL treatment significantly decreased heart, liver, spleen, and visceral fat weights; cardiac hypertrophy and percentage of fibrosis; lipid infiltration in liver and circulating levels of cholesterol and sodium. Moreover, SAC/VAL reduced glycerophospholipids, cholesterol, and cholesteryl esters while increasing triglyceride levels in cardiac tissue. In conclusion, SAC/VAL treatment improved diastolic and hepatic function, respiratory metabolism, reduced hypercholesterolemia and cardiac fibrosis and hypertrophy, and was able to modulate cardiac metabolic profile. Our findings might provide further insight into the therapeutic benefits of SAC/VAL treatment in obese patients with HFpEF.
Collapse
Affiliation(s)
- Sandra Moraña-Fernández
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Xocas Vázquez-Abuín
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Alana Aragón-Herrera
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain.
| | - Laura Anido-Varela
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier García-Seara
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Arrhytmia Unit, Cardiology Department, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Óscar Otero-García
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Cardiology Department, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain
| | - Diego Rodríguez-Penas
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Cardiology Department Clinical Trial Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain
| | - Manuel Campos-Toimil
- Physiology and Pharmacology of Chronic Diseases (FIFAEC), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Otero-Santiago
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Clinical Biochemistry Laboratory, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain
| | - Alexandre Rodrigues
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Alexandre Gonçalves
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Juliana Pereira Morais
- CINTESIS@RISE, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, UnIC@RISE - Cardiovascular Research Centre, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Inês N Alves
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Cláudia Sousa-Mendes
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Inês Falcão-Pires
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain
| | - Sandra Feijóo-Bandín
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Área Sanitaria Santiago de Compostela e Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
26
|
Lopaschuk GD, Sun Q, Ketema EB. Glycolysis in heart failure with preserved ejection fraction. Eur J Heart Fail 2024; 26:2576-2578. [PMID: 39192674 DOI: 10.1002/ejhf.3432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Qiuyu Sun
- Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Ezra B Ketema
- Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
Comella F, Aragón-Herrera A, Pirozzi C, Feijóo-Bandin S, Lama A, Opallo N, Melini S, Del Piano F, Gualillo O, Meli R, Mattace Raso G, Lago F. Oleoylethanolamide mitigates cardiometabolic disruption secondary to obesity induced by high-fat diet in mice. Life Sci 2024; 359:123226. [PMID: 39515418 DOI: 10.1016/j.lfs.2024.123226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Chronic lipid overnutrition has been demonstrated to promote cardiac dysfunction resulting from metabolic derangement, inflammation, and fibrosis. Oleoylethanolamide (OEA), an endogenous peroxisome proliferator activating receptor (PPAR)-α agonist, has been extensively studied for its metabolic properties. The aim of this study was to determine if OEA has beneficial effects on high-fat diet (HFD)-induced cardiac disruption in obese mice, focusing on the underlying pathological mechanisms. OEA treatment restores the metabolic pattern, improving serum glycaemic and lipid profile. OEA also reduces heart weight and serum creatine kinase-myocardial band (CK-MB), a marker of cardiac damage. Accordingly, OEA modulates cardiac metabolism, increasing insulin signaling and reducing lipid accumulation. OEA increases AMPK and AKT phosphorylation, converging in the rise of AS160 activation and glucose transporter (GLUT)4 protein level. Moreover, OEA reduces the transcription of the cardiac fatty acid transporter CD36 and fatty acid synthase and increases PPAR-α mRNA levels. Adiponectin and meteorite-like protein transcription levels were significantly reduced by OEA in HFD mice, as well as those of inflammatory cytokines and pro-fibrotic markers. An increased autophagic process was also shown, contributing to OEA's cardioprotective effects. Metabolomic analyses of cardiac tissue revealed the modulation of different lipids, including triglycerides, glycerophospholipids and sphingomyelins by OEA treatment. In vitro experiments on HL-1 cardiomyocytes showed OEA's capability in reducing inflammation and fibrosis following palmitate challenge, demonstrating a direct activity of OEA on cardiac cells, mainly mediated by PPAR-α activation. Our results indicate OEA as a potential therapeutic to restrain cardiac damage associated with metabolic disorders.
Collapse
Affiliation(s)
- Federica Comella
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Alana Aragón-Herrera
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Área Sanitaria de Santiago de Compostela y Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Sandra Feijóo-Bandin
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Área Sanitaria de Santiago de Compostela y Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Nicola Opallo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Stefania Melini
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Filomena Del Piano
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II, 80100 Naples, Italy
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Group (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Building C, Travesía da Choupana SIN, Santiago de Compostela 15706, Spain; International PhD School, University of Santiago de Compostela (EDIUS), Santiago de Compostela 15706, Spain
| | - Rosaria Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, 80131 Naples, Italy.
| | - Francisca Lago
- Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Área Sanitaria de Santiago de Compostela y Barbanza (SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
28
|
Zhang Z, Wang Y, Chen X, Wu C, Zhou J, Chen Y, Liu X, Tang X. The aging heart in focus: The advanced understanding of heart failure with preserved ejection fraction. Ageing Res Rev 2024; 101:102542. [PMID: 39396676 DOI: 10.1016/j.arr.2024.102542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 10/15/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for 50 % of heart failure (HF) cases, making it the most common type of HF, and its prevalence continues to increase in the aging society. HFpEF is a systemic syndrome resulting from many risk factors, such as aging, metabolic syndrome, and hypertension, and its clinical features are highly heterogeneous in different populations. HFpEF syndrome involves the dysfunction of multiple organs, including the heart, lung, muscle, and vascular system. The heart shows dysfunction of various cells, including cardiomyocytes, endothelial cells, fibroblasts, adipocytes, and immune cells. The complex etiology and pathobiology limit experimental research on HFpEF in animal models, delaying a comprehensive understanding of the mechanisms and making treatment difficult. Recently, many scientists and cardiologists have attempted to improve the clinical outcomes of HFpEF. Recent advances in clinically related animal models and systemic pathology studies have improved our understanding of HFpEF, and clinical trials involving sodium-glucose cotransporter 2 inhibitors have significantly enhanced our confidence in treating HFpEF. This review provides an updated comprehensive discussion of the etiology and pathobiology, molecular and cellular mechanisms, preclinical animal models, and therapeutic trials in animals and patients to enhance our understanding of HFpEF and improve clinical outcomes.
Collapse
Affiliation(s)
- Zhewei Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China; Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yu Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiangqi Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chuan Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China
| | - Jingyue Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China
| | - Yan Chen
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaojing Liu
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China.
| |
Collapse
|
29
|
Chen J, Wang B, Meng T, Li C, Liu C, Liu Q, Wang J, Liu Z, Zhou Y. Oxidative Stress and Inflammation in Myocardial Ischemia-Reperfusion Injury: Protective Effects of Plant-Derived Natural Active Compounds. J Appl Toxicol 2024. [PMID: 39482870 DOI: 10.1002/jat.4719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
Acute myocardial infarction (AMI) remains a leading cause of death among patients with cardiovascular diseases. Percutaneous coronary intervention (PCI) has been the preferred clinical treatment for AMI due to its safety and efficiency. However, research indicates that the rapid restoration of myocardial oxygen supply following PCI can lead to secondary myocardial injury, termed myocardial ischemia-reperfusion injury (MIRI), posing a grave threat to patient survival. Despite ongoing efforts, the mechanisms underlying MIRI are not yet fully elucidated. Among them, oxidative stress and inflammation stand out as critical pathophysiological mechanisms, playing significant roles in MIRI. Natural compounds have shown strong clinical therapeutic potential due to their high efficacy, availability, and low side effects. Many current studies indicate that natural compounds can mitigate MIRI by reducing oxidative stress and inflammatory responses. Therefore, this paper reviews the mechanisms of oxidative stress and inflammation during MIRI and the role of natural compounds in intervening in these processes, aiming to provide a basis and reference for future research and development of drugs for treating MIRI.
Collapse
Affiliation(s)
- Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiameng Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiping Liu
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
30
|
Martinez CS, Zheng A, Xiao Q. Mitochondrial Reactive Oxygen Species Dysregulation in Heart Failure with Preserved Ejection Fraction: A Fraction of the Whole. Antioxidants (Basel) 2024; 13:1330. [PMID: 39594472 PMCID: PMC11591317 DOI: 10.3390/antiox13111330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a multifarious syndrome, accounting for over half of heart failure (HF) patients receiving clinical treatment. The prevalence of HFpEF is rapidly increasing in the coming decades as the global population ages. It is becoming clearer that HFpEF has a lot of different causes, which makes it challenging to find effective treatments. Currently, there are no proven treatments for people with deteriorating HF or HFpEF. Although the pathophysiologic foundations of HFpEF are complex, excessive reactive oxygen species (ROS) generation and increased oxidative stress caused by mitochondrial dysfunction seem to play a critical role in the pathogenesis of HFpEF. Emerging evidence from animal models and human myocardial tissues from failed hearts shows that mitochondrial aberrations cause a marked increase in mitochondrial ROS (mtROS) production and oxidative stress. Furthermore, studies have reported that common HF medications like beta blockers, angiotensin receptor blockers, angiotensin-converting enzyme inhibitors, and mineralocorticoid receptor antagonists indirectly reduce the production of mtROS. Despite the harmful effects of ROS on cardiac remodeling, maintaining mitochondrial homeostasis and cardiac functions requires small amounts of ROS. In this review, we will provide an overview and discussion of the recent findings on mtROS production, its threshold for imbalance, and the subsequent dysfunction that leads to related cardiac and systemic phenotypes in the context of HFpEF. We will also focus on newly discovered cellular and molecular mechanisms underlying ROS dysregulation, current therapeutic options, and future perspectives for treating HFpEF by targeting mtROS and the associated signal molecules.
Collapse
Affiliation(s)
| | | | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (C.S.M.); (A.Z.)
| |
Collapse
|
31
|
Gibb AA, LaPenna K, Gaspar RB, Latchman NR, Tan Y, Choya-Foces C, Doiron JE, Li Z, Xia H, Lazaropoulos MP, Conwell M, Sharp TE, Goodchild TT, Lefer DJ, Elrod JW. Integrated systems biology identifies disruptions in mitochondrial function and metabolism as key contributors to heart failure with preserved ejection fraction (HFpEF). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.619450. [PMID: 39484400 PMCID: PMC11527111 DOI: 10.1101/2024.10.25.619450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Background Heart failure with preserved ejection fraction (HFpEF) accounts for ~50% of HF cases, with no effective treatments. The ZSF1-obese rat model recapitulates numerous clinical features of HFpEF including hypertension, obesity, metabolic syndrome, exercise intolerance, and LV diastolic dysfunction. Here, we utilized a systems-biology approach to define the early metabolic and transcriptional signatures to gain mechanistic insight into the pathways contributing to HFpEF development. Methods Male ZSF1-obese, ZSF1-lean hypertensive controls, and WKY (wild-type) controls were compared at 14w of age for extensive physiological phenotyping and LV tissue harvesting for unbiased metabolomics, RNA-sequencing, and assessment of mitochondrial morphology and function. Utilizing ZSF1-lean and WKY controls enabled a distinction between hypertension-driven molecular changes contributing to HFpEF pathology, versus hypertension + metabolic syndrome. Results ZSF1-obese rats displayed numerous clinical features of HFpEF. Comparison of ZSF1-lean vs WKY (i.e., hypertension-exclusive effects) revealed metabolic remodeling suggestive of increased aerobic glycolysis, decreased β-oxidation, and dysregulated purine and pyrimidine metabolism with few transcriptional changes. ZSF1-obese rats displayed worsened metabolic remodeling and robust transcriptional remodeling highlighted by the upregulation of inflammatory genes and downregulation of the mitochondrial structure/function and cellular metabolic processes. Integrated network analysis of metabolomic and RNAseq datasets revealed downregulation of nearly all catabolic pathways contributing to energy production, manifesting in a marked decrease in the energetic state (i.e., reduced ATP/ADP, PCr/ATP). Cardiomyocyte ultrastructure analysis revealed decreased mitochondrial area, size, and cristae density, as well as increased lipid droplet content in HFpEF hearts. Mitochondrial function was also impaired as demonstrated by decreased substrate-mediated respiration and dysregulated calcium handling. Conclusions Collectively, the integrated omics approach applied here provides a framework to uncover novel genes, metabolites, and pathways underlying HFpEF, with an emphasis on mitochondrial energy metabolism as a potential target for intervention.
Collapse
Affiliation(s)
- Andrew A. Gibb
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, KY, USA
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Kyle LaPenna
- Cardiovascular Center for Excellence, Department of Pharmacology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Ryan B. Gaspar
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Nadina R. Latchman
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Yinfei Tan
- Fox Chase Cancer Center, Temple University, Philadelphia, PA, USA
| | - Carmen Choya-Foces
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jake E. Doiron
- Cardiovascular Center for Excellence, Department of Pharmacology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Zhen Li
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Huijing Xia
- Cardiovascular Center for Excellence, Department of Pharmacology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Michael P. Lazaropoulos
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mariell Conwell
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Thomas E. Sharp
- Department of Molecular Pharmacology and Physiology, University of South Florida Health, Tampa, FL, USA
| | - Traci T. Goodchild
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David J. Lefer
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - John W. Elrod
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
32
|
Werbner B, Stephens SL, Stuart D, Hotchkiss TM, Chapman J, Funai K, Ramkumar N, Boudina S. Hypertension and obesity independently drive hypertrophy and alter mitochondrial metabolism in a mouse model of heart failure with preserved ejection fraction. Physiol Rep 2024; 12:e70072. [PMID: 39776299 PMCID: PMC11427896 DOI: 10.14814/phy2.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/31/2024] [Accepted: 09/20/2024] [Indexed: 01/11/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) has recently emerged as an insidiously and increasingly prevalent heart failure phenotype. HFpEF often occurs in the context of hypertension and obesity and presents with diastolic dysfunction, ventricular hypertrophy, and myocardial fibrosis. Despite growing study of HFpEF, the causal links between early metabolic changes, bioenergetic perturbations, and cardiac structural remodeling remain unclear. This study sought to elucidate the contribution of the respective pathophysiological drivers of the HFpEF symptom suite using a recently developed two-hit mouse model. By studying the independent and concomitant consequences of hypertension and obesity-driven metabolic dysfunction on cardiac structure and function, we revealed the causative drivers of cardiac functional, structural, and metabolic remodeling in male HFpEF mice. We found that hypertensive male mice developed diastolic dysfunction and cardiac hypertrophy regardless of obesity status and that obese mice exhibited altered systemic glucose metabolism and increased cardiac mitochondrial fatty-acid metabolism independent of hypertension status. Taken together, our results suggest that the cardiac structural and metabolic HFpEF symptoms in this two-hit model occur as direct results of each of the two "hits." The results of this study help to clarify the pathogenic HFpEF cascade, providing causal insights that may aid in the development of more precisely targeted therapeutics.
Collapse
Affiliation(s)
- Benjamin Werbner
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Sophie L. Stephens
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Deborah Stuart
- Division of Nephrology and HypertensionUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Travis M. Hotchkiss
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Jonathan Chapman
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Katsuhiko Funai
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Nirupama Ramkumar
- Division of Nephrology and HypertensionUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Sihem Boudina
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
33
|
Bhattarai N, Scott I. In the heart and beyond: Mitochondrial dysfunction in heart failure with preserved ejection fraction (HFpEF). Curr Opin Pharmacol 2024; 76:102461. [PMID: 38759430 PMCID: PMC11176012 DOI: 10.1016/j.coph.2024.102461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/19/2024] [Accepted: 04/21/2024] [Indexed: 05/19/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major cardiovascular disorder with increasing prevalence and a limited range of targeted treatment options. While HFpEF can be derived from several different etiologies, much of the current growth in the disease is being driven by metabolic dysfunction (e.g. obesity, diabetes, hypertension). Deleterious changes in mitochondrial energy metabolism are a common feature of HFpEF, and may help to drive the progression of the disease. In this brief article we aim to review various aspects of cardiac mitochondrial dysfunction in HFpEF, discuss the emerging topic of HFpEF-driven mitochondrial dysfunction in tissues beyond the heart, and examine whether supporting mitochondrial function may be a therapeutic approach to arrest or reverse disease development.
Collapse
Affiliation(s)
- Nisha Bhattarai
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Iain Scott
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
34
|
Kass DA. Benchmarking pre-clinical heart failure with preserved ejection fraction models: can we do better? Cardiovasc Res 2024; 120:e20-e21. [PMID: 38572828 PMCID: PMC11074786 DOI: 10.1093/cvr/cvae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/14/2024] [Indexed: 04/05/2024] Open
Affiliation(s)
- David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Russ Building, Rm 858, 720 Rutland Avenue, Baltimore 21205, MD, USA
| |
Collapse
|
35
|
Aidara ML, Walsh-Wilkinson É, Thibodeau SÈ, Labbé EA, Morin-Grandmont A, Gagnon G, Boudreau DK, Arsenault M, Bossé Y, Couët J. Cardiac reverse remodeling in a mouse model with many phenotypical features of heart failure with preserved ejection fraction: effects of modifying lifestyle. Am J Physiol Heart Circ Physiol 2024; 326:H1017-H1036. [PMID: 38363584 DOI: 10.1152/ajpheart.00462.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/17/2024]
Abstract
Multiple factors cause heart failure with preserved ejection fraction (HFpEF) and involve various systems. HFpEF prevalence is rapidly rising, and its prognosis remains poor after the first hospitalization. Adopting a more active lifestyle has been shown to provide beneficial clinical outcomes for patients with HFpEF. Using a two-hit HfpEF murine model, we studied cardiac reverse remodeling (RR) after stopping the causing stress and introducing voluntary exercise (VE). We checked in 2-mo-old male and female C57Bl6/J mice the heart's response to angiotensin II (ANG II; 1.5 mg/kg/day for 28 days) fed or not with a high-fat diet (HFD). Then, ANG II and/or the HFD were stopped, and VE was started for an additional 4 wk. ANG II and ANG II + HFD (metabolic-hypertensive stress, MHS) caused cardiac hypertrophy (CH) and myocardial fibrosis, left ventricular (LV) concentric remodeling, atrial enlargement, and reduced exercise capacity. HFD alone induced CH and LV concentric remodeling in female mice only. CH and LV concentric remodeling were reversed 4 wk after stopping ANG II, starting VE, and a low-fat diet. Left atrial enlargement and exercise capacity were improved but differed from controls. We performed bulk LV RNA sequencing and observed that MHS upregulated 58% of the differentially expressed genes (DEGs) compared with controls. In the RR group, compared with MHS animals, 60% of the DEGs were downregulated. In an HfpEF mouse model, we show that correcting hypertension, diet, and introducing exercise can lead to extensive cardiac reverse remodeling.NEW & NOTEWORTHY Using a two-hit murine model of heart failure with preserved ejection fraction (HfpEF), combining elevated blood pressure, obesity, and exercise intolerance in male and female animals, we showed that correction of hypertension, normalization of the diet, and introduction of voluntary exercise could help reverse the remodeling of the left ventricle and double exercise capacity. We also identify genes that escape normalization after myocardial recovery and differences between males' and females' responses to stress and recovery.
Collapse
Affiliation(s)
- Mohamed Lamine Aidara
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Élisabeth Walsh-Wilkinson
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Sara-Ève Thibodeau
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Emylie-Ann Labbé
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Audrey Morin-Grandmont
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Geneviève Gagnon
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Dominique K Boudreau
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Marie Arsenault
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Yohan Bossé
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Jacques Couët
- Groupe de recherche sur les valvulopathies, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| |
Collapse
|