1
|
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
|
2
|
Pugliese NR, Paneni F, Tricò D, Bacca AV, De Biase N, Dalpiaz H, Mengozzi A, Virdis A, Ghiadoni L, Taddei S, Kreutz R, Tsioufis K, Masi S. Refining the link between obesity and heart failure: insights from GLP-1 receptor agonist trials and studies adopting direct adiposity measures. Cardiovasc Diabetol 2025; 24:224. [PMID: 40405237 PMCID: PMC12096527 DOI: 10.1186/s12933-025-02778-6] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025] Open
Abstract
Overweight and obesity are major risk factors for heart failure (HF), contributing to its development through metabolic, neurohormonal, haemodynamic, and inflammatory alterations. While overweight/obesity increases the risk of developing HF, its impact on patient outcomes remains complex. The "obesity paradox" suggests that a higher BMI may be associated with improved survival in patients with established HF. However, recent GLP-1 receptor agonist (GLP-1 RA) trials suggest that intentional weight loss positively influences outcomes in overweight/obese patients with HF. This seemingly contradictory evidence highlights the need for a deeper understanding of the mechanisms linking adiposity to HF outcomes. A more precise characterization of adiposity phenotypes using alternative and accurate measures of pathological fat accumulation is crucial in identifying individuals who may benefit most from anti-obesity treatments. In this context, recent research underscores the role of epicardial adipose tissue (EAT) in HF pathophysiology, as it directly influences cardiac function and structure through inflammatory, metabolic, and mechanical effects. This narrative review summarises current evidence on the impact of weight loss on HF outcomes, focusing on recent GLP-1 RA trial results. Additionally, it highlights epidemiological and molecular data supporting EAT as a novel adiposity measure that might allow refining patient selection for pharmacological weight-loss treatments. Finally, it emphasizes the need for future research to identify causal pathways linking alternative measures of visceral fat accumulation to HF outcomes. These efforts will be essential in optimizing the benefits of novel weight-loss treatments, ensuring effective and individualized therapeutic strategies for overweight or obese patients with HF.
Collapse
Affiliation(s)
- Nicola Riccardo Pugliese
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | | | - Nicolò De Biase
- PhD Program in Clinical and Translational Science, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Hermann Dalpiaz
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Lorenzo Ghiadoni
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Reinhold Kreutz
- Institute of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Konstantinos Tsioufis
- Hippokration Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy.
| |
Collapse
|
3
|
Zhang SJ, Wang SW, Liu SY, Li P, Huang DL, Zeng XX, Lan T, Ruan YP, Shi HJ, Zhang X. Epicardial adipose tissue: a new link between type 2 diabetes and heart failure-a comprehensive review. Heart Fail Rev 2025; 30:477-491. [PMID: 39730926 DOI: 10.1007/s10741-024-10478-8] [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] [Accepted: 12/18/2024] [Indexed: 12/29/2024]
Abstract
Diabetic cardiomyopathy is a unique cardiomyopathy that is common in diabetic patients, and it is also a diabetic complication for which no effective treatment is currently available. Moreover, relevant studies have revealed that a link exists between type 2 diabetes and heart failure and that abnormal thickening of EAT is inextricably linked to the development of diabetic heart failure. Numerous clinical studies have demonstrated that EAT is implicated in the pathophysiologic process of diabetic myocardial disease. In this overview, we will introduce the physiology, pathophysiology of the disease and potential therapeutic strategies, knowledge gaps, and future directions of the role of epicardial adipose tissue in type 2 diabetes mellitus and heart failure to promote the development of novel therapeutic approaches to improve the prognosis of patients with diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Si-Jia Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
| | - Si-Wei Wang
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
- Laboratory Animal Resources Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Shi-Yu Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
| | - Ping Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
| | - De-Lian Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
| | - Xi-Xi Zeng
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Tian Lan
- Panvascular Diseases Research Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
- Laboratory Animal Resources Center, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Ye-Ping Ruan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hai-Jiao Shi
- The Third Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Liaoning, 116600, China.
| | - Xin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China.
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
4
|
Li B, Gao Y, Wang W, Zhu R, Yang X, Chen H, Wang X, Gu H. Epicardial Adipose Tissue and Extracellular Volume Fraction in Patients with Hypertrophic Cardiomyopathy: A Multi‑center Prognosis Study. Acad Radiol 2025:S1076-6332(25)00314-9. [PMID: 40268603 DOI: 10.1016/j.acra.2025.04.015] [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: 01/26/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025]
Abstract
RATIONALE AND OBJECTIVES Epicardial adipose tissue (EAT) is thought to have a deleterious effect on the progression of myocardial disease; the extracellular volume (ECV) fraction has been validated by histology to correlate with adverse myocardial remodeling. The aim of this study was to investigate the prognostic value of EAT volume index (EATVI) and ECV in patients with hypertrophic cardiomyopathy (HCM). MATERIALS AND METHODS ECV and EAT were measured using cardiac magnetic resonance (CMR) imaging in 201 subjects with HCM. All patients were followed up prospectively. Major adverse cardiovascular events (MACEs) were categorized into primary and secondary endpoint events. The primary endpoint was a composite of cardiac death, heart transplant, and cardiopulmonary resuscitation following syncope. The secondary endpoint was defined as rehospitalization for heart failure (HF). RESULTS After 26±16 months of follow-up, 43 patients experienced MACEs (14 patients experienced a primary endpoint, and 29 patients experienced a secondary endpoint). Patients suffering from MACEs had significantly higher ECV and EATVI (p<0.001). After adjustment for body mass index (BMI), EATVI showed a significant correlation with ECV (r=0.424, p<0.001) among HCM patients. In the Kaplan-Meier analysis, the incidence of MACE was significantly higher in patients with increased ECV (p<0.001) and higher EATVI (p<0.001). In multivariate Cox regression analysis, ECV (HR=1.12, p<0.001) and EATVI (HR=1.31, p<0.001) were significantly associated with MACEs. CONCLUSION In patients with HCM, ECV and EATVI measured by CMR are strong predictors of MACEs and improve risk stratification. CLINICAL IMPLICATION Extracellular volume fraction and epicardial adipose tissue are associated with major adverse cardiovascular events in patients with hypertrophic cardiomyopathy and can improve their risk stratification.
Collapse
Affiliation(s)
- Bowen Li
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250117, China (B.L., R.Z., X.Y., H.C.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.)
| | - Yan Gao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.); Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China (Y.G., X.Y., X.W.)
| | - Wenxian Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.); School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Yantai, Shandong 264003, PR China (W.W., X.Y.)
| | - Runze Zhu
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250117, China (B.L., R.Z., X.Y., H.C.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.)
| | - Xueqiao Yang
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250117, China (B.L., R.Z., X.Y., H.C.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.); Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China (Y.G., X.Y., X.W.); School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Yantai, Shandong 264003, PR China (W.W., X.Y.)
| | - Huiyu Chen
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250117, China (B.L., R.Z., X.Y., H.C.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.)
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.); Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China (Y.G., X.Y., X.W.)
| | - Hui Gu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.).
| |
Collapse
|
5
|
Hsu JC, Huang KC, Lin TT, Lee JK, Su MYM, Juang JMJ, Wu CK, Lin LY. Epicardial Adipose Tissue Is Associated With Geometry Alteration and Diastolic Dysfunction in Prediabetic Cardiomyopathy. J Clin Endocrinol Metab 2025; 110:1478-1487. [PMID: 38864548 DOI: 10.1210/clinem/dgae400] [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: 02/14/2024] [Revised: 05/19/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Diastolic dysfunction and alterations in cardiac geometry are early indicators of diabetic cardiomyopathy. However, the association between cardiac changes across the glucose continuum and the contribution of epicardial adipose tissue (EAT) to these changes has not yet been investigated. PURPOSE In this study, we aimed to investigate the EAT on cardiac diastolic function and structural alterations along the diabetic continuum using cardiac magnetic resonance imaging (CMRI). METHODS We enrolled individuals who were categorized into groups based on glucose tolerance status. Left ventricular structure and diastolic function were assessed using echocardiography and CMRI to determine the EAT, intramyocardial fat, and associated parameters. Multivariable logistic regression models were also used. RESULTS In a study of 370 patients (209 normal glucose tolerance, 82 prediabetes, 79 diabetes), those with prediabetes and diabetes showed increased heart dimensions and diastolic dysfunction, including the ratio of early mitral inflow velocity to mitral annular early diastolic velocity (7.9 ± 0.51 vs 8.5 ± 0.64 vs 10.0 ± 0.93, P = .010), left atrial volume index (28.21 ± 14.7 vs 33.2 ± 12.8 vs 37.4 ± 8.2 mL/m2, P < .001), and left ventricular peak filling rate (4.46 ± 1.75 vs 3.61 ± 1.55 vs 3.20 ± 1.30 mL/s, P < .001). EAT significantly increased in prediabetes and diabetes (26.3 ± 1.16 vs 31.3 ± 1.83 vs 33.9 ± 1.9 gm, P = .001), while intramyocardial fat did not differ significantly. Prediabetes altered heart geometry but not diastolic function (odds ratio [OR] 1.22 [1.02-1.83], P = .012; and 1.70 [0.79-3.68], P = .135). Diabetes significantly affected both heart structure and diastolic function (OR 1.42 [1.11-1.97], P = .032; and 2.56 [1.03-5.40], P = .034) after adjusting for covariates. CONCLUSION Elevated EAT was observed in patients with prediabetes and is associated with adverse alterations in cardiac structure and diastolic function, potentially serving as an underlying mechanism for the early onset of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Jung-Chi Hsu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Jinshan Branch, New Taipei City 20844, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei 100225, Taiwan
| | - Kuan-Chih Huang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei 100225, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 300195, Taiwan
| | - Ting-Tse Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei 100225, Taiwan
- Department of Internal Medicine, College of Medicine National Taiwan University, Taipei 100233, Taiwan
| | - Jen-Kuang Lee
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei 100225, Taiwan
- Department of Internal Medicine, College of Medicine National Taiwan University, Taipei 100233, Taiwan
| | - Mao-Yuan M Su
- Department of Medical Imaging, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Jyh-Ming Jimmy Juang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei 100225, Taiwan
- Department of Internal Medicine, College of Medicine National Taiwan University, Taipei 100233, Taiwan
- Heart Failure Center, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Cho-Kai Wu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei 100225, Taiwan
- Department of Internal Medicine, College of Medicine National Taiwan University, Taipei 100233, Taiwan
| | - Lian-Yu Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei 100225, Taiwan
- Department of Internal Medicine, College of Medicine National Taiwan University, Taipei 100233, Taiwan
- Master's Program in Smart Medicine and Health Informatics, National Taiwan University, Taipei 106319, Taiwan
| |
Collapse
|
6
|
Janssen-Telders C, Eringa EC, de Groot JR, de Man FS, Handoko ML. The role of epicardial adipose tissue remodelling in heart failure with preserved ejection fraction. Cardiovasc Res 2025:cvaf056. [PMID: 40238568 DOI: 10.1093/cvr/cvaf056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/12/2024] [Accepted: 01/22/2025] [Indexed: 04/18/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a growing global health problem characterized by high morbidity and mortality, with limited effective therapies available. Obesity significantly influences haemodynamic and structural changes in the myocardium and vasculature, primarily through the accumulation and action of visceral adipose tissue. Particularly, epicardial adipose tissue (EAT) contributes to HFpEF through inflammation and lipotoxic infiltration of the myocardium. However, the precise signalling pathways leading to diastolic stiffness in HFpEF require further elucidation. This review explores the dynamic role of EAT in health and disease. Drawing upon insights from studies in other conditions, we discuss potential EAT-mediated inflammatory pathways in HFpEF and how they may contribute to functional and structural myocardial and endothelial derangements, including intramyocardial lipid infiltration, fibrosis, endothelial dysfunction, cardiomyocyte stiffening, and left ventricular hypertrophy. Lastly, we propose potential targets for novel therapeutic avenues.
Collapse
Affiliation(s)
- Carolina Janssen-Telders
- Department of Cardiology Amsterdam UMC, Heart Centre, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Etto C Eringa
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Department of Physiology, Amsterdam UMC, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht UMC, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Joris R de Groot
- Department of Cardiology Amsterdam UMC, Heart Centre, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Frances S de Man
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht UMC, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - M Louis Handoko
- Department of Cardiology Amsterdam UMC, Heart Centre, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Pulmonology, Amsterdam UMC, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
7
|
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
|
8
|
Ding Y, Lin F, Liu Z, Zhou X, Liang X. Targeting Epicardial/Pericardial Adipose Tissue in Cardiovascular Diseases: A Novel Therapeutic Strategy. Rev Cardiovasc Med 2025; 26:26128. [PMID: 40160564 PMCID: PMC11951288 DOI: 10.31083/rcm26128] [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: 08/14/2024] [Revised: 11/01/2024] [Accepted: 11/14/2024] [Indexed: 04/02/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain a global health concern, prompting ongoing research into novel contributors to their pathogenesis. Due to the proximity of the coronary arteries and the myocardium in epicardial adipose tissue (EAT) and pericardial adipose tissue (PAT), these tissues have emerged as key areas of interest for their potential influence on cardiac function and vascular health. This review synthesizes current research on the physiological and biological characteristics of EAT and PAT, exploring their composition and clinical measurement approaches. The roles of EAT and PAT in coronary artery disease (CAD), atrial fibrillation, and heart failure are discussed, and the contributions of EAT and PAT to these cardiovascular conditions are highlighted alongside their potential as therapeutic targets.
Collapse
Affiliation(s)
- Yue Ding
- Department of Organ Transplantation, Changzheng Hospital, Second Military Medical University, 200003 Shanghai, China
| | - Fang Lin
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Zhongmin Liu
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Xiaohui Zhou
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Xiaoting Liang
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| |
Collapse
|
9
|
Chang KC, Su TH, Wu CK, Huang SC, Tseng TC, Hong CM, Hsu SJ, Liu CH, Yang HC, Liu CJ, Kao JH. Metabolic dysfunction-associated steatotic liver disease is associated with increased risks of heart failure. Eur J Heart Fail 2025; 27:512-520. [PMID: 39777761 DOI: 10.1002/ejhf.3567] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD), defined by steatotic liver disease (SLD) and cardiometabolic factors, is increasing in prevalence, but its association with heart failure (HF) is unclear. METHODS AND RESULTS Patients with SLD without a history of HF from 2006 to 2021 were retrospectively included and were classified into MASLD and non-MASLD groups that were followed longitudinally. The primary outcome was the new development of HF, which was sub-classified by echocardiography. Multivariable and propensity score matching analyses were conducted to adjust for confounding factors. Overall, 26 676 patients with SLD were included, with a median age of 51 years and 71% classified as MASLD. During a median follow-up of 6 years, 429 (1.61%) patients developed HF, and 76% were HF with preserved ejection fraction (HFpEF). The risk of HF was significantly higher in patients with MASLD than in those without (sub-distribution hazard ratio [SHR] 2.59, 95% confidence interval [CI] 1.84-3.64) after adjustment of competing mortality. There was a dose-dependent increase in HF risks in patients with more cardiometabolic risk factors (SHR 1.12, 95% CI 1.04-1.22). MASLD was also associated with higher risk of HF-related hospitalization (SHR 2.30, 95% CI 1.31-4.04) and specifically, the risk of HFpEF (SHR 1.91, 95% CI 1.27-2.86). In propensity score-matched cohorts, MASLD was also associated with a 2.52-fold higher risk of HF. CONCLUSION In patients with SLD, those with MASLD show a higher risk of HF, specifically HFpEF. Future studies are warranted to validate the association between HF and MASLD.
Collapse
Affiliation(s)
- Kai-Chun Chang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tung-Hung Su
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Cho-Kai Wu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shang-Chin Huang
- Department of Internal Medicine, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tai-Chung Tseng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Ming Hong
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Jer Hsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Chen-Hua Liu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Hung-Chih Yang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Jen Liu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Jia-Horng Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
10
|
Kramer CM, Borlaug BA, Zile MR, Ruff D, DiMaria JM, Menon V, Ou Y, Zarante AM, Hurt KC, Murakami M, Packer M. Tirzepatide Reduces LV Mass and Paracardiac Adipose Tissue in Obesity-Related Heart Failure: SUMMIT CMR Substudy. J Am Coll Cardiol 2025; 85:699-706. [PMID: 39566869 DOI: 10.1016/j.jacc.2024.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Obesity is a known risk factor for heart failure with preserved ejection fraction (HFpEF) and is considered a distinct phenotype with more concentric remodeling. Epicardial adipose tissue (EAT) is also increased in obesity-related HFpEF and is associated with adverse events. OBJECTIVES The cardiac magnetic resonance (CMR) substudy of the SUMMIT trial aimed to examine the effects of tirzepatide on cardiac structure and function with the underlying hypothesis that it would reduce left ventricular (LV) mass and EAT in obesity-related HFpEF. METHODS A total of 175 patients with obesity-related HFpEF from the parent study of tirzepatide (2.5 mg subcutaneously weekly, increasing to a maximum of 15 mg weekly) or matching placebo underwent CMR at baseline, which consisted of multiplanar cine imaging. A total of 106 patients completed the CMR and had adequate image quality for analysis of LV and left atrial structure and function and paracardiac (epicardial plus pericardial) adipose tissue at both baseline and 52 weeks. The prespecified primary endpoint of this substudy was between-group changes in LV mass. RESULTS LV mass decreased by 11 g (95% CI: -19 to -4 g) in the treated group (n = 50) when corrected for placebo (n = 56) (P = 0.004). Paracardiac adipose tissue decreased in the treated group by 45 mL (95% CI: -69 to -22 mL) when corrected for placebo (P < 0.001). The change in LV mass in the treated group correlated with changes in body weight (P < 0.02) and tended to correlate with changes in waist circumference and blood pressure (P = 0.06 for both). The LV mass change also correlated with changes in LV end-diastolic volume and left atrial end-diastolic and end-systolic volumes (P < 0.03 for all). CONCLUSIONS The CMR substudy of the SUMMIT trial demonstrated that tirzepatide therapy in obesity-related HFpEF led to reduced LV mass and paracardiac adipose tissue as compared with placebo, and the change in LV mass paralleled weight loss. These physiologic changes may contribute to the reduction in heart failure events seen in the main SUMMIT trial. (A Study of Tirzepatide [LY3298176] in Participants With Heart Failure With Preserved Ejection Fraction [HFpEF] and Obesity: The SUMMIT Trial; NCT04847557).
Collapse
Affiliation(s)
- Christopher M Kramer
- Cardiovascular Division, Department of Medicine, University of Virginia Health, Charlottesville, Virginia, USA.
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael R Zile
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Dustin Ruff
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Joseph M DiMaria
- Cardiovascular Division, Department of Medicine, University of Virginia Health, Charlottesville, Virginia, USA
| | - Venu Menon
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yang Ou
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Karla C Hurt
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas, USA; Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Radakrishnan A, Agrawal S, Singh N, Barbieri A, Shaw LJ, Gulati M, Lala A. Underpinnings of Heart Failure With Preserved Ejection Fraction in Women - From Prevention to Improving Function. A Co-publication With the American Journal of Preventive Cardiology and the Journal of Cardiac Failure. J Card Fail 2025:S1071-9164(25)00037-5. [PMID: 39971643 DOI: 10.1016/j.cardfail.2025.01.008] [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: 07/09/2024] [Revised: 10/30/2024] [Accepted: 01/08/2025] [Indexed: 02/21/2025]
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents a major clinical challenge with rising global prevalence. Women have a nearly double lifetime risk of developing HFpEF compared to heart failure with reduced ejection fraction (HFrEF). In HFpEF, sex differences emerge both in how traditional cardiovascular risk factors (such as hypertension, obesity, and diabetes) affect cardiac function and through distinct pathophysiological mechanisms triggered by sex-specific events like menopause and adverse pregnancy outcomes. These patterns influence not only disease development, but also therapeutic responses, necessitating sex-specific approaches to treatment. This review aims to synthesize existing knowledge regarding HFpEF in women including traditional and sex-specific risk factors, pathophysiology, presentation, and therapies, while outlining important knowledge gaps that warrant further investigation. The impact of HFpEF spans a woman's entire lifespan, requiring prevention and management strategies tailored to different life stages. While understanding of sex-based differences in HFpEF has improved, significant knowledge gaps persist. Through examination of current evidence and challenges, this review highlights promising opportunities for innovative research, therapeutic development, and clinical care approaches that could transform the management of HFpEF in women.
Collapse
Affiliation(s)
- Ankitha Radakrishnan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saloni Agrawal
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nausheen Singh
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Barbieri
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Leslee J Shaw
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Martha Gulati
- Department of Cardiology, Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, California, USA.
| | - Anuradha Lala
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
12
|
Theodorakis N, Nikolaou M. From Cardiovascular-Kidney-Metabolic Syndrome to Cardiovascular-Renal-Hepatic-Metabolic Syndrome: Proposing an Expanded Framework. Biomolecules 2025; 15:213. [PMID: 40001516 PMCID: PMC11853431 DOI: 10.3390/biom15020213] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiometabolic diseases represent an escalating global health crisis, slowing or even reversing earlier declines in cardiovascular disease (CVD) mortality. Traditionally, conditions such as obesity, type 2 diabetes mellitus (T2DM), atherosclerotic CVD, heart failure (HF), chronic kidney disease (CKD), and metabolic dysfunction-associated steatotic liver disease (MASLD) were managed in isolation. However, emerging evidence reveals that these disorders share overlapping pathophysiological mechanisms and treatment strategies. In 2023, the American Heart Association proposed the Cardiovascular-Kidney-Metabolic (CKM) syndrome, recognizing the interconnected roles of the heart, kidneys, and metabolic system. Yet, this model omits the liver-a critical organ impacted by metabolic dysfunction. MASLD, which can progress to metabolic dysfunction-associated steatohepatitis (MASH), is closely tied to insulin resistance and obesity, contributing directly to cardiovascular and renal impairment. Notably, MASLD is bidirectionally associated with the development and progression of CKM syndrome. As a result, we introduce an expanded framework-the Cardiovascular-Renal-Hepatic-Metabolic (CRHM) syndrome-to more comprehensively capture the broader inter-organ dynamics. We provide guidance for an integrated diagnostic approach aimed at halting progression to advanced stages and preventing further organ damage. In addition, we highlight advances in medical management that target shared pathophysiological pathways, offering benefits across multiple organ systems. Viewing these conditions as an integrated whole, rather than as discrete entities, and incorporating the liver into this framework fosters a more holistic management strategy and offers a promising path to addressing the cardiometabolic pandemic.
Collapse
Affiliation(s)
- Nikolaos Theodorakis
- NT-CardioMetabolics, Clinic for Metabolism and Athletic Performance, 47 Tirteou Str., 17564 Palaio Faliro, Greece
- Department of Cardiology & Preventive Cardiology Outpatient Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece;
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
| | - Maria Nikolaou
- Department of Cardiology & Preventive Cardiology Outpatient Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece;
| |
Collapse
|
13
|
Zhang H, Xu T, Mei X, Zhao Q, Yang Q, Zeng X, Ma Z, Zhou H, Zeng Q, Xu D, Ren H. PINK1 modulates Prdx2 to reduce lipotoxicity-induced apoptosis and attenuate cardiac dysfunction in heart failure mice with a preserved ejection fraction. Clin Transl Med 2025; 15:e70166. [PMID: 39763059 PMCID: PMC11705485 DOI: 10.1002/ctm2.70166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Heart failure with preserved ejection fraction (HFpEF) is a complex condition characterized by metabolic dysfunction and myocardial lipotoxicity. The roles of PTEN-induced kinase 1 (PINK1) and peroxiredoxin-2 (Prdx2) in HFpEF pathogenesis remain unclear. OBJECTIVE This study aimed to investigate the interaction between PINK1 and Prdx2 to mitigate cardiac diastolic dysfunction in HFpEF. METHODS In vivo, PINK1-knockout mice and cardiac-specific PINK1-overexpressing transgenic mice were used to establish an HFpEF mouse model via a high-fat diet and L-NAME. Myocardial lipotoxicity was induced by palmitic acid in vitro. Immunoprecipitation, western blotting and immunofluorescence analysis were performed to elucidate the molecular mechanisms involved. RESULTS We determined that PINK1 and Prdx2 were downregulated in the HFpEF mouse model. In vivo, PINK1 ablation exacerbated the reduction in Prdx2 expression, worsening cardiac dysfunction in HFpEF mice. Conversely, PINK1 overexpression restored Prdx2 levels and decreased reactive oxygen species and apoptosis, thereby reducing fibrosis and inflammation and ameliorating cardiac diastolic dysfunction in HFpEF mice. In vitro, an interaction between the N-terminal region (amino acids 1-133) of PINK1 and Prdx2 was identified. The overexpression of PINK1 induced Prdx2 expression and effectively attenuated palmitic acid-induced apoptosis through the c-Jun amino-terminal kinase (JNK) and mitogen-activated protein kinase (p38) pathways, whereas siRNA-mediated Prdx2 knockdown abolished the protective effect of PINK1. CONCLUSION PINK1 alleviates lipotoxicity-induced myocardial apoptosis and improves diastolic dysfunction in HFpEF through Prdx2, highlighting PINK1 overexpression as a potential therapeutic strategy for HFpEF. KEY POINTS Our investigation discloses a pivotal relationship between PINK1 and Prdx2 in the context of HFpEF. Notably, PINK1, in addition to its role in mitochondrial autophagy, can increase Prdx2 expression, effectively remove ROS and attenuate cardiomyocyte apoptosis by modulating the JNK and p38 pathways, thereby alleviating myocardial lipotoxicity and improving HFpEF cardiac function. Our studies offer valuable insights, opening avenues for the development of innovative therapeutic strategies in the prevention and treatment of HFpEF.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Tianyu Xu
- NHC Key Laboratory of Assisted Circulation, Department of CardiologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xiyuan Mei
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Qiming Zhao
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Qiling Yang
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Xianghui Zeng
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Zhuang Ma
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Haobin Zhou
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Dingli Xu
- State Key Laboratory of Organ Failure ResearchDepartment of CardiologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
| | - Hao Ren
- Key Laboratory For Organ Failure ResearchMinistry of Education of the People's Republic of ChinaGuangzhouChina
- Department of RheumatologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
14
|
Hsueh HW, Chao CC, Lin YH, Tseng PH, Su MY, Hsieh ST. Neck triangle nerve enlargement in hereditary transthyretin amyloidosis correlates with changes in the autonomic, cardiac, and gastrointestinal systems. J Intern Med 2024; 296:495-509. [PMID: 39436674 DOI: 10.1111/joim.20019] [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] [Indexed: 10/23/2024]
Abstract
BACKGROUND Hereditary transthyretin amyloidosis (ATTRv) is a hereditary disease that affects multiple bodily systems. Although sonography generally reveals enlargement of nerves in the limbs, the brachial plexus, and vagus nerve, the clinical significance of these findings remains unclear. METHODS We performed sonographic measurements of the median nerve, cervical spinal nerves at the C5-C7 level, and the vagus nerve in patients with ATTRv and healthy controls. Clinical profiles and cardiac and gastrointestinal examination results were also collected for linear regression analysis. RESULTS We recruited 47 patients with ATTRv (males/females: 34/13, age: 65.6 ± 5.3 years). The sampled segments were all significantly larger than those of the controls. In the clinical profiles, the sum of the Z scores of the neck triangle nerves (cervical spinal nerves and vagus nerve) and of all nerves (cervical spinal nerves, vagus nerve, and median nerve at the wrist) significantly correlated with the familial amyloid polyneuropathy stage, onset of autonomic nervous system (ANS) symptoms, and autonomic symptom scores. On cardiac examinations, several ultrasonography and magnetic resonance imaging parameters (primarily those that reflect heart volume) were found to be significantly correlated with the sum of the Z scores of the cervical spinal nerves but not with the Z score of the vagus nerve. In gastrointestinal evaluation, the cross-sectional area of the vagus nerve was correlated with gastric emptying time parameters on scintigraphy. CONCLUSIONS Neck triangle nerve enlargement on sonography correlated with parameters related to ANS dysfunction, indicating that nerve enlargement observed on ultrasonography may serve as a potential surrogate biomarker of ATTRv.
Collapse
Affiliation(s)
- Hsueh-Wen Hsueh
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Chao Chao
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ping-Huei Tseng
- Division of Hepatology & Gastroenterology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Mao-Yuan Su
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Tsang Hsieh
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
15
|
Paterek A, Załęska-Kocięcka M, Wojdyńska Z, Kalisz M, Litwiniuk A, Leszek P, Mączewski M. Epicardial fat in heart failure-Friend, foe, or bystander. Obes Rev 2024; 25:e13820. [PMID: 39187402 DOI: 10.1111/obr.13820] [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: 09/30/2023] [Revised: 07/12/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024]
Abstract
Epicardial adipose tissue (EAT) is a fat depot covering the heart. No physical barrier separates EAT from the myocardium, so EAT can easily affect the underlying cardiac muscle. EAT can participate in the development and progression of heart failure with preserved (HFpEF) and reduced ejection fraction (HFrEF). In healthy humans, excess EAT is associated with impaired cardiac function and worse outcomes. In HFpEF, this trend continues: EAT amount is usually increased, and excess EAT correlates with worse function/outcomes. However, in HFrEF, the opposite is true: reduced EAT amount correlates with worse cardiac function/outcomes. Surprisingly, although EAT has beneficial effects on cardiac function, it aggravates ventricular arrhythmias. Here, we dissect these phenomena, trying to explain these paradoxical findings to find a target for novel heart failure therapies aimed at EAT rather than the myocardium itself. However, the success of this approach depends on a thorough understanding of interactions between EAT and the myocardium.
Collapse
Affiliation(s)
- Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marta Załęska-Kocięcka
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Zuzanna Wojdyńska
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Małgorzata Kalisz
- Department of Clinical Neuroendocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Anna Litwiniuk
- Department of Clinical Neuroendocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Przemysław Leszek
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
16
|
Bodenstab ML, Varghese RT, Iacobellis G. Cardio-Lipotoxicity of Epicardial Adipose Tissue. Biomolecules 2024; 14:1465. [PMID: 39595641 PMCID: PMC11591820 DOI: 10.3390/biom14111465] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Epicardial adipose tissue is a unique visceral adipose tissue depot that plays a crucial role in myocardial metabolism. Epicardial adipose tissue is a major source of energy and free fatty acids for the adjacent myocardium. However, under pathological conditions, epicardial fat can affect the heart through the excessive and abnormal influx of lipids. The cardio-lipotoxicity of the epicardial adipose tissue is complex and involves different pathways, such as increased inflammation, the infiltration of lipid intermediates such as diacylglycerol and ceramides, mitochondrial dysfunction, and oxidative stress, ultimately leading to cardiomyocyte dysfunction and coronary artery ischemia. These changes can contribute to the pathogenesis of various cardio-metabolic diseases including atrial fibrillation, coronary artery disease, heart failure, and obstructive sleep apnea. Hence, the role of the cardio-lipotoxicity of epicardial fat and its clinical implications are discussed in this review.
Collapse
Affiliation(s)
- Monica L. Bodenstab
- Department of Internal Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Ron T. Varghese
- Sleep—Endocrinology Integrated Clinic, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Gianluca Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
17
|
Harada T, Tada A, Borlaug BA. Imaging and mechanisms of heart failure with preserved ejection fraction: a state-of-the-art review. Eur Heart J Cardiovasc Imaging 2024; 25:1475-1490. [PMID: 38912836 DOI: 10.1093/ehjci/jeae152] [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: 06/03/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
Understanding of the pathophysiology of heart failure with preserved ejection fraction (HFpEF) has advanced rapidly over the past two decades. Currently, HFpEF is recognized as a heterogeneous syndrome, and there is a growing movement towards developing personalized treatments based on phenotype-guided strategies. Left ventricular dysfunction is a fundamental pathophysiological abnormality in HFpEF; however, recent evidence also highlights significant roles for the atria, right ventricle, pericardium, and extracardiac contributors. Imaging plays a central role in characterizing these complex and highly integrated domains of pathophysiology. This review focuses on established evidence, recent insights, and the challenges that need to be addressed concerning the pathophysiology of HFpEF, with a focus on imaging-based evaluations and opportunities for further research.
Collapse
Affiliation(s)
- Tomonari Harada
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Atsushi Tada
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
18
|
Nogajski Ł, Mazuruk M, Kacperska M, Kurpias M, Mączewski M, Nowakowski M, Mączewski M, Michałowska I, Leszek P, Paterek A. Epicardial fat density obtained with computed tomography imaging - more important than volume? Cardiovasc Diabetol 2024; 23:389. [PMID: 39472958 PMCID: PMC11523889 DOI: 10.1186/s12933-024-02474-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
Epicardial adipose tissue (EAT) is a unique fat depot located between the myocardium and the visceral layer of pericardium. It can be further subdivided into pericoronary (PCAT), periatrial (PAAT) and periventricular adipose tissue (PVentAT), each of them exhibiting specific characteristics and association with the underlying tissue. Since no physical barrier separates EAT from the myocardium, this fat tissue can easily interact with the underlying cardiac structure. EAT can be visualized using various imaging modalities. Computed tomography provides not only information on EAT volume, but also on its density. Indeed, EAT density reflected by the recently developed fat attenuation index (FAI) is emerging as a useful index of PCAT inflammation, PAAT inflammation and fibrosis, while the relevance of density of PVentAT is much less known. The emerging data indicates that FAI can be an important diagnostic and prognostic tool in both coronary artery disease and atrial fibrillation. Future studies will demonstrate if it also could be used as a marker of efficacy of therapies and whether FAI PVentAT could indicate ventricular pathologies, such as heart failure. The aim of the review is to present computed tomography derived FAI as an important tool both to study and better understand the epicardial fat and as a potential predictive marker in cardiovascular disorders.
Collapse
Affiliation(s)
- Łukasz Nogajski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Maciej Mazuruk
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Marta Kacperska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Mikołaj Kurpias
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Maciej Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Maksymilian Nowakowski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Ilona Michałowska
- Department of Radiology, National Institute of Cardiology, Warsaw, Poland
| | - Przemysław Leszek
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
19
|
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
|
20
|
Dronkers J, van Veldhuisen DJ, van der Meer P, Meems LMG. Heart Failure and Obesity: Unraveling Molecular Mechanisms of Excess Adipose Tissue. J Am Coll Cardiol 2024; 84:1666-1677. [PMID: 39415402 DOI: 10.1016/j.jacc.2024.07.016] [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: 04/10/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
Obesity is an ongoing pandemic and is associated with the development of heart failure (HF), and especially HF with preserved ejection fraction. The definition of obesity is currently based on anthropometric measurements but neglects the location and molecular properties of excess fat. Important depots associated with HF development are subcutaneous adipose tissue and visceral adipose tissue, both located in the abdominal region, and epicardial adipose tissue (EAT) surrounding the myocardium. However, mechanisms linking these different adipose tissue depots to HF development are incompletely understood. EAT in particular is of great interest because of its close proximity to the heart. In this review, we therefore focus on the characteristics of different adipose tissue depots and their response to obesity. In addition, we evaluate how different mechanisms associated with EAT expansion potentially contribute to HF and in particular HF with preserved ejection fraction development.
Collapse
Affiliation(s)
- Just Dronkers
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Peter van der Meer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Laura M G Meems
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands.
| |
Collapse
|
21
|
Fukuta H, Goto T, Kamiya T. Association of epicardial fat with cardiac structure and function and exercise capacity in heart failure with preserved ejection fraction: A systematic review and meta-analysis. IJC HEART & VASCULATURE 2024; 54:101444. [PMID: 39415965 PMCID: PMC11481611 DOI: 10.1016/j.ijcha.2024.101444] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/12/2024] [Accepted: 06/08/2024] [Indexed: 10/19/2024]
Abstract
Background Studies have reported the association of epicardial adipose tissue (EAT) with cardiac structure and function as well as exercise capacity in patients with heart failure with preserved ejection fraction (HFpEF), yielding inconsistent results. We aimed to conduct a meta-analysis of studies on the association of EAT with cardiac structure and function and exercise capacity in HFpEF patients. Methods and Results We searched studies examining the association of EAT quantified by echocardiography, computed tomography, or magnetic resonance imaging (MRI) with cardiac structure and function or exercise capacity in HFpEF patients through PubMed, Web of Science, and Scopus. In cases of significant heterogeneity (I2 > 50 %), data were pooled using a random-effects model; otherwise, a fixed-effects model was used. We identified five echocardiography studies (n = 825) and six MRI studies (n = 562), but found no computed tomography studies. In the echocardiography studies, EAT thickness correlated positively with left ventricular (LV) mass (P random < 0.01) and negatively with LV global longitudinal strain (P random < 0.01) and peak exercise oxygen uptake (P fix < 0.001). In the MRI studies, EAT volume correlated positively with LV mass (P fix < 0.01), left atrial volume (P fix < 0.001), and the ratio of LV early diastolic mitral inflow to early diastolic mitral annular velocity (E/e'; P random < 0.01) and negatively with LV ejection fraction (P fix < 0.01) and LV global longitudinal strain (P fix < 0.001). Conclusion Our meta-analysis indicates a potential association of increased EAT with altered cardiac structure and function and exercise intolerance in HFpEF patients. However, our meta-analysis included only two or three studies for each outcome and thus further studies are necessary to confirm our findings.
Collapse
Affiliation(s)
- Hidekatsu Fukuta
- Core Laboratory, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Toshihiko Goto
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takeshi Kamiya
- Department of Medical Innovation, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
22
|
Sidhu GS, Rabkin SW. Epicardial Fat in Heart Failure with Preserved Ejection Fraction Compared with Reduced Ejection Fraction. J Clin Med 2024; 13:5533. [PMID: 39337020 PMCID: PMC11432675 DOI: 10.3390/jcm13185533] [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: 08/07/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Background: The role of epicardial adipose tissue (EAT) in heart failure with preserved ejection fraction (HFpEF) remains to be defined. Methods: A consecutive series of outpatients with chronic heart failure-heart failure with reduced ejection fraction (HFrEF) and HFpEF and/or diastolic dysfunction-had EAT assessed by echocardiographic measurement and related to indices of cardiac structure and function. Results: Epicardial fat thickness was significantly (p < 0.05) greater in HFpEF (N = 141) with a mean of 6.7 ± 1.6 mm compared with a mean of 5.1 ± 1.0 mm in HFrEF (n = 40). After adjusting for the relationship with BMI, in HFpEF, epicardial fat was significantly (p < 0.05) negatively correlated with left ventricular internal diameter end diastole (LVIDd), left ventricular internal diameter end systole (LVIDs), left ventricular (LV) end-diastolic volume (EDV) index, lateral e', septal e', right atrial (RA) volume index, and hemoglobin (Hgb). The association with Hgb was no longer significant after adjusting for the effect of age. HFpEF was associated with smaller LVIDd, LVIDs, LV EDV indexes, and left atrial (LA) and RA volume indexes. Conclusions: Epicardial fat is significantly (p < 0.05) greater in HFpEF than HFrEF. Epicardial fat is associated with smaller cardiac chamber sizes in HFpEF suggesting that epicardial fat acts as a constraint to cardiac dilation.
Collapse
Affiliation(s)
- Gurwinder S Sidhu
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Simon W Rabkin
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Medicine, Division of Cardiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
23
|
Vest AR, Schauer PR, Rodgers JE, Sanderson E, LaChute CL, Seltz J, Lavie CJ, Mandras SA, Tang WHW, daSilva-deAbreu A. Obesity and Weight Loss Strategies for Patients With Heart Failure. JACC. HEART FAILURE 2024; 12:1509-1527. [PMID: 39093256 DOI: 10.1016/j.jchf.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 08/04/2024]
Abstract
Obesity is a common comorbidity among patients with heart failure with reduced ejection fraction (HFrEF) or heart failure with preserved ejection fraction (HFpEF), with the strongest pathophysiologic link of obesity being seen for HFpEF. Lifestyle measures are the cornerstone of weight loss management, but sustainability is a challenge, and there are limited efficacy data in the heart failure (HF) population. Bariatric surgery has moderate efficacy and safety data for patients with preoperative HF or left ventricular dysfunction and has been associated with reductions in HF hospitalizations and medium-term mortality. Antiobesity medications historically carried concerns for cardiovascular adverse effects, but the safety and weight loss efficacy seen in general population trials of glucagon-like peptide 1 (GLP-1) and gastric inhibitory polypeptide/GLP-1 agonists are highly encouraging. Although there are safety concerns regarding GLP-1 agonists in advanced HFrEF, trials of the GLP-1 agonist semaglutide for treatment of obesity have confirmed safety and efficacy in patients with HFpEF.
Collapse
Affiliation(s)
- Amanda R Vest
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Kaufman Center for Heart Failure Treatment and Recovery, Cleveland Clinic, Cleveland, Ohio, USA.
| | - Philip R Schauer
- Metamor Metabolic Institute, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Jo E Rodgers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Emily Sanderson
- Friedman School of Nutrition Science and Policy at Tufts University, Boston, Massachusetts, USA
| | - Courtney L LaChute
- Department of Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Jessica Seltz
- Frances Stern Nutrition Center, Tufts Medical Center, Boston, Massachusetts, USA
| | - Carl J Lavie
- Department of Cardiology, Ochsner Medical Center, New Orleans, Louisiana, USA; University of Queensland Ochsner Clinical School, University of Queensland, New Orleans, Louisiana, USA
| | - Stacy A Mandras
- Transplant Institute, AdventHealth Orlando, Orlando, Florida, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Kaufman Center for Heart Failure Treatment and Recovery, Cleveland Clinic, Cleveland, Ohio, USA.
| | - Adrian daSilva-deAbreu
- Doctoral School, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
24
|
Fu Z, Wang Y, Wang Y, Shi S, Li Y, Zhang B, Wu H, Song Q. Linking abnormal fat distribution with HFpEF and diastolic dysfunction: a systematic review, meta-analysis, and meta-regression of observational studies. Lipids Health Dis 2024; 23:277. [PMID: 39217346 PMCID: PMC11365188 DOI: 10.1186/s12944-024-02266-y] [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] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The global prevalence of obesity has escalated into a formidable health challenge intricately linked with the risk of developing cardiac diastolic disfunction and heart failure with preserved ejection fraction (HFpEF). Abnormal fat distribution is potentially strongly associated with an increased risk of cardiac diastolic dysfunction, and we aimed to scrutinize and elucidate the correlation between them. METHODS Following the Cochrane Handbook and PRISMA 2020 guidelines, we systematically reviewed the literature from PubMed, Embase, and Web of Science. We focused on studies reporting the mean and standard deviation (SD) of abnormal fat in HFpEF or cardiac diastolic dysfunction patients and the Pearson/Spearman correlation coefficients for the relationship between abnormal fat distribution and the risk of developing cardiac diastolic dysfunction. Data were standardized to the standard mean difference (SMD) and Fisher's z value for meta-analysis. RESULTS After progressive filtering and selection, 63 studies (43,113 participants) were included in the quantitative analyses. Abnormal fat distribution was significantly greater in participants with cardiac diastolic dysfunction than in controls [SMD 0.88 (0.69, 1.08)], especially in epicardial adipose tissue [SMD 0.99 (0.73, 1.25)]. Abnormal fat distribution was significantly correlated with the risk of developing cardiac diastolic dysfunction [E/E': 0.23 (0.18, 0.27), global longitudinal strain: r=-0.11 (-0.24, 0.02)]. Meta-regression revealed sample size as a potential heterogeneous source, and subgroup analyses revealed a stronger association between abnormal fat distribution and the risk of developing cardiac diastolic dysfunction in the overweight and obese population. CONCLUSION Abnormal fat distribution was significantly associated with the risk of developing cardiac diastolic dysfunction. TRIAL REGISTRATION CRD42024543774.
Collapse
Affiliation(s)
- Zhenyue Fu
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Yajiao Wang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxin Wang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuqing Shi
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yumeng Li
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huaqin Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingqiao Song
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
25
|
Schulz A, Backhaus SJ, Lange T, Evertz R, Kutty S, Kowallick JT, Hasenfuß G, Schuster A. Impact of epicardial adipose tissue on cardiac function and morphology in patients with diastolic dysfunction. ESC Heart Fail 2024; 11:2013-2022. [PMID: 38480481 PMCID: PMC11287361 DOI: 10.1002/ehf2.14744] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/19/2024] [Accepted: 02/11/2024] [Indexed: 07/31/2024] Open
Abstract
AIMS This study aimed to identify the impact of increased epicardial adipose tissue (EAT) and its regional distribution on cardiac function in patients with diastolic dysfunction. METHODS AND RESULTS Sixty-eight patients with exertional dyspnoea (New York Heart Association ≥II), preserved ejection fraction (≥50%), and diastolic dysfunction (E/e' ≥ 8) underwent rest and stress right heart catheterization, transthoracic echocardiography, and cardiovascular magnetic resonance (CMR). EAT volumes were depicted from CMR short-axis stacks. First, the impact of increased EAT above the median was investigated. Second, the association of ventricular and atrial EAT with myocardial deformation at rest and during exercise stress was analysed in a multivariable regression analysis. Patients with high EAT had higher HFA-PEFF and H2FPEFF scores as well as N-terminal prohormone of brain natriuretic peptide levels (all P < 0.048). They were diagnosed with manifest heart failure with preserved ejection fraction (HFpEF) more frequently (low EAT: 37% vs. high EAT: 64%; P = 0.029) and had signs of adverse remodelling indicated by higher T1 times (P < 0.001). No differences in biventricular volumetry and left ventricular mass (all P > 0.074) were observed. Patients with high EAT had impaired atrial strain at rest and during exercise stress, and impaired ventricular strain during exercise stress. Regionally increased EAT was independently associated with functional impairment of the adjacent chambers. CONCLUSIONS Patients with diastolic dysfunction and increased EAT show more pronounced signs of diastolic functional failure and adverse structural remodelling. Despite similar morphological characteristics, patients with high EAT show significant cardiac functional impairment, in particular in the atria. Our results indicate that regionally increased EAT directly induces atrial functional failure, which represents a distinct pathophysiological feature in HFpEF.
Collapse
Affiliation(s)
- Alexander Schulz
- Department of Cardiology and PneumologyUniversity Medical Center Göttingen, Georg August University of GöttingenGöttingenGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GöttingenGöttingenGermany
| | - Sören J. Backhaus
- Department of CardiologyCampus Kerckhoff of the Justus‐Liebig‐University Giessen, Kerckhoff‐ClinicBad NauheimGermany
| | - Torben Lange
- Department of Cardiology and PneumologyUniversity Medical Center Göttingen, Georg August University of GöttingenGöttingenGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GöttingenGöttingenGermany
| | - Ruben Evertz
- Department of Cardiology and PneumologyUniversity Medical Center Göttingen, Georg August University of GöttingenGöttingenGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GöttingenGöttingenGermany
| | - Shelby Kutty
- Taussig Heart CenterJohns Hopkins Hospital and School of MedicineBaltimoreMDUSA
| | - Johannes T. Kowallick
- German Centre for Cardiovascular Research (DZHK), Partner Site GöttingenGöttingenGermany
- Institute for Diagnostic and Interventional RadiologyUniversity Medical Center Göttingen, Georg August University of GöttingenGöttingenGermany
| | - Gerd Hasenfuß
- Department of Cardiology and PneumologyUniversity Medical Center Göttingen, Georg August University of GöttingenGöttingenGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GöttingenGöttingenGermany
| | - Andreas Schuster
- Department of Cardiology and PneumologyUniversity Medical Center Göttingen, Georg August University of GöttingenGöttingenGermany
- German Centre for Cardiovascular Research (DZHK), Partner Site GöttingenGöttingenGermany
| |
Collapse
|
26
|
Di Vincenzo A, Crescenzi M, Granzotto M, Vecchiato M, Fioretto P, Vettor R, Rossato M. Treatment with dapagliflozin increases FGF-21 gene expression and reduces triglycerides content in myocardial tissue of genetically obese mice. J Endocrinol Invest 2024; 47:1777-1786. [PMID: 38194168 DOI: 10.1007/s40618-023-02273-3] [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: 07/19/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND The association between obesity and some cardiovascular complications such as heart failure (HF) is well established, and drugs affecting adiposity are supposed to be promising treatments for these conditions. The sodium-glucose cotransporter-2 inhibitors (SGLT2i) are antidiabetic drugs showing benefits in patients with HF, despite the underlying mechanisms have not been completely understood yet. SGLT2i are supposed to promote systemic effects, such as triglycerides mobilization, through the enhancement of fibroblast growth factor-21 (FGF-21) activity. So, in this study, we evaluated the effects of dapagliflozin treatment on FGF-21 and related receptors (FGF-Rs) gene expression and on lipid content in myocardial tissue in an animal model of genetically induced obesity to unravel possible metabolic mechanisms accounting for the cardioprotection of SGLT2i. METHODS Six-week-old C57BL/6J wild-type mice and B6.V-LEP (ob/ob) mice were randomly assigned to the control or treatment group (14 animals/group). Treatment was based on the administration of dapagliflozin 0.15 mg/kg/day for 4 weeks. The gene expression of FGF-21 and related receptors (FGF-R1, FGF-R3, FGF-R4, and β-klotho co-receptor) was assessed at baseline and after treatment by real-time PCR. Similarly, cardiac triglycerides concentration was measured in the control group and treated animals. RESULTS At baseline, FGF-21 mRNA expression in the heart did not differ between lean and obese ob/ob mice. Dapagliflozin administration significantly increased heart FGF-21 gene expression, but only in ob/ob mice (p < 0.005). Consistently, when measuring the amount of triglycerides in the cardiac tissue, SGLT2i treatment reduced the lipid content in obese ob/ob mice, while no significant effects were observed in treated lean animals (p < 0.001). The overall expression of the FGF-21 receptors was only minimally affected by dapagliflozin treatment both in obese ob/ob mice and in lean controls. CONCLUSIONS Dapagliflozin administration increases FGF-21gene expression and reduces triglyceride content in myocardial tissue of ob/ob mice, while no significant effect was observed in lean controls. These results might help understand the cardiometabolic effects of SGLT2i inducing increased FGF-21 synthesis while reducing lipid content in cardiomyocytes as a possible expression of the switch to different energy substrates. This mechanism could represent a potential target of SGLT2i in obesity-related heart diseases.
Collapse
Affiliation(s)
- A Di Vincenzo
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy.
| | - M Crescenzi
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - M Granzotto
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - M Vecchiato
- Sports and Exercise Medicine Division, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - P Fioretto
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - R Vettor
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| | - M Rossato
- Internal Medicine 3, Department of Medicine, University-Hospital of Padova, Padua, Italy
| |
Collapse
|
27
|
Kasperova BJ, Mraz M, Svoboda P, Hlavacek D, Kratochvilova H, Modos I, Vrzackova N, Ivak P, Janovska P, Kobets T, Mahrik J, Riecan M, Steiner Mrazova L, Stranecky V, Netuka I, Cajka T, Kuda O, Melenovsky V, Stemberkova Hubackova S, Haluzik M. Sodium-glucose cotransporter 2 inhibitors induce anti-inflammatory and anti-ferroptotic shift in epicardial adipose tissue of subjects with severe heart failure. Cardiovasc Diabetol 2024; 23:223. [PMID: 38943140 PMCID: PMC11214218 DOI: 10.1186/s12933-024-02298-9] [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: 03/03/2024] [Accepted: 06/05/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Sodium-glucose cotransporter 2 inhibitors (SGLT-2i) are glucose-lowering agents used for the treatment of type 2 diabetes mellitus, which also improve heart failure and decrease the risk of cardiovascular complications. Epicardial adipose tissue (EAT) dysfunction was suggested to contribute to the development of heart failure. We aimed to elucidate a possible role of changes in EAT metabolic and inflammatory profile in the beneficial cardioprotective effects of SGLT-2i in subjects with severe heart failure. METHODS 26 subjects with severe heart failure, with reduced ejection fraction, treated with SGLT-2i versus 26 subjects without treatment, matched for age (54.0 ± 2.1 vs. 55.3 ± 2.1 years, n.s.), body mass index (27.8 ± 0.9 vs. 28.8 ± 1.0 kg/m2, n.s.) and left ventricular ejection fraction (20.7 ± 0.5 vs. 23.2 ± 1.7%, n.s.), who were scheduled for heart transplantation or mechanical support implantation, were included in the study. A complex metabolomic and gene expression analysis of EAT obtained during surgery was performed. RESULTS SGLT-2i ameliorated inflammation, as evidenced by the improved gene expression profile of pro-inflammatory genes in adipose tissue and decreased infiltration of immune cells into EAT. Enrichment of ether lipids with oleic acid noted on metabolomic analysis suggests a reduced disposition to ferroptosis, potentially further contributing to decreased oxidative stress in EAT of SGLT-2i treated subjects. CONCLUSIONS Our results show decreased inflammation in EAT of patients with severe heart failure treated by SGLT-2i, as compared to patients with heart failure without this therapy. Modulation of EAT inflammatory and metabolic status could represent a novel mechanism behind SGLT-2i-associated cardioprotective effects in patients with heart failure.
Collapse
Affiliation(s)
- Barbora Judita Kasperova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
- First Faculty of Medicine, Charles University in Prague, Katerinska 1660/32, 121 08, Prague, Czech Republic
| | - Milos Mraz
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 499/2, 128 08, Prague, Czech Republic
| | - Petr Svoboda
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technicka 5, 166 28, Prague, Czech Republic
| | - Daniel Hlavacek
- Department of Cardiac Surgery, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
- Third Faculty of Medicine, Charles University in Prague, Ruska 87, 100 00, Prague, Czech Republic
| | - Helena Kratochvilova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
| | - Istvan Modos
- Department of Informatics, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
| | - Nikola Vrzackova
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technicka 5, 166 28, Prague, Czech Republic
| | - Peter Ivak
- Department of Cardiac Surgery, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
- Third Faculty of Medicine, Charles University in Prague, Ruska 87, 100 00, Prague, Czech Republic
| | - Petra Janovska
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00, Prague, Czech Republic
| | - Tatyana Kobets
- Department of Metabolomics, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00, Prague, Czech Republic
| | - Jakub Mahrik
- First Faculty of Medicine, Charles University in Prague, Katerinska 1660/32, 121 08, Prague, Czech Republic
- Department of Cardiac Anesthesia, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
| | - Martin Riecan
- Department of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00, Prague, Czech Republic
| | - Lenka Steiner Mrazova
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00, Prague, Czech Republic
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, 128 08, Prague, Czech Republic
| | - Viktor Stranecky
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Ke Karlovu 455/2, 128 08, Prague, Czech Republic
| | - Ivan Netuka
- Department of Cardiac Surgery, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
| | - Tomas Cajka
- Department of Metabolomics, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00, Prague, Czech Republic
| | - Ondrej Kuda
- Department of Metabolism of Bioactive Lipids, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00, Prague, Czech Republic
| | - Vojtech Melenovsky
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic
| | - Sona Stemberkova Hubackova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic.
| | - Martin Haluzik
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21, Prague, Czech Republic.
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 499/2, 128 08, Prague, Czech Republic.
| |
Collapse
|
28
|
Abstract
Heart failure (HF) management guidelines recommend individualized assessments based on HF phenotypes. Adiposity is a known risk factor for HF. Recently, there has been an increased interest in organ-specific adiposity, specifically the role of the epicardial adipose tissue (EAT), in HF risk. EAT is easily assessable through various imaging modalities and is anatomically and functionally connected to the myocardium. In pathological conditions, EAT secretes inflammatory cytokines, releases excessive fatty acids, and increases mechanical load on the myocardium, resulting in myocardial remodeling. EAT plays a pathophysiological role in characterizing both HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). In HFrEF, EAT volume is reduced, reflecting an impaired metabolic reservoir, whereas in HFpEF, the amount of EAT is associated with worse biomarker and hemodynamic profiles, indicating increased EAT activity. Studies have examined the possibility of therapeutically targeting EAT, and recent studies using sodium glucose cotransporter 2 inhibitors have shown potential in reducing EAT volume. However, further research is required to determine the clinical implications of reducing EAT activity in patients with HF.
Collapse
Affiliation(s)
- Dong-Hyuk Cho
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Seong-Mi Park
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
29
|
Shao JW, Chen BH, Abu-Shaban K, Baiyasi A, Wu LM, Ma J. Epicardial adipose tissue in obesity with heart failure with preserved ejection fraction: Cardiovascular magnetic resonance biomarker study. World J Cardiol 2024; 16:149-160. [PMID: 38576524 PMCID: PMC10989227 DOI: 10.4330/wjc.v16.i3.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/09/2024] [Accepted: 02/06/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Obesity has become a serious public health issue, significantly elevating the risk of various complications. It is a well-established contributor to Heart failure with preserved ejection fraction (HFpEF). Evaluating HFpEF in obesity is crucial. Epicardial adipose tissue (EAT) has emerged as a valuable tool for validating prognostic biomarkers and guiding treatment targets. Hence, assessing EAT is of paramount importance. Cardiovascular magnetic resonance (CMR) imaging is acknowledged as the gold standard for analyzing cardiac function and morphology. We hope to use CMR to assess EAT as a bioimaging marker to evaluate HFpEF in obese patients. AIM To assess the diagnostic utility of CMR for evaluating heart failure with preserved ejection fraction [HFpEF; left ventricular (LV) ejection fraction ≥ 50%] by measuring the epicardial adipose tissue (EAT) volumes and EAT mass in obese patients. METHODS Sixty-two obese patients were divided into two groups for a case-control study based on whether or not they had heart failure with HFpEF. The two groups were defined as HFpEF+ and HFpEF-. LV geometry, global systolic function, EAT volumes and EAT mass of all subjects were obtained using cine magnetic resonance sequences. RESULTS Forty-five patients of HFpEF- group and seventeen patients of HFpEF+ group were included. LV mass index (g/m2) of HFpEF+ group was higher than HFpEF- group (P < 0.05). In HFpEF+ group, EAT volumes, EAT volume index, EAT mass, EAT mass index and the ratio of EAT/[left atrial (LA) left-right (LR) diameter] were higher compared to HFpEF- group (P < 0.05). In multivariate analysis, Higher EAT/LA LR diameter ratio was associated with higher odds ratio of HFpEF. CONCLUSION EAT/LA LR diameter ratio is highly associated with HFpEF in obese patients. It is plausible that there may be utility in CMR for assessing obese patients for HFpEF using EAT/LA LR diameter ratio as a diagnostic biomarker. Further prospective studies, are needed to validate these proof-of-concept findings.
Collapse
Affiliation(s)
- Ju-Wei Shao
- Department of Radiology, The Affiliated Hospital of Yunnan University, Kunming 650021, Yunnan Province, China
| | - Bing-Hua Chen
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Kamil Abu-Shaban
- Department of Radiology, University of Toledo College of Medicine, Toledo, OH 43623, United States
| | - Ahmad Baiyasi
- Department of Radiology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| | - Lian-Ming Wu
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jing Ma
- Department of Endocrinology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
30
|
O’Sullivan JF, Li M, Koay YC, Wang XS, Guglielmi G, Marques FZ, Nanayakkara S, Mariani J, Slaughter E, Kaye DM. Cardiac Substrate Utilization and Relationship to Invasive Exercise Hemodynamic Parameters in HFpEF. JACC Basic Transl Sci 2024; 9:281-299. [PMID: 38559626 PMCID: PMC10978404 DOI: 10.1016/j.jacbts.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 04/04/2024]
Abstract
The authors conducted transcardiac blood sampling in healthy subjects and subjects with heart failure with preserved ejection fraction (HFpEF) to compare cardiac metabolite and lipid substrate use. We demonstrate that fatty acids are less used by HFpEF hearts and that lipid extraction is influenced by hemodynamic factors including pulmonary pressures and cardiac index. The release of many products of protein catabolism is apparent in HFpEF compared to healthy myocardium. In subgroup analyses, differences in energy substrate use between female and male hearts were identified.
Collapse
Affiliation(s)
- John F. O’Sullivan
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia
- Department of Medicine, TU Dresden, Dresden, Germany
| | - Mengbo Li
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Yen Chin Koay
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, Australia
| | - Xiao Suo Wang
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Giovanni Guglielmi
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Australia
- School of Mathematics, University of Birmingham, Birmingham, United Kingdom
| | - Francine Z. Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
| | - Shane Nanayakkara
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- Monash-Alfred-Baker Centre for Cardiovascular Research, Monash University, Melbourne, Australia
| | - Justin Mariani
- Victorian Heart Institute, Monash University, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- Monash-Alfred-Baker Centre for Cardiovascular Research, Monash University, Melbourne, Australia
| | - Eugene Slaughter
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - David M. Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- Monash-Alfred-Baker Centre for Cardiovascular Research, Monash University, Melbourne, Australia
| |
Collapse
|
31
|
Henry JA, Couch LS, Rider OJ. Myocardial Metabolism in Heart Failure with Preserved Ejection Fraction. J Clin Med 2024; 13:1195. [PMID: 38592048 PMCID: PMC10931709 DOI: 10.3390/jcm13051195] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/11/2024] [Accepted: 02/18/2024] [Indexed: 04/10/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasingly prevalent and now accounts for half of all heart failure cases. This rise is largely attributed to growing rates of obesity, hypertension, and diabetes. Despite its prevalence, the pathophysiological mechanisms of HFpEF are not fully understood. The heart, being the most energy-demanding organ, appears to have a compromised bioenergetic capacity in heart failure, affecting all phenotypes and aetiologies. While metabolic disturbances in heart failure with reduced ejection fraction (HFrEF) have been extensively studied, similar insights into HFpEF are limited. This review collates evidence from both animal and human studies, highlighting metabolic dysregulations associated with HFpEF and its risk factors, such as obesity, hypertension, and diabetes. We discuss how changes in substrate utilisation, oxidative phosphorylation, and energy transport contribute to HFpEF. By delving into these pathological shifts in myocardial energy production, we aim to reveal novel therapeutic opportunities. Potential strategies include modulating energy substrates, improving metabolic efficiency, and enhancing critical metabolic pathways. Understanding these aspects could be key to developing more effective treatments for HFpEF.
Collapse
Affiliation(s)
- John Aaron Henry
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK (O.J.R.)
- Department of Cardiology, Jersey General Hospital, Gloucester Street, St. Helier JE1 3QS, Jersey, UK
| | - Liam S. Couch
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK (O.J.R.)
| | - Oliver J. Rider
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK (O.J.R.)
| |
Collapse
|
32
|
Hao Y, Zhang R, Chen L, Fan G, Liu B, Jiang K, Zhu Y, Zhang M, Guo J. Distinguishing heart failure subtypes: the diagnostic power of different cardiac magnetic resonance imaging parameters. Front Cardiovasc Med 2024; 11:1291735. [PMID: 38385138 PMCID: PMC10879269 DOI: 10.3389/fcvm.2024.1291735] [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: 09/10/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
Objectives The aim of this retrospective study was to explore the diagnostic potential of various cardiac parameters in differentiating between heart failure with preserved ejection fraction (HFpEF) and heart failure with mid-ranged and reduced ejection fraction (HFm + rEF), and to discern their relationship with normal cardiac function. Methods This research encompassed a comparative analysis of heart failure subtypes based on multiple indicators. Participants were categorized into HFm + rEF, HFpEF, and control groups. For each participant, we investigated indicators of left ventricular function (LVEDVi, LVESVi, and LVEF) and myocardial strain parameters (GLS, GCS, GRS). Additionally, quantitative tissue evaluation parameters including native T1, enhanced T1, and extracellular volume (ECV) were examined.For comprehensive diagnostic performance analysis, receiver operating characteristic (ROC) curve evaluations for each parameters were conducted. Results HFm + rEF patients exhibited elevated LVEDVi and LVESVi and decreased LVEF compared to both HFpEF and control groups. Myocardial strain revealed significant reductions in GLS, GCS, and GRS for HFm + rEF patients compared to the other groups. HFpEF patients showed strain reductions relative to the control group. In cardiac magnetic resonance imaging (CMR) evaluations, HFm + rEF patients demonstrated heightened native T1 times and ECV fractions. Native T1 was particularly effective in distinguishing HFpEF from healthy subjects. Conclusion Native T1, ECV, and myocardial strain parameters have substantial diagnostic value in identifying HFpEF. Among them, native T1 displayed superior diagnostic efficiency relative to ECV, offering critical insights into early-stage HFpEF. These findings can play a pivotal role in refining clinical management and treatment strategies for heart failure patients.
Collapse
Affiliation(s)
- Yanhui Hao
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rui Zhang
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lihong Chen
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ganglian Fan
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bing Liu
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ke Jiang
- Clinical & Technical Support, Philips Healthcare, Beijing, China
| | - Yi Zhu
- Clinical & Technical Support, Philips Healthcare, Beijing, China
| | - Ming Zhang
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jianxin Guo
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
33
|
Załęska-Kocięcka M, Wojdyńska Z, Kalisz M, Litwiniuk A, Mączewski M, Leszek P, Paterek A. Epicardial fat and ventricular arrhythmias. Heart Rhythm 2024; 21:206-212. [PMID: 37972673 DOI: 10.1016/j.hrthm.2023.11.008] [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: 09/17/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
The arrhythmogenic role of epicardial adipose tissue (EAT) in atrial arrhythmias is well established, but its effect on ventricular arrhythmias has been significantly less investigated. Since ventricular arrhythmias are thought to cause 75%-80% of cases of sudden cardiac death, this is not a trivial issue. We provide an overview of clinical data as well as experimental and molecular data linking EAT to ventricular arrhythmias, attempting to dissect possible mechanisms and indicate future directions of research and possible clinical implications. However, despite a wealth of data indicating the role of epicardial and intramyocardial fat in the induction and propagation of ventricular arrhythmias, unfortunately there is currently no direct evidence that indeed EAT triggers arrhythmia or can be a target for antiarrhythmic strategies.
Collapse
Affiliation(s)
- Marta Załęska-Kocięcka
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Zuzanna Wojdyńska
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Małgorzata Kalisz
- Department of Clinical Neuroendocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Anna Litwiniuk
- Department of Clinical Neuroendocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Przemysław Leszek
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
34
|
Oneglia AP, Szczepaniak LS, Zaha VG, Nelson MD. Myocardial steatosis across the spectrum of human health and disease. Exp Physiol 2024; 109:202-213. [PMID: 38063136 PMCID: PMC10841709 DOI: 10.1113/ep091566] [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: 10/10/2023] [Accepted: 11/15/2023] [Indexed: 02/02/2024]
Abstract
Preclinical data strongly suggest that myocardial steatosis leads to adverse cardiac remodelling and left ventricular dysfunction. Using 1 H cardiac magnetic resonance spectroscopy, similar observations have been made across the spectrum of health and disease. The purpose of this brief review is to summarize these recent observations. We provide a brief overview of the determinants of myocardial triglyceride accumulation, summarize the current evidence that myocardial steatosis contributes to cardiac dysfunction, and identify opportunities for further research.
Collapse
Affiliation(s)
- Andrew P. Oneglia
- Applied Physiology and Advanced Imaging Laboratory, Department of Kinesiology, College of Nursing and Health InnovationUniversity of Texas at ArlingtonArlingtonTexasUSA
| | | | - Vlad G. Zaha
- Division of Cardiology, Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Advanced Imaging Research CenterUniversity of Texas Southwestern Medical CenterArlingtonTexasUSA
| | - Michael D. Nelson
- Applied Physiology and Advanced Imaging Laboratory, Department of Kinesiology, College of Nursing and Health InnovationUniversity of Texas at ArlingtonArlingtonTexasUSA
- Clinical Imaging Research CenterUniversity of Texas at ArlingtonArlingtonTexasUSA
- Center for Healthy Living and LongevityUniversity of Texas at ArlingtonArlingtonTexasUSA
| |
Collapse
|
35
|
Gao S, Liu XP, Li TT, Chen L, Feng YP, Wang YK, Yin YJ, Little PJ, Wu XQ, Xu SW, Jiang XD. Animal models of heart failure with preserved ejection fraction (HFpEF): from metabolic pathobiology to drug discovery. Acta Pharmacol Sin 2024; 45:23-35. [PMID: 37644131 PMCID: PMC10770177 DOI: 10.1038/s41401-023-01152-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is currently a preeminent challenge for cardiovascular medicine. It has a poor prognosis, increasing mortality, and is escalating in prevalence worldwide. Despite accounting for over 50% of all HF patients, the mechanistic underpinnings driving HFpEF are poorly understood, thus impeding the discovery and development of mechanism-based therapies. HFpEF is a disease syndrome driven by diverse comorbidities, including hypertension, diabetes and obesity, pulmonary hypertension, aging, and atrial fibrillation. There is a lack of high-fidelity animal models that faithfully recapitulate the HFpEF phenotype, owing primarily to the disease heterogeneity, which has hampered our understanding of the complex pathophysiology of HFpEF. This review provides an updated overview of the currently available animal models of HFpEF and discusses their characteristics from the perspective of energy metabolism. Interventional strategies for efficiently utilizing energy substrates in preclinical HFpEF models are also discussed.
Collapse
Affiliation(s)
- Si Gao
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Xue-Ping Liu
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Ting-Ting Li
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Li Chen
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yi-Ping Feng
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yu-Kun Wang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yan-Jun Yin
- School of Pharmacy, Bengbu Medical College, Bengbu, 233000, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, 4102, Australia
| | - Xiao-Qian Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Suo-Wen Xu
- Department of Endocrinology, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Xu-Dong Jiang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China.
| |
Collapse
|
36
|
Rossi VA, Nebunu D, Haider T, Laptseva N, Naegele MP, Ruschitzka F, Sudano I, Flammer AJ. Diverging role of epicardial adipose tissue across the entire heart failure spectrum. ESC Heart Fail 2023; 10:3419-3429. [PMID: 37697706 PMCID: PMC10682858 DOI: 10.1002/ehf2.14483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/06/2023] [Indexed: 09/13/2023] Open
Abstract
AIMS Epicardial adipose tissue (EAT) is a metabolically highly active tissue modulating numerous pathophysiological processes. The aim of this study was to investigate the association between EAT thickness and endothelial function in patients with heart failure (HF) across the entire ejection fraction spectrum. METHODS AND RESULTS A total of 258 patients with HF with an ejection fraction across the entire spectrum [HF with reduced ejection fraction (HFrEF), n = 168, age 60.6 ± 11.2 years; HF with preserved ejection fraction (HFpEF), n = 50, mean age 65.1 ± 11.9 years; HF with mildly reduced ejection fraction (HFmrEF), n = 32, mean age 65 ± 12] were included. EAT was measured with transthoracic echocardiography. Vascular function was assessed with flicker-light-induced vasodilation of retinal arterioles (FIDart%) and flow-mediated dilatation (FMD%) in conduit arteries. Patients with HFrEF have less EAT compared with patients with HFpEF (4.2 ± 2 vs. 5.3 ± 2 mm, respectively, P < 0.001). Interestingly, EAT was significantly associated with impaired microvascular function (FIDart%; r = -0.213, P = 0.012) and FMD% (r = -0.186, P = 0.022), even after multivariate correction for confounding factors (age, body mass index, hypertension, and diabetes; standardized regression coefficient (SRC) = -0.184, P = 0.049 for FIDart% and SRC = -0.178, P = 0.043 for FMD%) in HFrEF but not in HFpEF. CONCLUSIONS Although less EAT is present in HFrEF than in HFpEF, only in HFrEF EAT is associated with vascular dysfunction. The diverging role of EAT in HF and its switch to a functionally deleterious tissue promoting HF progression provide the rationale to specifically target EAT, in particular in patients with reduced ejection fraction.
Collapse
Affiliation(s)
- Valentina A. Rossi
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
| | - Delia Nebunu
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
| | - Thomas Haider
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
| | - Natallia Laptseva
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
| | - Matthias P. Naegele
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
| | - Frank Ruschitzka
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
- University of ZurichZurichSwitzerland
| | - Isabella Sudano
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
| | - Andreas J. Flammer
- Department of CardiologyUniversity Heart Centre, University Hospital of ZurichRaemistrasse 100Zurich8091Switzerland
- Centre for Translational and Experimental CardiologySchlierenSwitzerland
- University of ZurichZurichSwitzerland
| |
Collapse
|
37
|
Wu A, Yang Z, Zhang X, Lin Z, Lu H. Association Between Epicardial Adipose Tissue and Left Atrial and Ventricular Function in Patients With Heart Failure: A Systematic Review and Meta-Analysis. Curr Probl Cardiol 2023; 48:101979. [PMID: 37481217 DOI: 10.1016/j.cpcardiol.2023.101979] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Existing evidence suggested that the role of epicardial adipose tissue (EAT) in heart failure with reduced and preserved ejection fraction (HFrEF/HFpEF) might be divergent. Here, we conducted a systematic review and meta-analysis to evaluate the association between EAT and HF. Several databases were searched from their inception to January 20, 2023. We calculated the standard mean difference (SMD) in EAT between the HF and control groups, as well as the correlation coefficient between EAT and left atrial (LA) and left ventricular (LV) function. This meta-analysis included 23 studies, involving 1563 HFrEF and 1351 HFpEF patients. Our findings indicated that EAT was significantly higher in HFpEF patients (SMD: 0.61, 95% CI: 0.27-0.94), but not in total HF or HFrEF patients compared to controls. In HFrEF, EAT was positively correlated with LVEF, LV end-diastolic volume index (LVEDVI), LA global longitudinal strain (LAGLS), and negatively correlated with N-terminal pro-B-type natriuretic peptide (NT-ProBNP). However, no significant relationship existed between EAT and LV mass index (LVMI) or LVGLS. For HFpEF, EAT correlated positively with LVMI, LVEDVI, LV end-systolic volume index (LVESVI), LA volume index (LAVI), cardiac troponin T, and extracellular volume (ECV), but negatively with LVGLS and LAGLS. EAT was shown to be higher in HFpEF, but not in HFrEF. Less EAT was linked with worse LA function but not worse LV function in HFrEF, while more EAT was associated with worse LA/LV function in HFpEF.
Collapse
Affiliation(s)
- Anhu Wu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhuohao Yang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xinyu Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zongwei Lin
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Huixia Lu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
38
|
Banerjee M. Epicardial Fat Paradox and Differential Effects of GLP-1 Receptor Agonists Across Heart Failure Phenotypes. Circ Heart Fail 2023; 16:e010966. [PMID: 38010208 DOI: 10.1161/circheartfailure.123.010966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Affiliation(s)
- Mainak Banerjee
- Department of Endocrinology, Ramakrishna Mission Seva Pratishthan Vivekandanda Institute of Medical Sciences, Kolkata, India (M.B.)
- Narayana Health, Rabindranath Tagore International Institute of Cardiac Sciences, Mukundapur, Kolkata, India (M.B.)
| |
Collapse
|
39
|
Goldman SA, Requena-Ibanez JA, Devesa A, Santos-Gallego CG, Badimon JJ, Fuster V. Uncovering the Role of Epicardial Adipose Tissue in Heart Failure With Preserved Ejection Fraction. JACC. ADVANCES 2023; 2:100657. [PMID: 38938732 PMCID: PMC11198699 DOI: 10.1016/j.jacadv.2023.100657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 06/29/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is the most common form of heart failure. Obesity is a modifiable risk factor of HFpEF; however, body mass index provides limited information on visceral adiposity and patients with similar anthropometrics can present variable cardiovascular risk. Epicardial adipose tissue (EAT) is the closest fat deposit to the heart and has been proposed as a biomarker of visceral adiposity. EAT may be particularly important for cardiac function, because of its location (under the pericardium) and because it acts as a metabolically active endocrine organ (which can produce both beneficial and detrimental cytokines). In this paper, the authors review the role of EAT in normal and pathologic conditions and discuss the noninvasive imaging modalities that allow its identification. This review highlights EAT implications in HFpEF and discuss new therapies that act on EAT and might also exert beneficial effects on the cardiovascular system.
Collapse
Affiliation(s)
- Sarah A. Goldman
- Department of Internal Medicine, Zucker School of Medicine at Hofstra Northwell, Lenox Hill Hospital New York, New York, New York, USA
| | - Juan Antonio Requena-Ibanez
- Atherothrombosis Research Unit, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai School of Medicine, New York, New York, USA
| | - Ana Devesa
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- BioMedical Engineering and Imaging Institute (BMEII), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Carlos G. Santos-Gallego
- Atherothrombosis Research Unit, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai School of Medicine, New York, New York, USA
| | - Juan José Badimon
- Atherothrombosis Research Unit, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai School of Medicine, New York, New York, USA
| | - Valentin Fuster
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| |
Collapse
|
40
|
Zoico E, Giani A, Saatchi T, Rizzatti V, Mazzali G, Fantin F, Benfari G, Onorati F, Urbani S, Zamboni M. Myocardial Fibrosis and Steatosis in Patients with Aortic Stenosis: Roles of Myostatin and Ceramides. Int J Mol Sci 2023; 24:15508. [PMID: 37958492 PMCID: PMC10648018 DOI: 10.3390/ijms242115508] [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: 09/07/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Aortic stenosis (AS) involves progressive valve obstruction and a remodeling response of the left ventriculum (LV) with systolic and diastolic dysfunction. The roles of interstitial fibrosis and myocardial steatosis in LV dysfunction in AS have not been completely characterized. We enrolled 31 patients (19 women and 12 men) with severe AS undergoing elective aortic valve replacement. The subjects were clinically evaluated, and transthoracic echocardiography was performed pre-surgery. LV septal biopsies were obtained to assess fibrosis and apoptosis and fat deposition in myocytes (perilipin 5 (PLIN5)), or in the form of adipocytes within the heart (perilipin 1 (PLIN1)), the presence of ceramides and myostatin were assessed via immunohistochemistry. After BMI adjustment, we found a positive association between fibrosis and apoptotic cardiomyocytes, as well as fibrosis and the area covered by PLIN5. Apoptosis and PLIN5 were also significantly interrelated. LV fibrosis increased with a higher medium gradient (MG) and peak gradient (PG). Ceramides and myostatin levels were higher in patients within the higher MG and PG tertiles. In the linear regression analysis, increased fibrosis correlated with increased apoptosis and myostatin, independent from confounding factors. After adjustment for age and BMI, we found a positive relationship between PLIN5 and E/A and a negative correlation between septal S', global longitudinal strain (GLS), and fibrosis. Myostatin was inversely correlated with GLS and ejection fraction. Fibrosis and myocardial steatosis altogether contribute to ventricular dysfunction in severe AS. The association of myostatin and fibrosis with systolic dysfunction, as well as between myocardial steatosis and diastolic dysfunction, highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Elena Zoico
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Anna Giani
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Tanaz Saatchi
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Vanni Rizzatti
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Gloria Mazzali
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Francesco Fantin
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Giovanni Benfari
- Division of Cardiology, Department of Medicine, University of Verona, 37126 Verona, Italy
| | - Francesco Onorati
- Division of Cardiac Surgery, Department of Surgery, Dentistry, Pediatric and Gynecology, University of Verona, 37126 Verona, Italy
| | - Silvia Urbani
- Division of Geriatric Medicine, Department of Medicine, University of Verona, 37126 Verona, Italy; (A.G.)
| | - Mauro Zamboni
- Division of Geriatric Medicine, Department of Surgery, Dentistry, Pediatric and Gynecology, University of Verona, 37126 Verona, Italy
| |
Collapse
|
41
|
Lin T, Lee C, Huang K, Wu C, Lee J, Lan C, Su MM, Hwang J, Wang Y, Lin L. Differentiating the Prognostic Determinants of Myocardial Steatosis for Heart Failure With Preserved Ejection Fraction by Cardiac Magnetic Resonance Imaging. J Am Heart Assoc 2023; 12:e027781. [PMID: 37642018 PMCID: PMC10547328 DOI: 10.1161/jaha.122.027781] [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: 08/10/2022] [Accepted: 07/10/2023] [Indexed: 08/31/2023]
Abstract
Background Myocardial steatosis and fibrosis may play a role in the pathophysiology of heart failure with preserved ejection fraction. We therefore investigated the prognostic significance of epicardial fat (epicardial adipose tissue [EAT]) and myocardial diffuse fibrosis. Methods and Results Myocardial fibrosis, estimated as extracellular volume (ECV), and EAT were measured using cardiac magnetic resonance imaging in 163 subjects with heart failure with preserved ejection fraction. We also evaluated cardiac structure and diastolic and systolic function by echocardiography and cardiac magnetic resonance imaging. After 24 months' follow-up, 39 (24%) subjects had experienced cardiovascular events, including hospitalization for heart failure, acute coronary syndrome, and cardiovascular death. Median EAT and mean ECV were significantly higher in subjects with cardiovascular events than survivors (EAT, 35 [25-45] versus 31 [21-38], P=0.006 and ECV, 28.9±3.16% versus 27.2±3.56%, P=0.04). Subjects with high EAT (≥42 g) had increased risk of cardiovascular events (hazard ratio [HR], 2.528 [95% CI, 1.704-4.981]; P=0.032). High ECV (>29%) was also significantly associated with poorer outcomes (HR, 1.647 [95% CI, 1.263-2.548]; P=0.013). With respect to secondary end points, high EAT and high ECV were associated with increased risk of the incident acute coronary syndrome (HR, 1.982 [95% CI, 1.008-4.123]; P=0.049) and hospitalization for heart failure (HR, 1.789 [95% CI, 1.102-6.987]; P=0.033), respectively. Conclusions Our study suggested that increased epicardial fat and ECV detected by cardiac magnetic resonance imaging have an impact on cardiovascular prognosis, in particular acute coronary syndrome and hospitalization for heart failure, respectively.
Collapse
Affiliation(s)
- Ting‐Tse Lin
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Chih‐Kuo Lee
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University Hospital Hsin‐Chu BranchHsinchuTaiwan
| | - Kuan‐Chih Huang
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University Hospital Hsin‐Chu BranchHsinchuTaiwan
| | - Cho‐Kai Wu
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Jen‐Kuang Lee
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Chen‐Wei Lan
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Mao‐Yuan M. Su
- Department of Medical ImagingNational Taiwan University HospitalTaipeiTaiwan
| | - Juey‐Jen Hwang
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Yi‐Chih Wang
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| | - Lian‐Yu Lin
- Department of Internal Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Cardiology, Department of Internal MedicineNational Taiwan University College of Medicine and HospitalTaipeiTaiwan
| |
Collapse
|
42
|
Wang X, Butcher SC, Myagmardorj R, Liem SIE, Delgado V, Bax JJ, De Vries-Bouwstra JK, Marsan NA. Epicardial adipose tissue in patients with systemic sclerosis. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2023; 1:qyad037. [PMID: 39045073 PMCID: PMC11195713 DOI: 10.1093/ehjimp/qyad037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/11/2023] [Indexed: 07/25/2024]
Abstract
Aims Epicardial adipose tissue (EAT) has emerged as a mediator between systemic inflammatory disorders and cardiovascular disease, and may therefore play a role in the pathophysiology of cardiac involvement in systemic sclerosis (SSc). The aim of this study was to assess the correlation between EAT and left ventricular (LV) function, and to determine the prognostic value of EAT in patients with SSc. Methods and results Consecutive patients with SSc who underwent non-contrast thorax computed tomography and echocardiography were included. EAT mass was quantified using dedicated software. The study endpoint was all-cause mortality. A total of 230 SSc patients (age 53 ± 15 years, 14% male) were included. The median value of EAT mass was 67 g (interquartile range: 45-101 g). Patients with increased EAT mass (≥67 g) showed more impaired LV diastolic function as compared with patients with less EAT mass (<67 g), and even after adjusting for age and comorbidities, EAT mass was independently associated with LV diastolic function parameters. During a median follow-up of 8 years, 42 deaths occurred. Kaplan-Meier analysis showed that patients with increased EAT mass had higher all-cause mortality rate as compared with patients with less EAT mass (29% vs. 7%; P < 0.001). In the multivariable analysis, EAT was independently associated with all-cause mortality after adjusting for important covariates (HR: 1.006; 95% CI: 1.001-1.010). Conclusion In patients with SSc, EAT is independently associated with LV diastolic dysfunction and higher mortality rate.
Collapse
Affiliation(s)
- Xu Wang
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, XCF3+6R6, Chaoyang, Beijing 100029, China
| | - Steele C Butcher
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
- Department of Cardiology, Royal Perth Hospital, Victoria Square, Perth WA 6000, Western Australia, Australia
| | - Rinchyenkhand Myagmardorj
- Department of Cardiology, Mongolia-Japan Teaching Hospital, Mongolian National University of Medical Sciences, Botanic street, Ulaanbaatar 13270, Mongolia
| | - Sophie I E Liem
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Victoria Delgado
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
- Hospital University Germans Trias i Pujol, Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Carretera de Canyet, s/n, Badalona 08916, Spain
| | - Jeroen J Bax
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
- Heart Center, University of Turku and Turku University Hospital, U-sairaala, Kiinamyllynkatu 4-8, Turku 20521, Finland
| | - Jeska K De Vries-Bouwstra
- Department of Cardiology, Mongolia-Japan Teaching Hospital, Mongolian National University of Medical Sciences, Botanic street, Ulaanbaatar 13270, Mongolia
| | - Nina Ajmone Marsan
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| |
Collapse
|
43
|
Fan L, Meng C, Wang X, Wang Y, Li Y, Lv S, Zhang J. Driving force of deteriorated cellular environment in heart failure: Metabolic remodeling. Clinics (Sao Paulo) 2023; 78:100263. [PMID: 37557005 PMCID: PMC10432917 DOI: 10.1016/j.clinsp.2023.100263] [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: 01/07/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023] Open
Abstract
Heart Failure (HF) has been one of the leading causes of death worldwide. Though its latent mechanism and therapeutic manipulation are updated and developed ceaselessly, there remain great gaps in the cognition of heart failure. High morbidity and readmission rates among HF patients are waiting to be addressed. Recent studies have found that myocardial energy metabolism was closely related to heart failure, in which substrate utilization, as well as intermediate metabolism disorders, insulin resistance, oxidative stress, and mitochondrial dysfunction, might underlie systolic dysfunction and progression of HF. This article centers on the changes and counteraction of cardiac energy metabolism in the failing heart. Therefore, targeting impaired energy provision is of great potential in the treatment of HF. And shifting the objective from traditional neurohormones to improving the cellular environment is expected to further optimize the management of HF.
Collapse
Affiliation(s)
- Lu Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chenchen Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaoming Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunjiao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanyang Li
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin, China.
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
44
|
Hearon CM, Reddy S, Dias KA, Shankar A, MacNamara J, Levine B, Sarma S. Characterizing regional and global effects of epicardial adipose tissue on cardiac systolic and diastolic function. Obesity (Silver Spring) 2023; 31:1884-1893. [PMID: 37368514 DOI: 10.1002/oby.23782] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVE The aim of this retrospective study was to determine whether regional epicardial adipose tissue (EAT) exerts localized effects on adjacent myocardial left ventricular (LV) function. METHODS Cardiac magnetic resonance imaging (MRI), echocardiography, dual-energy x-ray absorptiometry, and exercise testing were performed in 71 patients with obesity with elevated cardiac biomarkers and visceral fat. Total and regional (anterior, inferior, lateral, right ventricular) EAT was quantified by MRI. Diastolic function was quantified by echocardiography. MRI was used to quantify regional longitudinal LV strain. RESULTS EAT was associated with visceral adiposity (r = 0.47, p < 0.0001) but not total fat mass. Total EAT was associated with markers of diastolic function (early tissue Doppler relaxation velocity [e'], mitral inflow velocity ratio [E/A], early mitral inflow/e' ratio [E/e']), but only E/A remained significant after adjustment for visceral adiposity (r = -0.30, p = 0.015). Right ventricular and LV EAT had similar associations with diastolic function. There was no evidence for localized effects of regional EAT deposition on adjacent regional longitudinal strain. CONCLUSIONS There was no association between regional EAT deposition and corresponding regional LV segment function. Furthermore, the association between total EAT and diastolic function was attenuated after adjustment for visceral fat, indicating that systemic metabolic impairments contribute to diastolic dysfunction in high-risk middle-aged adults.
Collapse
Affiliation(s)
- Christopher M Hearon
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Shiva Reddy
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Katrin A Dias
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Aditi Shankar
- Department of Internal Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
| | - James MacNamara
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Benjamin Levine
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Satyam Sarma
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Dallas, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
45
|
Hundertmark MJ, Adler A, Antoniades C, Coleman R, Griffin JL, Holman RR, Lamlum H, Lee J, Massey D, Miller JJ, Milton JE, Monga S, Mózes FE, Nazeer A, Raman B, Rider O, Rodgers CT, Valkovič L, Wicks E, Mahmod M, Neubauer S. Assessment of Cardiac Energy Metabolism, Function, and Physiology in Patients With Heart Failure Taking Empagliflozin: The Randomized, Controlled EMPA-VISION Trial. Circulation 2023; 147:1654-1669. [PMID: 37070436 PMCID: PMC10212585 DOI: 10.1161/circulationaha.122.062021] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/16/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Sodium-glucose co-transporter 2 inhibitors (SGLT2i) have emerged as a paramount treatment for patients with heart failure (HF), irrespective of underlying reduced or preserved ejection fraction. However, a definite cardiac mechanism of action remains elusive. Derangements in myocardial energy metabolism are detectable in all HF phenotypes, and it was proposed that SGLT2i may improve energy production. The authors aimed to investigate whether treatment with empagliflozin leads to changes in myocardial energetics, serum metabolomics, and cardiorespiratory fitness. METHODS EMPA-VISION (Assessment of Cardiac Energy Metabolism, Function and Physiology in Patients With Heart Failure Taking Empagliflozin) is a prospective, randomized, double-blind, placebo-controlled, mechanistic trial that enrolled 72 symptomatic patients with chronic HF with reduced ejection fraction (HFrEF; n=36; left ventricular ejection fraction ≤40%; New York Heart Association class ≥II; NT-proBNP [N-terminal pro-B-type natriuretic peptide] ≥125 pg/mL) and HF with preserved ejection fraction (HFpEF; n=36; left ventricular ejection fraction ≥50%; New York Heart Association class ≥II; NT-proBNP ≥125 pg/mL). Patients were stratified into respective cohorts (HFrEF versus HFpEF) and randomly assigned to empagliflozin (10 mg; n=35: 17 HFrEF and 18 HFpEF) or placebo (n=37: 19 HFrEF and 18 HFpEF) once daily for 12 weeks. The primary end point was a change in the cardiac phosphocreatine:ATP ratio (PCr/ATP) from baseline to week 12, determined by phosphorus magnetic resonance spectroscopy at rest and during peak dobutamine stress (65% of age-maximum heart rate). Mass spectrometry on a targeted set of 19 metabolites was performed at baseline and after treatment. Other exploratory end points were investigated. RESULTS Empagliflozin treatment did not change cardiac energetics (ie, PCr/ATP) at rest in HFrEF (adjusted mean treatment difference [empagliflozin - placebo], -0.25 [95% CI, -0.58 to 0.09]; P=0.14) or HFpEF (adjusted mean treatment difference, -0.16 [95% CI, -0.60 to 0.29]; P=0.47]. Likewise, there were no changes in PCr/ATP during dobutamine stress in HFrEF (adjusted mean treatment difference, -0.13 [95% CI, -0.35 to 0.09]; P=0.23) or HFpEF (adjusted mean treatment difference, -0.22 [95% CI, -0.66 to 0.23]; P=0.32). No changes in serum metabolomics or levels of circulating ketone bodies were observed. CONCLUSIONS In patients with either HFrEF or HFpEF, treatment with 10 mg of empagliflozin once daily for 12 weeks did not improve cardiac energetics or change circulating serum metabolites associated with energy metabolism when compared with placebo. Based on our results, it is unlikely that enhancing cardiac energy metabolism mediates the beneficial effects of SGLT2i in HF. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03332212.
Collapse
Affiliation(s)
- Moritz J. Hundertmark
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
- Department of Internal Medicine I, University Hospital Wuerzburg, Germany (M.J.H.)
| | - Amanda Adler
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (A.A., R.C., R.R.H., J.E.M.), University of Oxford, UK
| | - Charalambos Antoniades
- Acute Multidisciplinary Imaging and Interventional Centre (C.A., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Ruth Coleman
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (A.A., R.C., R.R.H., J.E.M.), University of Oxford, UK
| | | | - Rury R. Holman
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (A.A., R.C., R.R.H., J.E.M.), University of Oxford, UK
| | - Hanan Lamlum
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Jisoo Lee
- John Radcliffe Hospital, Oxford University Hospitals National Health Service Foundation Trust, UK (J.L., E.W.)
| | - Daniel Massey
- Elderbrook Solutions GmbH on behalf of Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany (D.M.)
| | - Jack J.J.J. Miller
- Department of Physics (J.M.), University of Oxford, UK
- Department of Clinical Medicine, Aarhus University, Denmark (J.J.M.)
| | - Joanne E. Milton
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (A.A., R.C., R.R.H., J.E.M.), University of Oxford, UK
| | - Shveta Monga
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Ferenc E. Mózes
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Areesha Nazeer
- Oxford National Institutes of Health and Care Research Biomedical Research Centre, Oxford University Hospitals, Oxford, UK (R.R.H.)
| | - Betty Raman
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Oliver Rider
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Christopher T. Rodgers
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, Cambridge Biomedical Campus, UK (C.T.R.)
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava (L.V.)
| | - Eleanor Wicks
- John Radcliffe Hospital, Oxford University Hospitals National Health Service Foundation Trust, UK (J.L., E.W.)
| | - Masliza Mahmod
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
- Acute Multidisciplinary Imaging and Interventional Centre (C.A., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| |
Collapse
|
46
|
Oduah MT, Sundaram V, Reddy YNV. Epicardial Fat in Heart Failure with Preserved Ejection Fraction: Bad Actor or Just Lying Around? Card Fail Rev 2023; 9:e06. [PMID: 37397241 PMCID: PMC10311396 DOI: 10.15420/cfr.2022.25] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/04/2022] [Indexed: 07/04/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasingly recognised to be strongly associated with obesity and abnormalities in fat distribution. Epicardial fat has been associated with abnormal haemodynamics in HFpEF, with potential for direct mechanical effects on the heart causing constriction-like physiology and local myocardial remodelling effects from secretion of inflammatory and profibrotic mediators. However, patients with epicardial fat generally have more systemic and visceral adipose tissue making determination of causality between epicardial fat and HFpEF complex. In this review, we will summarise the evidence for epicardial fat being either directly causal in HFpEF pathogenesis or merely being a correlate of worse systemic inflammatory and generalised adiposity. We will also discuss therapies that directly target epicardial fat and may have potential for treating HFpEF and elucidating the independent role of epicardial fat in its pathogenesis.
Collapse
Affiliation(s)
| | - Varun Sundaram
- Division of Cardiovascular Diseases, Louis Stokes Cleveland Department of Veterans Affairs Medical CenterCleveland, OH, US
| | - Yogesh NV Reddy
- Department of Cardiovascular Disease, Mayo ClinicRochester, MN, US
| |
Collapse
|
47
|
Rossi VA, Gruebler M, Monzo L, Galluzzo A, Beltrami M. The Different Pathways of Epicardial Adipose Tissue across the Heart Failure Phenotypes: From Pathophysiology to Therapeutic Target. Int J Mol Sci 2023; 24:6838. [PMID: 37047810 PMCID: PMC10095298 DOI: 10.3390/ijms24076838] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
Epicardial adipose tissue (EAT) is an endocrine and paracrine organ constituted by a layer of adipose tissue directly located between the myocardium and visceral pericardium. Under physiological conditions, EAT exerts protective effects of brown-like fat characteristics, metabolizing excess fatty acids, and secreting anti-inflammatory and anti-fibrotic cytokines. In certain pathological conditions, EAT acquires a proatherogenic transcriptional profile resulting in increased synthesis of biologically active adipocytokines with proinflammatory properties, promoting oxidative stress, and finally causing endothelial damage. The role of EAT in heart failure (HF) has been mainly limited to HF with preserved ejection fraction (HFpEF) and related to the HFpEF obese phenotype. In HFpEF, EAT seems to acquire a proinflammatory profile and higher EAT values have been related to worse outcomes. Less data are available about the role of EAT in HF with reduced ejection fraction (HFrEF). Conversely, in HFrEF, EAT seems to play a nutritive role and lower values may correspond to the expression of a catabolic, adverse phenotype. As of now, there is evidence that the beneficial systemic cardiovascular effects of sodium-glucose cotransporter-2 receptors-inhibitors (SGLT2-i) might be partially mediated by inducing favorable modifications on EAT. As such, EAT may represent a promising target organ for the development of new drugs to improve cardiovascular prognosis. Thus, an approach based on detailed phenotyping of cardiac structural alterations and distinctive biomolecular pathways may change the current scenario, leading towards a precision medicine model with specific therapeutic targets considering different individual profiles. The aim of this review is to summarize the current knowledge about the biomolecular pathway of EAT in HF across the whole spectrum of ejection fraction, and to describe the potential of EAT as a therapeutic target in HF.
Collapse
Affiliation(s)
- Valentina A. Rossi
- University Heart Center, Department of Cardiology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Martin Gruebler
- Regional Hospital Neustadt, 2700 Wiener Neustadt, Austria
- Faculty of Medicine, Medical University of Graz, 8036 Graz, Austria
- Faculty of Medicine, Sigmund Freud University Vienna, 1020 Vienna, Austria
| | - Luca Monzo
- Centre d’Investigations Cliniques Plurithématique 1433 and Inserm U1116, Université de Lorraine, CHRU Nancy, FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), 54035 Nancy, France
| | | | - Matteo Beltrami
- Azienda USL Toscana Centro, Cardiology Unit, San Giovanni di Dio Hospital, 50143 Florence, Italy;
| |
Collapse
|
48
|
Zhang TY, An DA, Zhou H, Ni Z, Wang Q, Chen B, Lu R, Huang J, Zhou Y, Kim DH, Wilson M, Wu LM, Mou S. Texture analysis of native T1 images as a novel method for non-invasive assessment of heart failure with preserved ejection fraction in end-stage renal disease patients. Eur Radiol 2023; 33:2027-2038. [PMID: 36260118 DOI: 10.1007/s00330-022-09177-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To explore the diagnostic potential of texture analysis applied to native T1 maps obtained from cardiac magnetic resonance (CMR) images for the assessment of heart failure with preserved ejection fraction (HFpEF) among patients with end-stage renal disease (ESRD). METHODS This study, conducted from June 2018 to November 2020, included 119 patients (35 on hemodialysis, 55 on peritoneal dialysis, and 29 with kidney transplants) in Renji Hospital. Native T1 maps were assessed with texture analysis, using a freely available software package, in participants who underwent cardiac MRI at 3.0 T. Four texture features, selected by dimension reduction specific to the diagnosis of HFpEF, were analyzed. Multivariate logistic regression was performed to examine the independent association between the selected features and HFpEF in ESRD patients. RESULTS Seventy-six of 119 patients were diagnosed with HFpEF. Demographic, laboratory, cardiac MRI, and echocardiogram characteristics were compared between HFpEF and non-HFpEF groups. The four texture features that were analyzed showed statistically significant differences between groups. In multivariate analysis, age, left atrial volume index (LAVI), and sum average 4 (SA4) turned out to be independent predictors for HFpEF in ESRD patients. Combining the texture feature, SA4, with typical predictive factors resulted in higher C-index (0.923 vs. 0.898, p = 0.045) and a sensitivity and specificity of 79.2% and 95.2%, respectively. CONCLUSIONS Texture analysis of T1 maps adds diagnostic value to typical clinical parameters for the assessment of heart failure with preserved ejection fraction in patients with end-stage renal disease. KEY POINTS • Non-invasive assessment of HFpEF can help predict prognosis in ESRD patients and help them take timely preventative measures. • Texture analysis of native T1 maps adds diagnostic value to the typical clinical parameters for the assessment of HFpEF in patients with ESRD.
Collapse
Affiliation(s)
- Tian-Yi Zhang
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Dong-Aolei An
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Hang Zhou
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Zhaohui Ni
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Qin Wang
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Binghua Chen
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Renhua Lu
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Jiaying Huang
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Yin Zhou
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China
| | - Doo Hee Kim
- Department of Radiology, Wayne State University, Detroit, MI, 48201, USA
| | - Molly Wilson
- Department of Radiology, Wayne State University, Detroit, MI, 48201, USA
| | - Lian-Ming Wu
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China.
| | - Shan Mou
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
49
|
Theofilis P, Oikonomou E, Tsioufis K, Tousoulis D. Diabetes Mellitus and Heart Failure: Epidemiology, Pathophysiologic Mechanisms, and the Role of SGLT2 Inhibitors. Life (Basel) 2023; 13:497. [PMID: 36836854 PMCID: PMC9968235 DOI: 10.3390/life13020497] [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: 01/15/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Diabetes mellitus (DM) and heart failure (HF) are frequently encountered afflictions that are linked by a common pathophysiologic background. According to landmark studies, those conditions frequently coexist, and this interaction represents a poor prognostic indicator. Based on mechanistic studies, HF can be propagated by multiple pathophysiologic pathways, such as inflammation, oxidative stress, endothelial dysfunction, fibrosis, cardiac autonomic neuropathy, and alterations in substrate utilization. In this regard, DM may augment myocardial inflammation, fibrosis, autonomic dysfunction, and lipotoxicity. As the interaction between DM and HF appears critical, the new cornerstone in DM and HF treatment, sodium-glucose cotransporter-2 inhibitors (SGLT2i), may be able to revert the pathophysiology of those conditions and lead to beneficial HF outcomes. In this review, we aim to highlight the deleterious pathophysiologic interaction between DM and HF, as well as demonstrate the beneficial role of SGLT2i in this field.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Evangelos Oikonomou
- Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece
| | - Konstantinos Tsioufis
- Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Dimitris Tousoulis
- Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
| |
Collapse
|
50
|
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., G.G.S.).,Division of Internal Medicine, Department of Medicine, University of Padua, Italy (F.C., R.V.)
| | - Roberto Vettor
- Division of Internal Medicine, Department of Medicine, University of Padua, Italy (F.C., R.V.)
| | - 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., G.G.S.).,Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité - Universitätsmedizin Berlin, Germany (G.G.S.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (G.G.S.).,Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (G.G.S.)
| |
Collapse
|