1
|
Stefanska K, Sattar N. Incretin-based weight loss therapies and heart failure with preserved ejection fraction: guideline impactful results, but mechanisms unclear. Cardiovasc Res 2025; 121:1-3. [PMID: 39887081 DOI: 10.1093/cvr/cvaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 02/01/2025] Open
Affiliation(s)
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
2
|
Ruperez C, Madeo F, de Cabo R, Kroemer G, Abdellatif M. Obesity accelerates cardiovascular ageing. Eur Heart J 2025:ehaf216. [PMID: 40197620 DOI: 10.1093/eurheartj/ehaf216] [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: 10/13/2024] [Revised: 12/11/2024] [Accepted: 03/17/2025] [Indexed: 04/10/2025] Open
Abstract
A global obesity pandemic, coupled with an increasingly ageing population, is exacerbating the burden of cardiovascular disease. Indeed, clinical and experimental evidence underscores a potential connection between obesity and ageing in the pathogenesis of various cardiovascular disorders. This is further supported by the notion that weight reduction not only effectively reduces major cardiovascular events in elderly individuals but is also considered the gold standard for lifespan extension, in obese and non-obese model organisms. This review evaluates the intricate interplay between obesity and ageing from molecular mechanisms to whole organ function within the cardiovascular system. By comparatively analysing their characteristic features, shared molecular and cell biological signatures between obesity and ageing are unveiled, with the intent to shed light on how obesity accelerates cardiovascular ageing. This review also elaborates on how emerging metabolic interventions targeting obesity might protect from cardiovascular diseases largely through antagonizing key molecular mechanisms of the ageing process itself. In sum, this review aims to provide valuable insight into how understanding these interconnected processes could guide the development of novel and effective cardiovascular therapeutics for a growing aged population with a concerning obesity problem.
Collapse
Affiliation(s)
- Celia Ruperez
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, Graz 8036, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 15 Rue de l'École de Médecine, Paris 75006, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif 94805, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, 20 Rue Leblanc, Paris 75015, France
| | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, Graz 8036, Austria
- BioTechMed-Graz, 8010 Graz, Austria
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 15 Rue de l'École de Médecine, Paris 75006, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif 94805, France
| |
Collapse
|
3
|
Luo L, Zuo Y, Dai L. Metabolic rewiring and inter-organ crosstalk in diabetic HFpEF. Cardiovasc Diabetol 2025; 24:155. [PMID: 40186193 PMCID: PMC11971867 DOI: 10.1186/s12933-025-02707-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents a significant and growing clinical challenge. Initially, for an extended period, HFpEF was simply considered as a subset of heart failure, manifesting as haemodynamic disorders such as hypertension, myocardial hypertrophy, and diastolic dysfunction. However, the rising prevalence of obesity and diabetes has reshaped the HFpEF phenotype, with nearly 45% of cases coexisting with diabetes. Currently, it is recognized as a multi-system disorder that involves the heart, liver, kidneys, skeletal muscle, adipose tissue, along with immune and inflammatory signaling pathways. In this review, we summarize the landscape of metabolic rewiring and the crosstalk between the heart and other organs/systems (e.g., adipose, gut, liver and hematopoiesis system) in diabetic HFpEF for the first instance. A diverse array of metabolites and cytokines play pivotal roles in this intricate crosstalk process, with metabolic rewiring, chronic inflammatory responses, immune dysregulation, endothelial dysfunction, and myocardial fibrosis identified as the central mechanisms at the heart of this complex interplay. The liver-heart axis links nonalcoholic steatohepatitis and HFpEF through shared lipid accumulation, inflammation, and fibrosis pathways, while the gut-heart axis involves dysbiosis-driven metabolites (e.g., trimethylamine N-oxide, indole-3-propionic acid and short-chain fatty acids) impacting cardiac function and inflammation. Adipose-heart crosstalk highlights epicardial adipose tissue as a source of local inflammation and mechanical stress, whereas the hematopoietic system contributes via immune cell activation and cytokine release. We contend that, based on the viewpoints expounded in this review, breaking this inter-organ/system vicious cycle is the linchpin of treating diabetic HFpEF.
Collapse
Affiliation(s)
- Lingyun Luo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China
| | - Yuyue Zuo
- Department of Dermatology, Wuhan No. 1 Hospital, Wuhan, 430030, Hubei, China.
| | - Lei Dai
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China.
| |
Collapse
|
4
|
Sequeira V, Theisen J, Ermer KJ, Oertel M, Xu A, Weissman D, Ecker K, Dudek J, Fassnacht M, Nickel A, Kohlhaas M, Maack C, Dischinger U. Semaglutide normalizes increased cardiomyocyte calcium transients in a rat model of high fat diet-induced obesity. ESC Heart Fail 2025; 12:1386-1397. [PMID: 39482267 PMCID: PMC11911617 DOI: 10.1002/ehf2.15152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024] Open
Abstract
AIMS Obesity increases the risk of heart failure with preserved (HFpEF), but not reduced ejection fraction (HFrEF). The glucagon-like peptide-1 receptor agonist (GLP-1-RA) semaglutide improves outcome of patients with obesity with or without HFpEF, while GLP-1-RAs were associated with adverse outcome in patients with HFrEF. Here, we investigate the effect of in vivo treatment with semaglutide on excitation-contraction coupling in a rat model of obesity. METHODS AND RESULTS Rats received high-fat/high-fructose diet for 8 weeks and were then randomized to semaglutide (HFD/Sema) or vehicle (HFD/Veh) for another 8 weeks, during which they could choose between HFD and a low-fat/high-fructose diet (LFD). Control rats received either standard chow (CON), HFD or LFD only, without treatment. After 16 weeks, sarcomere shortening and cytosolic Ca2+ concentrations ([Ca2+]c) were determined in isolated cardiomyocytes. Compared with CON, HFD/Veh increased the amplitude of [Ca2+]c transients and systolic sarcomere shortening in absence or presence of β-adrenergic stimulation, which was reversed by HFD/Sema. Caffeine-induced sarcoplasmic reticulum (SR) Ca2+ release and L-type Ca2+ channel (LTCC) currents were reduced by HFD/Sema versus HFD/Veh, while SR Ca2+ ATPase activity remained unaffected. Compared with HFD, LFD increased [Ca2+]c transients and sarcomere shortening further despite similar effects on body weight. CONCLUSIONS While HFD increased cardiomyocyte [Ca2+]c transients and systolic sarcomere shortening, semaglutide normalized these alterations, mediated by reduced SR Ca2+ load and LTCC currents. Because increased LTCC currents were previously traced to cardiac hypertrophy, these effects may explain why GLP-1-RAs provide benefits for patients with obesity with or without HFpEF, but rather adverse outcome in HFrEF.
Collapse
Affiliation(s)
- Vasco Sequeira
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Julia Theisen
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Katharina J Ermer
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Marie Oertel
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany
| | - Anton Xu
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - David Weissman
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Katharina Ecker
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Jan Dudek
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Martin Fassnacht
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany
| | - Alexander Nickel
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Michael Kohlhaas
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
| | - Ulrich Dischinger
- Department of Translational Science Universitätsklinikum, DZHI, Würzburg, Germany
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
Krammer T, Baier MJ, Hegner P, Zschiedrich T, Lukas D, Wolf M, Le Phu C, Lutz V, Evert K, Kozakov K, Li J, Holzamer A, Maier LS, Provaznik Z, Bers DM, Wagner S, Mustroph J. Cardioprotective effects of semaglutide on isolated human ventricular myocardium. Eur J Heart Fail 2025. [PMID: 40107718 DOI: 10.1002/ejhf.3644] [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: 10/18/2024] [Revised: 12/19/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
AIMS Semaglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, has shown promising effects in reducing cardiovascular events in patients with obesity and heart failure (HF) with preserved ejection fraction (HFpEF) irrespective of concomitant diabetes. However, the exact mechanisms underlying its cardioprotective actions remain unclear. Our study investigates the direct effects of semaglutide on human cardiomyocytes, focusing on calcium (Ca) and sodium (Na) handling and its potential to improve myocardial contractility. METHODS AND RESULTS Human left ventricular cardiomyocytes were isolated from non-failing (NF) hearts, patients with aortic stenosis and a HFpEF-like phenotype (AS), and those with end-stage HF with reduced ejection fraction (HFrEF). Late Na current (INa), sarcoplasmic reticulum (SR) Ca leak, and contractility were assessed in isolated cardiomyocytes treated with semaglutide. CaMKII inhibitor autocamtide-2-related inhibitory peptide and GLP-1 receptor antagonist exendin 9-39 (Ex-9-39) were used to elucidate signalling pathways. Semaglutide reduced late INa in AS and HFrEF cardiomyocytes to levels comparable to NF. Additionally, semaglutide decreased diastolic SR Ca leak and improved systolic Ca transients and contractility in AS and HFrEF tissue. These effects were mediated through GLP-1 receptor agonism and were comparable to CaMKII inhibition. In multicellular preparations, semaglutide differentially improved myocardial contractility in AS and HFrEF in a dose-dependent manner. CONCLUSION Semaglutide directly modulates ion homeostasis in human cardiomyocytes, reducing proarrhythmic diastolic SR Ca leak and enhancing systolic function, which may explain its observed clinical benefits. These findings provide mechanistic insights into the cardioprotective effects of semaglutide and suggest its potential therapeutic use in HF.
Collapse
Affiliation(s)
- Thomas Krammer
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Maria J Baier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Tilman Zschiedrich
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - David Lukas
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Wolf
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Christian Le Phu
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Vanessa Lutz
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Katja Evert
- Institute for Pathology, University of Regensburg, Regensburg, Germany
| | - Kostiantyn Kozakov
- Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Jing Li
- Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Holzamer
- Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lars S Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Zdenek Provaznik
- Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Julian Mustroph
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
6
|
Ogurtsova E, Arefieva T, Filatova A, Radyukhina N, Ovchinnikov A. Cardiometabolic Phenotype in HFpEF: Insights from Murine Models. Biomedicines 2025; 13:744. [PMID: 40149720 PMCID: PMC11940576 DOI: 10.3390/biomedicines13030744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) remains a significant challenge in modern healthcare. It accounts for the majority of heart failure cases and their number worldwide is steadily increasing. With its high prevalence and substantial clinical impact, therapeutic strategies for HFpEF are still inadequate. This review focuses on the cardiometabolic phenotype of HFpEF which is characterised by such conditions as obesity, type 2 diabetes mellitus, and hypertension. Various murine models that mimic this phenotype are discussed. Each model's pathophysiological aspects, namely inflammation, oxidative stress, endothelial dysfunction, changes in cardiomyocyte protein function, and myocardial metabolism alterations are examined in detail. Understanding these models can provide insight into the mechanisms underlying HFpEF and aid in the development of effective therapeutic interventions.
Collapse
Affiliation(s)
- Ekaterina Ogurtsova
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (E.O.); (T.A.); (N.R.)
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Prospekt, 27/1, 117192 Moscow, Russia
| | - Tatiana Arefieva
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (E.O.); (T.A.); (N.R.)
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Prospekt, 27/1, 117192 Moscow, Russia
| | - Anastasiia Filatova
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (E.O.); (T.A.); (N.R.)
- Laboratory of Myocardial Fibrosis and Heart Failure with Preserved Ejection Fraction, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia;
| | - Natalya Radyukhina
- Laboratory of Cell Immunology, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia; (E.O.); (T.A.); (N.R.)
| | - Artem Ovchinnikov
- Laboratory of Myocardial Fibrosis and Heart Failure with Preserved Ejection Fraction, National Medical Research Center of Cardiology Named After Academician E.I. Chazov, Academician Chazov St., 15a, 121552 Moscow, Russia;
- Department of Clinical Functional Diagnostics, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, 127473 Moscow, Russia
| |
Collapse
|
7
|
Jani VP, Yoo EJ, Binek A, Guo A, Kim JS, Aguilan J, Keykhaei M, Jenkin SR, Sidoli S, Sharma K, Van Eyk JE, Kass DA, Hahn VS. Myocardial Proteome in Human Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2025; 14:e038945. [PMID: 40079330 DOI: 10.1161/jaha.124.038945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/16/2024] [Indexed: 03/15/2025]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) constitutes more than half of all HF but has few effective therapies. Recent human myocardial transcriptomics and metabolomics have identified major differences between HFpEF and controls. How this translates at the protein level is unknown. METHODS AND RESULTS Myocardial tissue from patients with HFpEF and nonfailing donor controls was analyzed by data-dependent acquisition (n=10 HFpEF, n=10 controls) and data-independent acquisition (n=44 HFpEF, n=5 controls) mass spectrometry-based proteomics. Differential protein expression analysis, pathway overrepresentation, weighted coexpression network analysis, and machine learning were integrated with clinical characteristics and previously reported transcriptomics. Principal component analysis (data-dependent acquisition-mass spectrometry) found HFpEF separated into 2 subgroups: one similar to controls and the other disparate. Downregulated proteins in HFpEF versus controls were enriched in mitochondrial transport/organization, translation, and metabolism including oxidative phosphorylation. Proteins upregulated in HFpEF were related to immune activation, reactive oxygen species, and inflammatory response. Ingenuity pathway analysis predicted downregulation of protein translation, mitochondrial function, and glucose and fat metabolism in HFpEF. Expression of oxidative phosphorylation and metabolism genes (higher) versus proteins (lower) was discordant in HFpEF versus controls. Data-independent acquisition-mass spectrometry proteomics also yielded 2 HFpEF subgroups; the one most different from controls had a higher proportion of patients with severe obesity and exhibited lower proteins related to fuel metabolism, oxidative phosphorylation, and protein translation. Three modules of correlated proteins in HFpEF that correlated with left ventricular hypertrophy and right ventricular load related to (1) proteasome; (2) fuel metabolism; and (3) protein translation, oxidative phosphorylation, and sarcomere organization. CONCLUSIONS Integrative proteomics, transcriptomics, and pathway analysis supports a defect in both metabolism and translation in HFpEF. Patients with HFpEF with more distinct proteomic signatures from control more often had severe obesity, supporting therapeutic efforts targeting metabolism and translation, particularly in this subgroup.
Collapse
Affiliation(s)
- Vivek P Jani
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Edwin J Yoo
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Aleksandra Binek
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center Los Angeles CA USA
| | - Alina Guo
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Julie S Kim
- Department of Biochemistry Albert Einstein College of Medicine Bronx NY USA
| | - Jennifer Aguilan
- Department of Pathology Albert Einstein College of Medicine Bronx NY USA
| | - Mohammad Keykhaei
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Sydney R Jenkin
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Simone Sidoli
- Department of Biochemistry Albert Einstein College of Medicine Bronx NY USA
| | - Kavita Sharma
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center Los Angeles CA USA
| | - David A Kass
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
- Department of Pharmacology and Molecular Sciences, and Department of Biomedical Engineering Johns Hopkins University Baltimore MD USA
| | - Virginia S Hahn
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| |
Collapse
|
8
|
Alansari H, Lazzara G, Taha MB, Gorthi JR. The Impact of Obesity on Cardiovascular Diseases: Heart Failure. Methodist Debakey Cardiovasc J 2025; 21:44-52. [PMID: 39990757 PMCID: PMC11844025 DOI: 10.14797/mdcvj.1511] [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: 10/26/2024] [Accepted: 01/14/2025] [Indexed: 02/25/2025] Open
Abstract
Obesity and heart failure (HF) are two intersecting public health challenges, each with rising prevalence worldwide. Obesity alters cardiac structure and function, leading to ventricular systolic and diastolic dysfunction. However, weight loss interventions, whether through lifestyle changes, pharmacological agents, or bariatric surgery, can improve cardiac function, reduce symptoms, and lower hospitalization rates. Interestingly, the "obesity paradox" suggests that HF patients with obesity may experience better survival outcomes than HF patients with normal weight despite the adverse cardiac effects of obesity. Most importantly, focusing on strategies that aim to prevent HF in patients with obesity can potentially curb the burden of this chronic condition. This review explores the complex relationship between obesity and HF, emphasizing pathophysiological mechanisms, the paradoxical survival benefit, and the impact of weight loss strategies. A deeper understanding of this relationship is critical for optimizing care and outcomes in HF patients with obesity.
Collapse
Affiliation(s)
- Hatem Alansari
- Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, TX, US
| | - Gina Lazzara
- Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, TX, US
| | - Mohamad B. Taha
- Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, TX, US
| | - Janardhana R. Gorthi
- Houston Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, TX, US
| |
Collapse
|
9
|
Ala M, Moheb Aleaba M. The blood pressure-lowering property of subcutaneous semaglutide: a systematic review, meta-analysis, and meta-regression. J Endocrinol Invest 2025; 48:283-294. [PMID: 39347905 DOI: 10.1007/s40618-024-02459-3] [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: 05/06/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024]
Abstract
PURPOSE Semaglutide is a glucagon-like peptide (GLP1) receptor agonist with unprecedented weight-lowering and anti-hyperglycemic properties. Recent clinical trials reported that subcutaneous semaglutide can modulate blood pressure; however, its effect on blood pressure widely varied in different studies and different subgroups of patients. METHODS PubMed, Web of Science, Scopus, and the Cochrane Library were systematically searched from the inception to July 18, 2024. Due to high heterogeneity, a random-effects model was adopted to pool data. RESULTS Twenty clinical trials with 15,312 participants in the placebo group and 18,231 participants in the semaglutide group were included in this study. Subcutaneous semaglutide significantly decreased both systolic (WMD - 3.71 mmHg, 95% CI (-4.29, -3.13), I2: 50.2%) and diastolic (WMD - 1.10 mmHg, 95% CI (-1.58, -0.63), I2: 69.7%) blood pressure. Subgroup analyses indicated that the blood pressure-lowering property of subcutaneous semaglutide was greater among patients without diabetes, with lower baseline hemoglobin A1c (HbA1c), baseline body mass index (BMI) greater than 35 kg/m2, dose of semaglutide more than 1 mg/week, baseline systolic blood pressure equal or less than 130 mmHg, weight loss greater than 10 kg, and BMI reduction greater than 3 kg/m2. In addition, a treatment length of 50 to 100 weeks was associated with greater blood pressure-lowering effects in subgroup analysis. After adjusting for other factors, meta-regression revealed that placebo-adjusted weight change was independently correlated with the effect of semaglutide on systolic and diastolic blood pressure. CONCLUSION Subcutaneous semaglutide can significantly decrease systolic and diastolic blood pressure, particularly in selected groups of patients.
Collapse
Affiliation(s)
- Moein Ala
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Mohammadreza Moheb Aleaba
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
10
|
Dürschmied D, Weidner K, Nitschmann S. [Semaglutide for heart failure with preserved ejection fraction]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2025; 66:244-246. [PMID: 39820736 DOI: 10.1007/s00108-024-01844-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/19/2025]
Affiliation(s)
- Daniel Dürschmied
- Medizinische Klinik (Kardiologie, Hämostaseologie und Internistische Intensivmedizin), Universitätsmedizin Mannheim, Universitätsklinikum Mannheim GmbH, Medizinische Fakultät Mannheim der Universität Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Deutschland.
| | - Kathrin Weidner
- Medizinische Klinik (Kardiologie, Hämostaseologie und Internistische Intensivmedizin), Universitätsmedizin Mannheim, Universitätsklinikum Mannheim GmbH, Medizinische Fakultät Mannheim der Universität Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Deutschland.
| | | |
Collapse
|
11
|
Fuerlinger A, Stockner A, Sedej S, Abdellatif M. Caloric restriction and its mimetics in heart failure with preserved ejection fraction: mechanisms and therapeutic potential. Cardiovasc Diabetol 2025; 24:21. [PMID: 39827109 PMCID: PMC11742808 DOI: 10.1186/s12933-024-02566-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025] Open
Abstract
The global increase in human life expectancy, coupled with an unprecedented rise in the prevalence of obesity, has led to a growing clinical and socioeconomic burden of heart failure with preserved ejection fraction (HFpEF). Mechanistically, the molecular and cellular hallmarks of aging are omnipresent in HFpEF and are further exacerbated by obesity and associated metabolic diseases. Conversely, weight loss strategies, particularly caloric restriction, have shown promise in improving health status in patients with HFpEF and are considered the gold standard for promoting longevity and healthspan (disease-free lifetime) in model organisms. In this review, we implicate fundamental mechanisms of aging in driving HFpEF and elucidate how caloric restriction mitigates the disease progression. Furthermore, we discuss the potential for pharmacologically mimicking the beneficial effects of caloric restriction in HFpEF using clinically approved and emerging caloric restriction mimetics. We surmise that these compounds could offer novel therapeutic avenues for HFpEF and alleviate the challenges associated with the implementation of caloric restriction and other lifestyle modifications to reduce the burden of HFpEF at a population level.
Collapse
Affiliation(s)
- Alexander Fuerlinger
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
- BioTechMed-Graz, 8010, Graz, Austria
| | - Alina Stockner
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
- BioTechMed-Graz, 8010, Graz, Austria
- Faculty of Medicine, University of Maribor, 2000, Maribor, Slovenia
| | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria.
- BioTechMed-Graz, 8010, Graz, Austria.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805, Villejuif, France.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, 75006, France.
| |
Collapse
|
12
|
Pillarisetti L, Agrawal DK. Semaglutide: Double-edged Sword with Risks and Benefits. ARCHIVES OF INTERNAL MEDICINE RESEARCH 2025; 8:1-13. [PMID: 39902055 PMCID: PMC11790292 DOI: 10.26502/aimr.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Type 2 Diabetes Mellitus therapy has evolved over the years to now include a new class of therapeutics, semaglutide. This article reviews the mechanism of action and formulation of semaglutide therapy, potential benefits, contraindications, adverse effects, and drug interactions. Oral and subcutaneous semaglutide therapies have shown effectiveness in improving glycemic control, weight loss, and reducing cardiovascular risks associated with diabetes mellitus. Semaglutide has also shown potential in being used as a therapeutic strategy in Alzheimer's disease due to its anti-neuroinflammatory effects and being used to treat polycystic ovary syndrome. However, semaglutide therapy is also associated with concerning adverse effects like acute pancreatitis, anesthetic risks like pulmonary aspiration or residual gastric content, acute kidney injury, acute gallbladder injury, nonarteritic anterior ischemic optic neuropathy and diabetic retinopathy. Contraindications of semaglutide include history of medullary thyroid carcinoma or multiple endocrine neoplasia syndrome type 2, and pregnancy. Drug interactions to consider with semaglutide therapy include those also used in diabetes treatment, like metformin, as well as anti-psychotics, due to anti-psychotics associated weight gain. The findings of this article emphasize the need for a cross-disciplinary approach to understand the molecular mechanisms and clinical implications of semaglutide on patients with complex medical histories and treatment regimens. The potential anesthetic risks of semaglutide therapy warrant careful consideratiion with ethical concerns. Further studies can assess if there is a need to modify pre-operative guidelines to account for patient using semaglutide and how delayed gastric emptying and constitpation will affect surgical outcomes and complications. While semaglutide therapy for diabetes mellitus has been established, there is a need for extensive research on repurposing semaglutide in neurodegenerative disease treatment.
Collapse
Affiliation(s)
- Lekha Pillarisetti
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766 USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766 USA
| |
Collapse
|
13
|
Wang TY, Yang Q, Cheng XY, Ding JC, Hu PF. Beyond weight loss: the potential of glucagon-like peptide-1 receptor agonists for treating heart failure with preserved ejection fraction. Heart Fail Rev 2025; 30:17-38. [PMID: 39269643 DOI: 10.1007/s10741-024-10438-2] [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: 09/05/2024] [Indexed: 09/15/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome with various phenotypes, and obesity is one of the most common and clinically relevant phenotypes of HFpEF. Obesity contributes to HFpEF through multiple mechanisms, including sodium retention, neurohormonal dysregulation, altered energy substrate metabolism, expansion of visceral adipose tissue, and low-grade systemic inflammation. Glucagon-like peptide-1 (GLP-1) is a hormone in the incretin family. It is produced by specialized cells called neuroendocrine L cells located in the distal ileum and colon. GLP-1 reduces blood glucose levels by promoting glucose-dependent insulin secretion from pancreatic β cells, suppressing glucagon release from pancreatic α cells, and blocking hepatic gluconeogenesis. Recent evidence suggests that GLP-1 receptor agonists (GLP-1 RAs) can significantly improve physical activity limitations and exercise capacity in obese patients with HFpEF. The possible cardioprotective mechanisms of GLP-1 RAs include reducing epicardial fat tissue thickness, preventing activation of the renin-angiotensin-aldosterone system, improving myocardial energy metabolism, reducing systemic inflammation and cardiac oxidative stress, and delaying the progression of atherosclerosis. This review examines the impact of obesity on the underlying mechanisms of HFpEF, summarizes the trial data on cardiovascular outcomes of GLP-1 RAs in patients with type 2 diabetes mellitus, and highlights the potential cardioprotective mechanisms of GLP-1 RAs to give a pathophysiological and clinical rationale for using GLP-1 RAs in obese HFpEF patients.
Collapse
Affiliation(s)
- Tian-Yu Wang
- Department of The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Yang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin-Yi Cheng
- Department of The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun-Can Ding
- Department of The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peng-Fei Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
14
|
Lin K, Wang A, Zhai C, Zhao Y, Hu H, Huang D, Zhai Q, Yan Y, Ge J. Semaglutide protects against diabetes-associated cardiac inflammation via Sirt3-dependent RKIP pathway. Br J Pharmacol 2024. [PMID: 39710830 DOI: 10.1111/bph.17327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/12/2024] [Accepted: 06/27/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND AND PURPOSE Glucagon-like peptide-1 receptor agonists (GLP-1RAs) exert cardiovascular benefits in diabetic patients, but the underlying mechanisms remain incompletely understood. Semaglutide, a novel long-acting GLP-1RA, has shown a reduced risk of cardiovascular events. Based on these results, we investigated the therapeutic potential of semaglutide in diabetic cardiomyopathy and sought to elucidate the underlying mechanisms. EXPERIMENTAL APPROACH Mice with diabetes induced by high-fat diet/streptozotocin were treated with semaglutide. The mechanisms underlying the cardioprotective effects of semaglutide were analysed using animal and cell experiments. KEY RESULTS In diabetic mice, semaglutide alleviated metabolic disorders, ameliorated myocardial fibrosis, improved cardiac function, antagonized oxidative stress and suppressed cardiomyocyte apoptosis. More significantly, semaglutide attenuated cardiac inflammation through restoring Raf kinase inhibitor protein (RKIP) expression and inhibiting downstream TANK-binding kinase 1 (TBK1)-NF-κB pathway. Meanwhile, decreased RKIP expression and activated TBK1-NF-κB signalling pathway were also found in tissues from human diabetic hearts. RKIP deficiency exacerbated cardiac inflammation and offset the cardioprotective effect of semaglutide in diabetic mice. Moreover, semaglutide also restored the expression level of Sirtuin 3(Sirt3), which served as a modulator against cardiac inflammation by regulating RKIP-dependent pathway. In diabetic mice, RKIP deficiency abolished the cardioprotective benefits conferred by the Sirt3 activator honokiol. We also found that cAMP/PKA signalling, rather than glucose lowering, contributed to the anti-inflammatory effect of semaglutide through Sirt3-dependent RKIP pathway. CONCLUSIONS AND IMPLICATIONS Semaglutide exerted cardioprotective effects against diabetic heart failure by alleviating cardiac inflammation through Sirt3-dependent RKIP signalling pathway.
Collapse
Affiliation(s)
- Kaibin Lin
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ai Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changlin Zhai
- Department of Cardiovascular Diseases, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yun Zhao
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huilin Hu
- Department of Cardiovascular Diseases, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Dong Huang
- Heart Center, Shanghai Jiaotong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qiwei Zhai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yan Yan
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Cool D, Coventon J, Sharma A. Semaglutide Inducing Resolution of Proliferative Diabetic Retinopathy: A Case Report. Case Rep Ophthalmol Med 2024; 2024:5834769. [PMID: 39691771 PMCID: PMC11651726 DOI: 10.1155/crop/5834769] [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: 08/29/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024] Open
Abstract
Purpose: To describe a case of regression of proliferative diabetic retinopathy (PDR) following treatment with semaglutide. Methods: Case report. Results: The case describes a 47-year-old woman with Type 2 diabetes, obesity, hypertension, and dyslipidaemia who had difficulty controlling her blood sugar levels despite oral hypoglycaemic medications. She presented with PDR in her right eye and severe nonproliferative diabetic retinopathy (NPDR) in her left eye. After missing her follow-up appointment for panretinal photocoagulation (PRP), her general practitioner initiated semaglutide therapy. Despite minimal changes in glycaemic control, the patient exhibited resolution of neovascularisation in her right eye and improved diabetic macular oedema (DMO) within 6 weeks of semaglutide therapy. Conclusion: This case report suggests a potential independent role for semaglutide in managing PDR.
Collapse
Affiliation(s)
- Daniel Cool
- Ophthalmology Department, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - James Coventon
- Ophthalmology Department, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- Ophthalmology Department, Cairns Hospital, Cairns, Queensland, Australia
| | | |
Collapse
|
16
|
Muayad J, Loya A, Hussain ZS, Lee DH, Chauhan MZ, Lee AG, Movahedan A, Dahr SS. Influence of Common Medications on Diabetic Macular Edema in Type 2 Diabetes Mellitus. Ophthalmol Retina 2024:S2468-6530(24)00582-7. [PMID: 39644923 DOI: 10.1016/j.oret.2024.12.006] [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: 05/21/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
PURPOSE This study aimed to assess the impact of systemic medications, including glucagon-like peptide-1 receptor agonists (GLP-1 RAs), fenofibrates, thiazolidinediones (TZDs), and calcium channel blockers (CCBs), on the risk of developing diabetic macular edema (DME) in patients with type 2 diabetes mellitus (T2DM). DESIGN A retrospective cohort study was conducted using electronic medical records (EMR) data from the TriNetX health research network, covering a period from October 2004 to 2024. PARTICIPANTS The study population comprised patients diagnosed with T2DM who were newly initiated on GLP-1 RAs, fenofibrates, TZDs, or CCBs. Propensity score matched (PSM) controls were patients with T2DM who did not receive these medications within the same timeframe. METHODS Patients were observed for 1 to 2 years postmedication initiation to monitor the development of DME. The study used 1:1 propensity score matching to adjust for baseline characteristics and comorbidities. MAIN OUTCOME MEASURES The primary outcome measure was the incidence rate of DME within the 2-year follow-up period. Hazard ratios (HRs) with 95% confidence interval (CI) were calculated to compare the risk of DME between treatment and control groups. RESULTS After PSM, the study analyzed data from 107 193 patients in the CCB cohort, 76 583 in the GLP-1 agonists cohort, 25 657 in the TZDs cohort, and 18 606 in the fenofibrates cohort. Calcium channel blocker-treated patients demonstrated a higher risk of DME development compared with controls (HR: 1.66, 95% CI: 1.54-1.78). In contrast, GLP-1 RA-treated patients showed a decreased risk of DME (HR: 0.77, 95% CI: 0.70-0.85), as did fenofibrate-treated patients (HR: 0.83, 95% CI: 0.68-0.98). No significant difference in DME risk was observed in the TZDs cohort (HR: 1.08, 95% CI: 0.94-1.25). CONCLUSIONS Patients on GLP-1 RAs and fenofibrates experienced a lower risk of DME diagnosis, suggesting a protective effect against DME development in patients with T2DM, whereas those on CCBs experienced an increased risk. These findings suggest that systemic medications may significantly influence DME outcomes, warranting further investigation into their effects on retinal health. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Jawad Muayad
- School of Medicine, Texas A&M University, Houston, Texas
| | - Asad Loya
- Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas
| | - Zain S Hussain
- Department of Ophthalmology, University of Medicine and Health Sciences, New York, New York
| | - Debora H Lee
- Ruiz Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston, Texas
| | - Muhammad Z Chauhan
- Department of Ophthalmology, Harvey and Bernice Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Andrew G Lee
- School of Medicine, Texas A&M University, Houston, Texas; Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas; Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, Texas; Center for Space Medicine, Baylor College of Medicine, Houston, Texas; The Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas; Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine, New York, New York; Department of Ophthalmology, University of Texas Medical Branch, Galveston, Texas; Department of Ophthalmology, University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Ophthalmology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Asadolah Movahedan
- Department of Ophthalmology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Sami S Dahr
- Ruiz Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston, Texas.
| |
Collapse
|
17
|
Martens MD, Abuetabh Y, Schmidt MA, Zolondek MC, Silver HL, Levasseur JL, Ferdaoussi M, Dyck JR. Semaglutide Reduces Cardiomyocyte Size and Cardiac Mass in Lean and Obese Mice. JACC Basic Transl Sci 2024; 9:1429-1431. [PMID: 39822607 PMCID: PMC11733746 DOI: 10.1016/j.jacbts.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
|
18
|
Li S, Withaar C, Rodrigues PG, Zijlstra SN, de Boer RA, Silljé HHW, Meems LMG. The NLRP3-inflammasome inhibitor MCC950 improves cardiac function in a HFpEF mouse model. Biomed Pharmacother 2024; 181:117711. [PMID: 39616735 DOI: 10.1016/j.biopha.2024.117711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is posing a significant medical challenge due to its growing prevalence, high hospitalization rates and limited response to current treatment options. Accumulating evidence suggests that a comorbidity-driven systemic pro-inflammatory state, including activation of the NLRP3 inflammasome, contributes to the pathogenesis of HFpEF. This study aimed to investigate the potential cardiac protective effects of the selective NLRP3 inhibitor MCC950, in a mouse model of HFpEF. HFpEF was obtained in 18-22 months old female mice using high-fat diet (HFD) and angiotensin II (AngII) infusion. Mice developed HFpEF and comorbidities such as obesity, type 2 diabetes, and hypertension. MCC950 was added to HFD and groups were treated for four weeks until the study endpoint. MCC950 treatment resulted in lower plasma IL-18 levels (-47.3 %), illustrating target engagement. First, we observed that MCC950 treatment improved left ventricular function, demonstrated by enhanced global longitudinal strain (GLS, 3.9 %, P<0.01) and reverse peak longitudinal strain (RPLSR, +46.8 %, P<0.05). Second, MCC950 reduced cardiac hypertrophy (cardiomyocyte size -19.5 %, P<0.001) and fibrosis (-32.5 %, P<0.05), accompanied by lower expression of pro-fibrotic genes. Finally, MCC950 treatment reduced macrophage infiltration in left ventricular tissue and attenuated macrophage accumulation in visceral adipose tissue, even more as compared to caloric restriction. Overall, this suggests that NLRP3 inhibition could be a promising treatment for HFpEF patients with a pro-inflammatory profile, potentially improving heart function, systemic inflammation, and metabolic parameters.
Collapse
Affiliation(s)
- Sunhuo Li
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Coenraad Withaar
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Patricia G Rodrigues
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sietske N Zijlstra
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands; Cardiovascular Research Institute, Thorax Center, Department of Cardiology, Erasmus Medical Center, Rotterdam 3015 GD, the Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Laura M G Meems
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands.
| |
Collapse
|
19
|
Balata M, Becher MU. Impact of semaglutide on weight and functional outcomes among obese heart failure patients: a propensity scores matching analysis. BMC Cardiovasc Disord 2024; 24:590. [PMID: 39462311 PMCID: PMC11515153 DOI: 10.1186/s12872-024-04275-2] [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: 02/04/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND & OBJECTIVES Obesity is a common comorbidity in heart failure, yet effective pharmacological options for weight loss in these patients are limited. Semaglutide, a glucagon-like peptide 1 receptor agonist, has shown promise for weight reduction in obese adults. This study aims to evaluate semaglutide's impact on weight loss, functional status, and clinical outcomes in obese patients with heart failure. METHODS A retrospective analysis was conducted on all consecutive obese (BMI > 30 kg/m²) patients with heart failure at the University Hospital Bonn outpatient clinic from July 2019 to July 2022. Propensity score matching paired patients receiving semaglutide as an add-on therapy (SEMA) with those on medical therapy alone (Control). RESULTS Among 1,942 patients with heart failure screened, 26 matched pairs were identified. At one year, the SEMA group exhibited significant weight loss, with a mean BMI reduction of -2.91 kg/m² (95% CI: -4.27 to -1.55; p < 0.001), while the control group showed a non-significant mean change of -0.41 kg/m² (95% CI: -1.08 to 0.26; p = 0.22). The difference in BMI between the two groups was statistically significant (mean difference: 3.42 kg/m², 95% CI: 1.43 to 5.42; p = 0.001). Improvements by at least one NYHA class were observed in 65% of the SEMA group (p < 0.001) compared to 15% of the control group (p = 0.18). The SEMA group also showed a significant increase in 6-minute walk distance (6MWD), with a mean difference of 75 m between the groups at one year (95% CI: 0.53 to 150.02; p = 0.049). NT-proBNP levels significantly decreased in the SEMA group (p < 0.001) compared to the control group (p = 0.78), with a statistically significant difference in NT-proBNP between the groups (p = 0.048). Both improvements in 6MWD and reductions in NT-proBNP were significantly correlated with BMI percentage reductions. CONCLUSIONS Semaglutide was associated with significant weight reduction in obese patients with heart failure, accompanied by improved NYHA classification and 6-minute walk distance. Larger, multi-center trials and prospective, randomized controlled trials are warranted. These studies should focus on assessing long-term outcomes, optimizing dosage, and exploring the potential cardiovascular benefits beyond weight reduction.
Collapse
Affiliation(s)
- Mahmoud Balata
- University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| | - Marc Ulrich Becher
- University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Internal Medicine and Cardiology, City hospital Solingen, Gotenstraße 1, 42653, Solingen, Germany
| |
Collapse
|
20
|
Yang X, Lin R, Feng C, Kang Q, Yu P, Deng Y, Jin Y. Research Progress on Peptide Drugs for Type 2 Diabetes and the Possibility of Oral Administration. Pharmaceutics 2024; 16:1353. [PMID: 39598478 PMCID: PMC11597531 DOI: 10.3390/pharmaceutics16111353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Diabetes is a global disease that can lead to a range of complications. Currently, the treatment of type 2 diabetes focuses on oral hypoglycemic drugs and insulin analogues. Studies have shown that drugs such as oral metformin are useful in the treatment of diabetes but can limit the liver's ability to release sugar. The development of glucose-lowering peptides has provided new options for the treatment of type 2 diabetes. Peptide drugs have low oral utilization due to their easy degradation, short half-life, and difficulty passing through the intestinal mucosa. Therefore, improving the oral utilization of peptide drugs remains an urgent problem. This paper reviews the research progress of peptide drugs in the treatment of diabetes mellitus and proposes that different types of nano-formulation carriers, such as liposomes, self-emulsifying drug delivery systems, and polymer particles, should be combined with peptide drugs for oral administration to improve their absorption in the gastrointestinal tract.
Collapse
Affiliation(s)
- Xinxin Yang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.Y.); (R.L.)
| | - Ruiting Lin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.Y.); (R.L.)
| | - Changzhuo Feng
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China; (C.F.); (Q.K.); (P.Y.)
| | - Qiyuan Kang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China; (C.F.); (Q.K.); (P.Y.)
| | - Peng Yu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China; (C.F.); (Q.K.); (P.Y.)
| | - Yongzhi Deng
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.Y.); (R.L.)
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.Y.); (R.L.)
| |
Collapse
|
21
|
Martos-Guillami N, Vergara A, Llorens-Cebrià C, Motto AE, Martínez-Díaz I, Gonçalves F, Garcias-Ramis MM, Allo-Urzainqui E, Narváez A, Bermejo S, Muñoz V, León-Román J, Ferrer-Costa R, Jacobs-Cachá C, Vilardell-Vilà J, Soler MJ. SGLT2i and GLP1-RA exert additive cardiorenal protection with a RAS blocker in uninephrectomized db/db mice. Front Pharmacol 2024; 15:1415879. [PMID: 39434906 PMCID: PMC11491409 DOI: 10.3389/fphar.2024.1415879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/20/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Diabetic Kidney Disease (DKD) is the main cause of end-stage renal disease in the developed world. The current treatment of the DKD with renin-angiotensin system (RAS) blockade does not totally halt the progression to end stage kidney disease. Currently, several drugs have shown to delay DKD progression such as sodium-glucose co-transporter-2 inhibitors (SGLT2i) and glucagon-like-1 receptor agonists (GLP-1RA). We hypothesized that by combining several drugs that prevent DKD progression on top of RAS blockade a synergistic effect would be achieved in terms of cardiorenal protection. In the present study, we analysed if the combination of a RAS blocker (ramipril) with a SGLT2i (empagliflozin) and/or GLP-1RA (semaglutide) in a type 2 diabetic mouse model could have add-on effects in kidney and heart protection. Methods Male and female uninephrectomized type 2 diabetic db/db mice were treated with empagliflozin and/or semaglutide on top of ramipril during 8 weeks. During the study body weight, water and food intake were weekly monitored, glycaemia biweekly and albuminuria and glomerular filtration rate (GFR) before and after the treatment. At the end of the experiment, kidney and heart were isolated for histological and gene expression studies as well as for intrarenal RAS state assessment. Results Semaglutide combined with ramipril and/or empagliflozin significantly decreased albuminuria but only when combined with both compounds, semaglutide further decreased blood glucose, glomerular hyperfiltration in male mice and glomerular mesangial matrix expansion. In kidney, only the triple treatment with empagliflozin, semaglutide and ramipril reduced the expression of the proinflammatory and profibrotic genes ccl2 and TGFß1. In addition, the combination of empagliflozin and semaglutide on top of RAS blockade was superior in decreasing cardiomyocyte hypertrophy and heart fibrosis in db/db mice. Discussion Our results suggest that the combination of SGLT2i with GLP-1RA is superior in cardiorenal protection in DKD than the drugs administered alone on top of RAS blockade.
Collapse
Affiliation(s)
- Nerea Martos-Guillami
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ander Vergara
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III (RD21/0005/0016), Madrid, Spain
| | - Carmen Llorens-Cebrià
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Aku Enam Motto
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Laboratory of Physiology/ Pharmacology, Unit of Pathophysiology, Bioactive Substances and Safety, Faculty of Sciences, University of Lomé, Lomé, Togo
| | - Irene Martínez-Díaz
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Francisco Gonçalves
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Maria Magdalena Garcias-Ramis
- Clinical Biochemistry Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus. Barcelona, Barcelona, Spain
| | - Estibaliz Allo-Urzainqui
- Clinical Biochemistry Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus. Barcelona, Barcelona, Spain
| | - Alonso Narváez
- Urology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Sheila Bermejo
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III (RD21/0005/0016), Madrid, Spain
| | - Vicent Muñoz
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Juan León-Román
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Roser Ferrer-Costa
- Clinical Biochemistry Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus. Barcelona, Barcelona, Spain
| | - Conxita Jacobs-Cachá
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III (RD21/0005/0016), Madrid, Spain
- Clinical Biochemistry Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus. Barcelona, Barcelona, Spain
| | - Jordi Vilardell-Vilà
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - María José Soler
- Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Nephrology Department, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III (RD21/0005/0016), Madrid, Spain
| |
Collapse
|
22
|
Valentine Y, Nikolajczyk BS. T cells in obesity-associated inflammation: The devil is in the details. Immunol Rev 2024; 324:25-41. [PMID: 38767210 PMCID: PMC11694249 DOI: 10.1111/imr.13354] [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] [Indexed: 05/22/2024]
Abstract
Obesity presents a significant health challenge, affecting 41% of adults and 19.7% of children in the United States. One of the associated health challenges of obesity is chronic low-grade inflammation. In both mice and humans, T cells in circulation and in the adipose tissue play a pivotal role in obesity-associated inflammation. Changes in the numbers and frequency of specific CD4+ Th subsets and their contribution to inflammation through cytokine production indicate declining metabolic health, that is, insulin resistance and T2D. While some Th subset alterations are consistent between mice and humans with obesity, some changes mainly characterize male mice, whereas female mice often resist obesity and inflammation. However, protection from obesity and inflammation is not observed in human females, who can develop obesity-related T-cell inflammation akin to males. The decline in female sex hormones after menopause is also implicated in promoting obesity and inflammation. Age is a second underappreciated factor for defining and regulating obesity-associated inflammation toward translating basic science findings to the clinic. Weight loss in mice and humans, in parallel with these other factors, does not resolve obesity-associated inflammation. Instead, inflammation persists amid modest changes in CD4+ T cell frequencies, highlighting the need for further research into resolving changes in T-cell function after weight loss. How lingering inflammation after weight loss affecting the common struggle to maintain lower weight is unknown. Semaglutide, a newly popular pharmaceutical used for treating T2D and reversing obesity, holds promise for alleviating obesity-associated health complications, yet its impact on T-cell-mediated inflammation remains unexplored. Further work in this area could significantly contribute to the scientific understanding of the impacts of weight loss and sex/hormones in obesity and obesity-associated metabolic decline.
Collapse
Affiliation(s)
- Yolander Valentine
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Barbara S. Nikolajczyk
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
23
|
Yaribeygi H, Maleki M, Jamialahmadi T, Sahebkar A. Anti-inflammatory benefits of semaglutide: State of the art. J Clin Transl Endocrinol 2024; 36:100340. [PMID: 38576822 PMCID: PMC10992717 DOI: 10.1016/j.jcte.2024.100340] [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: 02/05/2024] [Revised: 03/06/2024] [Accepted: 03/20/2024] [Indexed: 04/06/2024] Open
Abstract
Individuals with diabetes often have chronic inflammation and high levels of inflammatory cytokines, leading to insulin resistance and complications. Anti-inflammatory agents are proposed to prevent these issues, including using antidiabetic medications with anti-inflammatory properties like semaglutide, a GLP-1 analogue. Semaglutide not only lowers glucose but also shows potential anti-inflammatory effects. Studies suggest it can modulate inflammatory responses and benefit those with diabetes. However, the exact mechanisms of its anti-inflammatory effects are not fully understood. This review aims to discuss the latest findings on semaglutide's anti-inflammatory effects and the potential pathways involved.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mina Maleki
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Rahmouni K. Neural Circuits Underlying Reciprocal Cardiometabolic Crosstalk: 2023 Arthur C. Corcoran Memorial Lecture. Hypertension 2024; 81:1233-1243. [PMID: 38533662 PMCID: PMC11096079 DOI: 10.1161/hypertensionaha.124.22066] [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] [Indexed: 03/28/2024]
Abstract
The interplay of various body systems, encompassing those that govern cardiovascular and metabolic functions, has evolved alongside the development of multicellular organisms. This evolutionary process is essential for the coordination and maintenance of homeostasis and overall health by facilitating the adaptation of the organism to internal and external cues. Disruption of these complex interactions contributes to the development and progression of pathologies that involve multiple organs. Obesity-associated cardiovascular risks, such as hypertension, highlight the significant influence that metabolic processes exert on the cardiovascular system. This cardiometabolic communication is reciprocal, as indicated by substantial evidence pointing to the ability of the cardiovascular system to affect metabolic processes, with pathophysiological implications in disease conditions. In this review, I outline the bidirectional nature of the cardiometabolic interaction, with special emphasis on the impact that metabolic organs have on the cardiovascular system. I also discuss the contribution of the neural circuits and autonomic nervous system in mediating the crosstalk between cardiovascular and metabolic functions in health and disease, along with the molecular mechanisms involved.
Collapse
Affiliation(s)
- Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
25
|
Kusayev J, Levy Y, Weininger D, Frishman WH, Aronow WS. Semaglutide in Heart Failure With Preserved Ejection Fraction: Exploring Recent Evidence in Therapeutic Potential for the Obese Population. Cardiol Rev 2024:00045415-990000000-00269. [PMID: 38757954 DOI: 10.1097/crd.0000000000000726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is an increasingly prevalent condition, particularly among the aging population in the United States, and is associated with significant challenges due to its complex pathophysiology and limited therapeutic options. Historically, few pharmacological therapies have successfully mitigated HFpEF, making the emergence of effective treatments particularly significant. This review evaluates recent evidence on the therapeutic potential of semaglutide for managing HFpEF, especially in the obese population. Results from the STEP-HFpEF and STEP-HFpEF DM trials demonstrate that semaglutide, a glucagon-like peptide-1 receptor agonist originally developed for type 2 diabetes but now also approved for obesity treatment, significantly improves clinical outcomes such as symptom scores, body weight, exercise capacity, and inflammation markers in the obese population suffering from HFpEF. These improvements are attributed to both the weight loss induced by semaglutide and its direct effects on the congestive pathophysiology of HFpEF. The efficacy of semaglutide offers new hope for addressing a condition that has long lacked effective pharmacological interventions.
Collapse
Affiliation(s)
- Josef Kusayev
- From the Department of Medicine, New York Medical College, Valhalla, NY
| | - Yisrael Levy
- From the Department of Medicine, New York Medical College, Valhalla, NY
| | - David Weininger
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| | | | - Wilbert S Aronow
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
26
|
Capone F, Nambiar N, Schiattarella GG. Beyond Weight Loss: the Emerging Role of Incretin-Based Treatments in Cardiometabolic HFpEF. Curr Opin Cardiol 2024; 39:148-153. [PMID: 38294187 DOI: 10.1097/hco.0000000000001117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
PURPOSE OF REVIEW Incretin-based drugs are potent weight-lowering agents, emerging as potential breakthrough therapy for the treatment of obesity-related phenotype of heart failure with preserved ejection fraction (HFpEF). In this review article, we will discuss the contribution of weight loss as part of the benefits of incretin-based medications in obese patients with HFpEF. Furthermore, we will describe the potential effects of glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptor agonists on the heart, particularly in relation to HFpEF pathophysiology. RECENT FINDINGS In the STEP-HFpEF trial, the GLP-1 receptor agonist semaglutide significantly improved quality of life outcomes in obese HFpEF patients. Whether the beneficial effects of semaglutide in obese patients with HFpEF are merely a consequence of body weight reduction is unclear. Considering the availability of other weight loss strategies (e.g., caloric restriction, exercise training, bariatric surgery) to be used in obese HFpEF patients, answering this question is crucial to provide tailored therapeutic options in these subjects. SUMMARY Incretin-based drugs may represent a milestone in the treatment of obesity in HFpEF. Elucidating the contribution of weight loss in the overall benefit observed with these drugs is critical in the management of obese HFpEF patients, considering that other weight-lowering strategies are available and might represent potential alternative options for these patients.
Collapse
Affiliation(s)
- Federico Capone
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Division of Internal Medicine, Department of Medicine, University of Padua, Padua, Italy
| | - Natasha Nambiar
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gabriele G Schiattarella
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité -Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
27
|
Park B, Bakbak E, Teoh H, Krishnaraj A, Dennis F, Quan A, Rotstein OD, Butler J, Hess DA, Verma S. GLP-1 receptor agonists and atherosclerosis protection: the vascular endothelium takes center stage. Am J Physiol Heart Circ Physiol 2024; 326:H1159-H1176. [PMID: 38426865 DOI: 10.1152/ajpheart.00574.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Atherosclerotic cardiovascular disease is a chronic condition that often copresents with type 2 diabetes and obesity. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are incretin mimetics endorsed by major professional societies for improving glycemic status and reducing atherosclerotic risk in people living with type 2 diabetes. Although the cardioprotective efficacy of GLP-1RAs and their relationship with traditional risk factors are well established, there is a paucity of publications that have summarized the potentially direct mechanisms through which GLP-1RAs mitigate atherosclerosis. This review aims to narrow this gap by providing comprehensive and in-depth mechanistic insight into the antiatherosclerotic properties of GLP-1RAs demonstrated across large outcome trials. Herein, we describe the landmark cardiovascular outcome trials that triggered widespread excitement around GLP-1RAs as a modern class of cardioprotective agents, followed by a summary of the origins of GLP-1RAs and their mechanisms of action. The effects of GLP-1RAs at each major pathophysiological milestone of atherosclerosis, as observed across clinical trials, animal models, and cell culture studies, are described in detail. Specifically, this review provides recent preclinical and clinical evidence that suggest GLP-1RAs preserve vessel health in part by preventing endothelial dysfunction, achieved primarily through the promotion of angiogenesis and inhibition of oxidative stress. These protective effects are in addition to the broad range of atherosclerotic processes GLP-1RAs target downstream of endothelial dysfunction, which include systemic inflammation, monocyte recruitment, proinflammatory macrophage and foam cell formation, vascular smooth muscle cell proliferation, and plaque development.
Collapse
Affiliation(s)
- Brady Park
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Ehab Bakbak
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Hwee Teoh
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Aishwarya Krishnaraj
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Fallon Dennis
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Quan
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Ori D Rotstein
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Division of General Surgery, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, Texas, United States
- Department of Medicine, University of Mississippi, Jackson, Mississippi, United States
| | - David A Hess
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Keenan Research Centre of Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Butler J, Shah SJ, Petrie MC, Borlaug BA, Abildstrøm SZ, Davies MJ, Hovingh GK, Kitzman DW, Møller DV, Verma S, Einfeldt MN, Lindegaard ML, Rasmussen S, Abhayaratna W, Ahmed FZ, Ben-Gal T, Chopra V, Ezekowitz JA, Fu M, Ito H, Lelonek M, Melenovský V, Merkely B, Núñez J, Perna E, Schou M, Senni M, Sharma K, van der Meer P, Von Lewinski D, Wolf D, Kosiborod MN. Semaglutide versus placebo in people with obesity-related heart failure with preserved ejection fraction: a pooled analysis of the STEP-HFpEF and STEP-HFpEF DM randomised trials. Lancet 2024; 403:1635-1648. [PMID: 38599221 PMCID: PMC11317105 DOI: 10.1016/s0140-6736(24)00469-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND In the STEP-HFpEF (NCT04788511) and STEP-HFpEF DM (NCT04916470) trials, the GLP-1 receptor agonist semaglutide improved symptoms, physical limitations, bodyweight, and exercise function in people with obesity-related heart failure with preserved ejection fraction. In this prespecified pooled analysis of the STEP-HFpEF and STEP-HFpEF DM trials, we aimed to provide a more definitive assessment of the effects of semaglutide across a range of outcomes and to test whether these effects were consistent across key patient subgroups. METHODS We conducted a prespecified pooled analysis of individual patient data from STEP-HFpEF and STEP-HFpEF DM, randomised, double-blind, placebo-controlled trials at 129 clinical research sites in 18 countries. In both trials, eligible participants were aged 18 years or older, had heart failure with a left ventricular ejection fraction of at least 45%, a BMI of at least 30 kg/m2, New York Heart Association class II-IV symptoms, and a Kansas City Cardiomyopathy Questionnaire Clinical Summary Score (KCCQ-CSS; a measure of heart failure-related symptoms and physical limitations) of less than 90 points. In STEP-HFpEF, people with diabetes or glycated haemoglobin A1c concentrations of at least 6·5% were excluded, whereas for inclusion in STEP-HFpEF DM participants had to have been diagnosed with type 2 diabetes at least 90 days before screening and to have an HbA1c of 10% or lower. In both trials, participants were randomly assigned to either 2·4 mg semaglutide once weekly or matched placebo for 52 weeks. The dual primary endpoints were change from baseline to week 52 in KCCQ-CSS and bodyweight in all randomly assigned participants. Confirmatory secondary endpoints included change from baseline to week 52 in 6-min walk distance, a hierarchical composite endpoint (all-cause death, heart failure events, and differences in changes in KCCQ-CSS and 6-min walk distance); and C-reactive protein (CRP) concentrations. Heterogeneity in treatment effects was assessed across subgroups of interest. We assessed safety in all participants who received at least one dose of study drug. FINDINGS Between March 19, 2021 and March 9, 2022, 529 people were randomly assigned in STEP-HFpEF, and between June 27, 2021 and Sept 2, 2022, 616 were randomly assigned in STEP-HFpEF DM. Overall, 1145 were included in our pooled analysis, 573 in the semaglutide group and 572 in the placebo group. Improvements in KCCQ-CSS and reductions in bodyweight between baseline and week 52 were significantly greater in the semaglutide group than in the placebo group (mean between-group difference for the change from baseline to week 52 in KCCQ-CSS 7·5 points [95% CI 5·3 to 9·8]; p<0·0001; mean between-group difference in bodyweight at week 52 -8·4% [-9·2 to -7·5]; p<0·0001). For the confirmatory secondary endpoints, 6-min walk distance (mean between-group difference at week 52 17·1 metres [9·2 to 25·0]) and the hierarchical composite endpoint (win ratio 1·65 [1·42 to 1·91]) were significantly improved, and CRP concentrations (treatment ratio 0·64 [0·56 to 0·72]) were significantly reduced, in the semaglutide group compared with the placebo group (p<0·0001 for all comparisons). For the dual primary endpoints, the efficacy of semaglutide was largely consistent across multiple subgroups, including those defined by age, race, sex, BMI, systolic blood pressure, baseline CRP, and left ventricular ejection fraction. 161 serious adverse events were reported in the semaglutide group compared with 301 in the placebo group. INTERPRETATION In this prespecified pooled analysis of the STEP-HFpEF and STEP-HFpEF DM trials, semaglutide was superior to placebo in improving heart failure-related symptoms and physical limitations, and reducing bodyweight in participants with obesity-related heart failure with preserved ejection fraction. These effects were largely consistent across patient demographic and clinical characteristics. Semaglutide was well tolerated. FUNDING Novo Nordisk.
Collapse
Affiliation(s)
- Javed Butler
- Baylor Scott & White Research Institute, Dallas, TX, USA; Department of Medicine, University of Mississippi, Jackson, MS, USA
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Melanie J Davies
- Diabetes Research Centre, University of Leicester, Leicester, UK; National Institute for Health and Care Research Leicester Biomedical Research Centre, Leicester, UK
| | | | - Dalane W Kitzman
- Department of Cardiovascular Medicine and Section on Geriatrics and Gerontology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Subodh Verma
- Division of Cardiac Surgery, Li Ka Shing Knowledge Institute of St Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | | | | | | | - Walter Abhayaratna
- College of Health and Medicine, Australian National University, Canberra, ACT, Australia
| | - Fozia Z Ahmed
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Tuvia Ben-Gal
- Heart Failure Unit, Department of Cardiology, Rabin Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vijay Chopra
- Max Super Speciality Hospital, Saket, New Delhi, India
| | | | - Michael Fu
- Section of Cardiology, Department of Medicine, Sahlgrenska University Hospital-Ostra, Gothenburg, Sweden
| | - Hiroshi Ito
- Department of General Internal Medicine 3, Kawasaki Medical School, Okayama, Japan
| | - Małgorzata Lelonek
- Department of Noninvasive Cardiology, Medical University of Lodz, Lodz, Poland
| | - Vojtěch Melenovský
- Institute for Clinical and Experimental Medicine-IKEM, Prague, Czech Republic
| | - Bela Merkely
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Julio Núñez
- Hospital Clínico Universitario de Valencia, INCLIVA, Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red Cardiovascular, Valencia, Spain
| | - Eduardo Perna
- Instituto de Cardiologia J F Cabral, Corrientes, Argentina
| | - Morten Schou
- Department of Cardiology, Herlev-Gentofte Hospital, Hellerup, Denmark; Department of Clinical Medicine, University of Copenhagen, Herlev, Denmark
| | - Michele Senni
- Azienda Socio Sanitaria Territorial Papa Giovanni XXIII, Bergamo, Italy
| | - Kavita Sharma
- Heart Failure & Cardiac Transplantation, Johns Hopkins University Heart Failure with Preserved Ejection Fraction Program, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Dennis Wolf
- Cardiology and Angiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mikhail N Kosiborod
- Department of Cardiovascular Disease, Saint Luke's Mid America Heart Institute, Kansas City, MO, USA; University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.
| |
Collapse
|
29
|
Fu WJ, Huo JL, Mao ZH, Pan SK, Liu DW, Liu ZS, Wu P, Gao ZX. Emerging role of antidiabetic drugs in cardiorenal protection. Front Pharmacol 2024; 15:1349069. [PMID: 38384297 PMCID: PMC10880452 DOI: 10.3389/fphar.2024.1349069] [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: 12/06/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
The global prevalence of diabetes mellitus (DM) has led to widespread multi-system damage, especially in cardiovascular and renal functions, heightening morbidity and mortality. Emerging antidiabetic drugs sodium-glucose cotransporter 2 inhibitors (SGLT2i), glucagon-like peptide-1 receptor agonists (GLP-1RAs), and dipeptidyl peptidase-4 inhibitors (DPP-4i) have demonstrated efficacy in preserving cardiac and renal function, both in type 2 diabetic and non-diabetic individuals. To understand the exact impact of these drugs on cardiorenal protection and underlying mechanisms, we conducted a comprehensive review of recent large-scale clinical trials and basic research focusing on SGLT2i, GLP-1RAs, and DPP-4i. Accumulating evidence highlights the diverse mechanisms including glucose-dependent and independent pathways, and revealing their potential cardiorenal protection in diabetic and non-diabetic cardiorenal disease. This review provides critical insights into the cardiorenal protective effects of SGLT2i, GLP-1RAs, and DPP-4i and underscores the importance of these medications in mitigating the progression of cardiovascular and renal complications, and their broader clinical implications beyond glycemic management.
Collapse
Affiliation(s)
- Wen-Jia Fu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Jin-Ling Huo
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zi-Hui Mao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shao-Kang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dong-Wei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhang-Suo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Peng Wu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhong-Xiuzi Gao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| |
Collapse
|
30
|
Maeder MT. Natriuretic peptides - Biomarker companions through thick and thin. Eur J Heart Fail 2024; 26:270-273. [PMID: 38287692 DOI: 10.1002/ejhf.3153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/31/2024] Open
Affiliation(s)
- Micha T Maeder
- Cardiology Department, Kantonsspital St. Gallen, St. Gallen, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
31
|
Kobak KA, Chiao YA. Semaglutide in Heart Failure With Preserved Ejection Fraction: Benefits Above and Beyond Weight Loss. JACC Basic Transl Sci 2023; 8:1315-1317. [PMID: 38094694 PMCID: PMC10714162 DOI: 10.1016/j.jacbts.2023.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
Affiliation(s)
- Kamil A. Kobak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| |
Collapse
|