1
|
Houbachi L, Walker PM, Fournel I, Ksiazek E, Petit JM, Cochet A, Leclercq T, Roger A, Simoneau I, Bouillet B, Guenancia C. Evolution of myocardial steatosis in high cardiovascular risk T2DM patients treated by GLP1 receptor agonists: LICAS study. Diabetes Res Clin Pract 2025; 221:112017. [PMID: 39900263 DOI: 10.1016/j.diabres.2025.112017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND We hypothesized that the reduction of intramyocardial fat content may be involved in the cardioprotective effect of glucagon-like peptide-1 receptor agonists (GLP1-RA) in patients with type 2 diabetes (T2D). Therefore, we aimed to evaluate the change in intramyocardial triglyceride content in T2D patients treated with GLP1-RA. METHODS This monocentric proof-of-concept cohort study included patients with unbalanced T2D prior to the introduction of GLP1-RA. Patients underwent cardiac magnetic resonance imaging (MRI) coupled with nuclear magnetic resonance (NMR) spectroscopy at baseline and six months after the introduction (M6) of a GLP1-RA to assess changes in intramyocardial triglyceride levels and morphological, functional, and cardiac tissue parameters. The relative delta (Δr) between baseline and M6 was calculated and analyzed by Student test or sign test. RESULTS Twenty-six patients (mean age = 62.2 ± 6.7 years, median HbA1c = 9.1 %) fulfilled inclusion criteria and had both NMR measures. Compared with baseline, relative intramyocardial triglyceride levels significantly decreased after six months of treatment (mean Δr = -26 % [95 %CI:-39; -13]p = 0.003), as well as glycated hemoglobin (HbA1c) (median Δr = -26 % [IQR:25], p < 0.0001), body mass index (BMI) (mean Δr = -6% [-9; -4], p < 0.0001) and left ventricular mass (mean Δr = -6 [-12; -1] p = 0.02). The relative evolution of intramyocardial triglyceride content was not correlated with the relative evolution of HbA1c (r = 0.10) and BMI (r = -0.02). CONCLUSIONS We demonstrate a significant reduction in intramyocardial triglyceride content in patients with T2D after six months of treatment with GLP1-RA. The lack of correlation with reductions in HbA1c and BMI suggests a specific effect of GLP1-RA on myocardial steatosis, which might contribute to their previously demonstrated cardiovascular benefits.
Collapse
Affiliation(s)
- Lina Houbachi
- Department of Endocrinology and Diabetology, CHU Dijon 21000 Dijon, France
| | - Paul Michael Walker
- Medical Imaging Department, CHU Dijon / ICMUB Laboratory, CNRS UMR 6302, University of Burgundy, France
| | - Isabelle Fournel
- CHU Dijon Bourgogne, INSERM, Université de Bourgogne, CIC 1432, Module Epidémiologique Clinique F21000 Dijon, France
| | - Elea Ksiazek
- CHU Dijon Bourgogne, INSERM, Université de Bourgogne, CIC 1432, Module Epidémiologique Clinique F21000 Dijon, France
| | - Jean-Michel Petit
- Department of Endocrinology and Diabetology, CHU Dijon 21000 Dijon, France; INSERM Research Center U1231, Team Padys 21000 Dijon, France; University of Burgundy 21000 Dijon, France
| | - Alexandre Cochet
- Medical Imaging Department, CHU Dijon / ICMUB Laboratory, CNRS UMR 6302, University of Burgundy, France
| | | | - Antoine Roger
- Cardiology Department, CHU Dijon 21000 Dijon, France
| | - Isabelle Simoneau
- Department of Endocrinology and Diabetology, CHU Dijon 21000 Dijon, France
| | - Benjamin Bouillet
- Department of Endocrinology and Diabetology, CHU Dijon 21000 Dijon, France; INSERM Research Center U1231, Team Padys 21000 Dijon, France; University of Burgundy 21000 Dijon, France
| | - Charles Guenancia
- Cardiology Department, CHU Dijon 21000 Dijon, France; Epidemiology of Cerebro- and Cardiovascular Diseases (EA 7460), UFR Science de Santé, Université de Bourgogne 21000 Dijon, France.
| |
Collapse
|
2
|
Tsai HH, Hsiao FC, Yu AL, Juang JH, Yu J, Chu PH. Empagliflozin Reduces High Glucose-Induced Cardiomyopathy in hiPSC-Derived Cardiomyocytes : Glucose-induced Lipotoxicity in hiPSC-Derived Cardiomyocytes. Stem Cell Rev Rep 2025:10.1007/s12015-024-10839-8. [PMID: 39841369 DOI: 10.1007/s12015-024-10839-8] [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] [Accepted: 12/26/2024] [Indexed: 01/23/2025]
Abstract
Human-induced pluripotent stem cell (hiPSC) technology has been applied in pathogenesis studies, drug screening, tissue engineering, and stem cell therapy, and patient-specific hiPSC-derived cardiomyocytes (hiPSC-CMs) have shown promise in disease modeling, including diabetic cardiomyopathy. High glucose (HG) treatment induces lipotoxicity in hiPSC-CMs, as evidenced by changes in cell size, beating rate, calcium handling, and lipid accumulation. Empagliflozin, an SGLT2 inhibitor, effectively mitigates the hypertrophic changes, abnormal calcium handling, and contractility impairment induced by HG. Glucose concentration influences SGLT2 expression in cardiomyocytes, highlighting its potential role in diabetic cardiomyopathy. These findings support the potential utility of hiPSC-CMs in studying diabetic cardiomyopathy and the efficacy of empagliflozin in ameliorating HG-induced cardiomyocyte dysfunction. Such research may advance developments in precision medicine and therapeutic interventions for patients with diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hsiu-Hui Tsai
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Fu-Chih Hsiao
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Pediatrics, University of California in San Diego, San Diego, CA, USA
| | - Jyuhn-Huarng Juang
- Division of Endocrinology and Metabolism, Department of Internal Medicine and Center for Tissue Engineering, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Pao-Hsien Chu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
3
|
McLean E, Roo CD, Maag A, Coble M, Cano J, Liu R. ERK1/2 Inhibition Alleviates Diabetic Cardiomyopathy by Suppressing Fatty Acid Metabolism. FRONT BIOSCI-LANDMRK 2025; 30:26700. [PMID: 39862096 DOI: 10.31083/fbl26700] [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: 09/24/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Diabetes mellitus is associated with morphological and functional impairment of the heart primarily due to lipid toxicity caused by increased fatty acid metabolism. Extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) have been implicated in the metabolism of fatty acids in the liver and skeletal muscles. However, their role in the heart in diabetes remains unclear. In this study, we tested our hypothesis that pharmacological inhibition of ERK1/2 alleviates cardiac remodeling in diabetic mice through a reduction in fatty acid metabolism. METHODS ERK1/2 phosphorylation in diabetes was determined both in vitro and in vivo. H9C2 cells were subjected to high glucose, high palmitic acid, or both high glucose and palmitic acid. db/db and streptozotocin (STZ)-induced diabetic mice were analyzed for ERK1/2 phosphorylation levels as well as the effects of U0126 treatment on cardiac remodeling. Administration of STZ and U0126 in mice was performed via intraperitoneal injection. Blood glucose levels in mice were measured using a glucometer. Mouse heart total RNAs were purified for reverse transcription. Real-time polymerase chain reaction (PCR) analysis of the messenger ribonucleic acid (mRNA) expression was performed for hypertrophy (ANF, BNP, and βMHC), fibrosis (Col3α1), and fatty acid metabolism genes (PPARα, CPT1A, and FACS). Interstitial fibrosis of the myocardium was analyzed using Masson's trichrome staining of the paraffin-embedded tissues. RESULTS ERK1/2 phosphorylation was significantly increased in diabetic conditions. Inhibition of ERK1/2 by U0126 in both streptozotocin-induced diabetic mice and db/db mice resulted in a significant reduction in the expression of genes associated with hypertrophy and fibrosis. In contrast, elevated phosphorylation of ERK1/2 in Dusp6/8 knockout (DKO) mice resulted in fibrosis. Mechanistically, ERK1/2 activation enhanced the expression of fatty acid metabolism genes PPARα, CPT1A, and FACS in the heart, which was reversed by U0126 treatment. CONCLUSION ERK1/2 are potential therapeutic targets for diabetic cardiomyopathy by modulating fatty acid metabolism in the heart.
Collapse
Affiliation(s)
- Erin McLean
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI 49401, USA
| | - Caroline De Roo
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI 49401, USA
| | - Annabel Maag
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI 49401, USA
| | - Megan Coble
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI 49401, USA
| | - Jefferson Cano
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI 49401, USA
| | - Ruijie Liu
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI 49401, USA
| |
Collapse
|
4
|
Roos PR, van den Burg EL, Schoonakker MP, van Peet PG, Numans ME, Pijl H, Westenberg JJM, Lamb HJ. Fasting-mimicking diet in type 2 diabetes reduces myocardial triglyceride content: A 12-month randomised controlled trial. Nutr Metab Cardiovasc Dis 2025:103860. [PMID: 39934050 DOI: 10.1016/j.numecd.2025.103860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND AND AIMS Type 2 diabetes is associated with a heightened risk of cardiovascular complications, including myocardial steatosis. Fasting-mimicking diets (FMDs) may mimic the metabolic benefits of fasting, while being less intensive than fasting. This study aims to investigate the effect of following an FMD program on myocardial triglyceride content (MTGC), as assessed by Magnetic Resonance Spectroscopy (MRS), in patients with type 2 diabetes. METHODS AND RESULTS 100 patients with type 2 diabetes, who used metformin as the only glucose-lowering drug or no medication were randomly assigned to either an FMD group or a control group. The FMD group received the FMD program for 5 consecutive days a month alongside usual care, while the control group received usual care only. Both groups underwent baseline, 6-months and 12-months examinations, including single voxel cardiac 1H-MRS to assess MTGC. N = 13 participants of the FMD and n = 13 of the control group had complete data at baseline and twelve month follow-up. The FMD group exhibited a significant reduction in MTGC over the twelve month period (-0.235 % MTGC, p = 0.027), while the control group saw no significant change (0.143 % MTGC, p = 0.236). The decrease of MTGC in the FMD group was statistically different (p = 0.018) from control. CONCLUSION Following an FMD program reduces MTGC, which indicates a favorable effect on cardiac metabolism and thereby may be an effective strategy to reduce the cardiovascular risk in patients with type 2 diabetes. CLINICAL TRIALS REGISTRATION NUMBER NCT03811587. TRIAL REGISTRATION ClinicalTrials.gov; NCT03811587, submitted January 13th, 2019.
Collapse
Affiliation(s)
- Paul R Roos
- Department of Radiology, Leiden University Medical Center (LUMC), the Netherlands.
| | - Elske L van den Burg
- Department of Public Health and Primary Care, Leiden University Medical Center, the Netherlands
| | - Marjolein P Schoonakker
- Department of Public Health and Primary Care, Leiden University Medical Center, the Netherlands
| | - Petra G van Peet
- Department of Public Health and Primary Care, Leiden University Medical Center, the Netherlands
| | - Mattijs E Numans
- Department of Public Health and Primary Care, Leiden University Medical Center, the Netherlands
| | - Hanno Pijl
- Department of Public Health and Primary Care, Leiden University Medical Center, the Netherlands; Department of Internal Medicine, Leiden University Medical Center, the Netherlands
| | - Jos J M Westenberg
- Department of Radiology, Leiden University Medical Center (LUMC), the Netherlands
| | - Hildo J Lamb
- Department of Radiology, Leiden University Medical Center (LUMC), the Netherlands
| |
Collapse
|
5
|
Ng YH, Koay YC, Marques FZ, Kaye DM, O’Sullivan JF. Leveraging metabolism for better outcomes in heart failure. Cardiovasc Res 2024; 120:1835-1850. [PMID: 39351766 PMCID: PMC11630082 DOI: 10.1093/cvr/cvae216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/26/2024] [Accepted: 08/07/2024] [Indexed: 12/11/2024] Open
Abstract
Whilst metabolic inflexibility and substrate constraint have been observed in heart failure for many years, their exact causal role remains controversial. In parallel, many of our fundamental assumptions about cardiac fuel use are now being challenged like never before. For example, the emergence of sodium-glucose cotransporter 2 inhibitor therapy as one of the four 'pillars' of heart failure therapy is causing a revisit of metabolism as a key mechanism and therapeutic target in heart failure. Improvements in the field of cardiac metabolomics will lead to a far more granular understanding of the mechanisms underpinning normal and abnormal human cardiac fuel use, an appreciation of drug action, and novel therapeutic strategies. Technological advances and expanding biorepositories offer exciting opportunities to elucidate the novel aspects of these metabolic mechanisms. Methodologic advances include comprehensive and accurate substrate quantitation such as metabolomics and stable-isotope fluxomics, improved access to arterio-venous blood samples across the heart to determine fuel consumption and energy conversion, high quality cardiac tissue biopsies, biochemical analytics, and informatics. Pairing these technologies with recent discoveries in epigenetic regulation, mitochondrial dynamics, and organ-microbiome metabolic crosstalk will garner critical mechanistic insights in heart failure. In this state-of-the-art review, we focus on new metabolic insights, with an eye on emerging metabolic strategies for heart failure. Our synthesis of the field will be valuable for a diverse audience with an interest in cardiac metabolism.
Collapse
Affiliation(s)
- Yann Huey Ng
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
| | - Yen Chin Koay
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, VIC 3800, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3800, Australia
- Victorian Heart Institute, Monash University, Melbourne, VIC 3800, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3800, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Monash-Alfred-Baker Centre for Cardiovascular Research, Monash University, Melbourne, VIC 3800, Australia
| | - John F O’Sullivan
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- Department of Medicine, Technische Univeristat Dresden, 01062 Dresden, Germany
| |
Collapse
|
6
|
Rizza V, Tondi L, Patti AM, Cecchi D, Lombardi M, Perone F, Ambrosetti M, Rizzo M, Cianflone D, Maranta F. Diabetic cardiomyopathy: pathophysiology, imaging assessment and therapeutical strategies. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2024; 23:200338. [PMID: 39734497 PMCID: PMC11681223 DOI: 10.1016/j.ijcrp.2024.200338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/14/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024]
Abstract
Diabetes mellitus (DM) is one of the most prevalent cardiovascular risk factors in the general population, being associated with high morbidity and socioeconomic burden. Diabetic cardiomyopathy (DCM) is a non-negligible complication of DM, whose pathophysiological fundaments are the altered cardiac metabolism, the hyperglycemia-triggered formation of advanced glycation end-products (AGEs) and the inflammatory milieu which are typical in diabetic patients. These metabolic abnormalities lead to cardiomyocytes apoptosis, interstitial fibrosis and mechanical cardiac dysfunction, which can be identified with non-invasive imaging techniques, like echocardiography and cardiac magnetic resonance. This review aims to: 1) describe the major imaging features of DCM; 2) highlight how early identification of DCM-related anatomical and functional remodeling might allow patients' therapy optimization and prognosis improvement.
Collapse
Affiliation(s)
| | - Lara Tondi
- Multimodality Cardiac Imaging Section, Policlinico San Donato, San Donato Milanese, Italy
- Postgraduate School of Radiology, University of Milan, Milan, Italy
| | - Angelo Maria Patti
- Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | | | - Massimo Lombardi
- Multimodality Cardiac Imaging Section, Policlinico San Donato, San Donato Milanese, Italy
| | - Francesco Perone
- Cardiac Rehabilitation Unit, Rehabilitation Clinic ‘Villa Delle Magnolie', Castel Morrone, Caserta, Italy
| | - Marco Ambrosetti
- Cardiovascular Rehabilitation Unit, ASST Crema, Santa Marta Hospital, Rivolta D'Adda, Italy
| | - Manfredi Rizzo
- Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Domenico Cianflone
- IRCCS Ospedale San Raffaele, Milan, Italy
- Cardiovascular Rehabilitation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Maranta
- IRCCS Ospedale San Raffaele, Milan, Italy
- Cardiovascular Rehabilitation Unit, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
7
|
Bode D, Pronto JRD, Schiattarella GG, Voigt N. Metabolic remodelling in atrial fibrillation: manifestations, mechanisms and clinical implications. Nat Rev Cardiol 2024; 21:682-700. [PMID: 38816507 DOI: 10.1038/s41569-024-01038-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 06/01/2024]
Abstract
Atrial fibrillation (AF) is a continually growing health-care burden that often presents together with metabolic disorders, including diabetes mellitus and obesity. Current treatments often fall short of preventing AF and its adverse outcomes. Accumulating evidence suggests that metabolic disturbances can promote the development of AF through structural and electrophysiological remodelling, but the underlying mechanisms that predispose an individual to AF are aetiology-dependent, thus emphasizing the need for tailored therapeutic strategies to treat AF that target an individual's metabolic profile. AF itself can induce changes in glucose, lipid and ketone metabolism, mitochondrial function and myofibrillar energetics (as part of a process referred to as 'metabolic remodelling'), which can all contribute to atrial dysfunction. In this Review, we discuss our current understanding of AF in the setting of metabolic disorders, as well as changes in atrial metabolism that are relevant to the development of AF. We also describe the potential of available and emerging treatment strategies to target metabolic remodelling in the setting of AF and highlight key questions and challenges that need to be addressed to improve outcomes in these patients.
Collapse
Affiliation(s)
- David Bode
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
8
|
Chakraborty P, Nattel S, Nanthakumar K, Connelly KA, Husain M, Po SS, Ha ACT. Sudden cardiac death due to ventricular arrhythmia in diabetes mellitus: A bench to bedside review. Heart Rhythm 2024; 21:1827-1837. [PMID: 38848857 DOI: 10.1016/j.hrthm.2024.05.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024]
Abstract
Diabetes mellitus (DM) confers an increased risk of sudden cardiac death (SCD) independent of its associated cardiovascular comorbidities. DM induces adverse structural, electrophysiologic, and autonomic cardiac remodeling that can increase one's risk of ventricular arrhythmias and SCD. Although glycemic control and prevention of microvascular and macrovascular complications are cornerstones in the management of DM, they are not adequate for the prevention of SCD. In this narrative review, we describe the contribution of DM to the pathophysiologic mechanism of SCD beyond its role in atherosclerotic cardiovascular disease and heart failure. On the basis of this pathophysiologic framework, we outline potential preventive and therapeutic strategies to mitigate the risk of SCD in this population of high-risk patients.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Kumaraswamy Nanthakumar
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kim A Connelly
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Mansoor Husain
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Sunny S Po
- Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andrew C T Ha
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Seferović PM, Paulus WJ, Rosano G, Polovina M, Petrie MC, Jhund PS, Tschöpe C, Sattar N, Piepoli M, Papp Z, Standl E, Mamas MA, Valensi P, Linhart A, Lalić N, Ceriello A, Döhner W, Ristić A, Milinković I, Seferović J, Cosentino F, Metra M, Coats AJS. Diabetic myocardial disorder. A clinical consensus statement of the Heart Failure Association of the ESC and the ESC Working Group on Myocardial & Pericardial Diseases. Eur J Heart Fail 2024; 26:1893-1903. [PMID: 38896048 DOI: 10.1002/ejhf.3347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024] Open
Abstract
The association between type 2 diabetes mellitus (T2DM) and heart failure (HF) has been firmly established; however, the entity of diabetic myocardial disorder (previously called diabetic cardiomyopathy) remains a matter of debate. Diabetic myocardial disorder was originally described as the occurrence of myocardial structural/functional abnormalities associated with T2DM in the absence of coronary heart disease, hypertension and/or obesity. However, supporting evidence has been derived from experimental and small clinical studies. Only a minority of T2DM patients are recognized as having this condition in the absence of contributing factors, thereby limiting its clinical utility. Therefore, this concept is increasingly being viewed along the evolving HF trajectory, where patients with T2DM and asymptomatic structural/functional cardiac abnormalities could be considered as having pre-HF. The importance of recognizing this stage has gained interest due to the potential for current treatments to halt or delay the progression to overt HF in some patients. This document is an expert consensus statement of the Heart Failure Association of the ESC and the ESC Working Group on Myocardial & Pericardial Diseases. It summarizes contemporary understanding of the association between T2DM and HF and discuses current knowledge and uncertainties about diabetic myocardial disorder that deserve future research. It also proposes a new definition, whereby diabetic myocardial disorder is defined as systolic and/or diastolic myocardial dysfunction in the presence of diabetes. Diabetes is rarely exclusively responsible for myocardial dysfunction, but usually acts in association with obesity, arterial hypertension, chronic kidney disease and/or coronary artery disease, causing additive myocardial impairment.
Collapse
Affiliation(s)
- Petar M Seferović
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Walter J Paulus
- Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Giuseppe Rosano
- Department of Human Sciences and Promotion of Quality of Life, San Raffaele Open University of Rome, Rome, Italy
- Cardiology, San Raffaele Cassino Hospital, Cassino, Italy
| | - Marija Polovina
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Department of Cardiology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Pardeep S Jhund
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Carsten Tschöpe
- Berlin Institute of Health at Charité - Center for Regenerative Therapies, Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology (CVK) and German Centre for Cardiovascular Research (DZHK)- Partner Site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Massimo Piepoli
- Cardiology University Department, RCCS Policlinico San Donato, San Donato Milanese, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eberhard Standl
- Diabetes Research Group e.V. at Munich Helmholtz Center, Munich, Germany
| | - Mamas A Mamas
- Cardiovascular Research Group, Keele University, Keele, UK
| | - Paul Valensi
- Polyclinique d'Aubervilliers, Aubervilliers, and Paris Nord University, Bobigny, France
| | - Ales Linhart
- Department of Internal Medicine, School of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Nebojša Lalić
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
- Department of Endocrinology, University Clinical Centre of Serbia, Belgrade, Serbia
| | | | - Wolfram Döhner
- Berlin Institute of Health at Charité - Center for Regenerative Therapies, Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité, Department of Cardiology (CVK) and German Centre for Cardiovascular Research (DZHK)- Partner Site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin (CSB), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Arsen Ristić
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Department of Cardiology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Ivan Milinković
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Department of Cardiology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Jelena Seferović
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Department of Endocrinology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Francesco Cosentino
- Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Marco Metra
- Institute of Cardiology, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | | |
Collapse
|
10
|
Ke J, Pan J, Lin H, Huang S, Zhang J, Wang C, Chang ACY, Gu J. Targeting Rab7-Rilp Mediated Microlipophagy Alleviates Lipid Toxicity in Diabetic Cardiomyopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401676. [PMID: 38837607 PMCID: PMC11304244 DOI: 10.1002/advs.202401676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/25/2024] [Indexed: 06/07/2024]
Abstract
Diabetic cardiomyopathy (DbCM) is characterized by diastolic dysfunction, which progresses into heart failure and aberrant electrophysiology in diabetic patients. Dyslipidemia in type 2 diabetic patients leads to the accumulation of lipid droplets (LDs) in cardiomyocytes and results in lipid toxicity which has been suggested to drive DbCM. It is aimed to explore potential pathways that may boost LDs degradation in DbCM and restore cardiac function. LDs accumulation resulted in an increase in lipid toxicity in DbCM hearts is confirmed. Microlipophagy pathway, rather than traditional macrolipophagy, is activated in DbCM hearts. RNA-Seq data and Rab7-CKO mice implicate that Rab7 is a major modulator of the microlipophagy pathway. Mechanistically, Rab7 is phosphorylated at Tyrosine 183, which allows the recruitment of Rab-interacting lysosome protein (Rilp) to proceed LDs degradation by lysosome. Treating DbCM mice with Rab7 activator ML-098 enhanced Rilp level and rescued the observed cardiac dysfunction. Overall, Rab7-Rilp-mediated microlipophagy may be a promising target in the treatment of lipid toxicity in DbCM is suggested.
Collapse
Affiliation(s)
- Jiahan Ke
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200001China
| | - Jianan Pan
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200001China
| | - Hao Lin
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200001China
| | - Shuying Huang
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200001China
| | - Junfeng Zhang
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200001China
| | - Changqian Wang
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200001China
| | - Alex Chia Yu Chang
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200001China
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200120China
| | - Jun Gu
- Department of CardiologyShanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghai200001China
| |
Collapse
|
11
|
Bornstein MR, Tian R, Arany Z. Human cardiac metabolism. Cell Metab 2024; 36:1456-1481. [PMID: 38959861 PMCID: PMC11290709 DOI: 10.1016/j.cmet.2024.06.003] [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: 01/06/2024] [Revised: 04/12/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024]
Abstract
The heart is the most metabolically active organ in the human body, and cardiac metabolism has been studied for decades. However, the bulk of studies have focused on animal models. The objective of this review is to summarize specifically what is known about cardiac metabolism in humans. Techniques available to study human cardiac metabolism are first discussed, followed by a review of human cardiac metabolism in health and in heart failure. Mechanistic insights, where available, are reviewed, and the evidence for the contribution of metabolic insufficiency to heart failure, as well as past and current attempts at metabolism-based therapies, is also discussed.
Collapse
Affiliation(s)
- Marc R Bornstein
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Zoltan Arany
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Plötz T, Lenzen S. Mechanisms of lipotoxicity-induced dysfunction and death of human pancreatic beta cells under obesity and type 2 diabetes conditions. Obes Rev 2024; 25:e13703. [PMID: 38327101 DOI: 10.1111/obr.13703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 12/06/2023] [Accepted: 12/20/2023] [Indexed: 02/09/2024]
Abstract
The term "pancreatic beta-cell lipotoxicity" refers to the detrimental effects of free fatty acids (FFAs) on a wide variety of cellular functions. Basic research in the field has primarily analyzed the effects of palmitic acid and oleic acid. The focus on these two physiological FFAs, however, ignores differences in chain length and degree of saturation. In order to gain a comprehensive understanding of the lipotoxic mechanisms, a wide range of structurally related FFAs should be investigated. Structure-activity relationship analyses of FFAs in the human EndoC-βH1 beta-cell line have provided a deep insight into the mechanisms of beta-cell lipotoxicity. This review focuses on the effects of a wide range of FFAs with crucial structural determinants for the development of lipotoxicity in human beta cells and documents an association between increased triglyceride stores in obesity and in type 2 diabetes.
Collapse
Affiliation(s)
- Thomas Plötz
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Sigurd Lenzen
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Institute of Experimental Diabetes Research, Hannover Medical School, Hannover, Germany
| |
Collapse
|
13
|
Ye W, Han K, Xie M, Li S, Chen G, Wang Y, Li T. Mitochondrial energy metabolism in diabetic cardiomyopathy: Physiological adaption, pathogenesis, and therapeutic targets. Chin Med J (Engl) 2024; 137:936-948. [PMID: 38527931 PMCID: PMC11046025 DOI: 10.1097/cm9.0000000000003075] [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: 07/08/2023] [Indexed: 03/27/2024] Open
Abstract
ABSTRACT Diabetic cardiomyopathy is defined as abnormal structure and function of the heart in the setting of diabetes, which could eventually develop heart failure and leads to the death of the patients. Although blood glucose control and medications to heart failure show beneficial effects on this disease, there is currently no specific treatment for diabetic cardiomyopathy. Over the past few decades, the pathophysiology of diabetic cardiomyopathy has been extensively studied, and an increasing number of studies pinpoint that impaired mitochondrial energy metabolism is a key mediator as well as a therapeutic target. In this review, we summarize the latest research in the field of diabetic cardiomyopathy, focusing on mitochondrial damage and adaptation, altered energy substrates, and potential therapeutic targets. A better understanding of the mitochondrial energy metabolism in diabetic cardiomyopathy may help to gain more mechanistic insights and generate more precise mitochondria-oriented therapies to treat this disease.
Collapse
Affiliation(s)
- Wanlin Ye
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kun Han
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan 610041, China
| | - Maodi Xie
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Sheyu Li
- Department of Endocrinology and Metabolism, Division of Guideline and Rapid Recommendation, Cochrane China Center, MAGIC China Center, Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Guo Chen
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanyan Wang
- Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondria and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
14
|
Nakamura M. Lipotoxicity as a therapeutic target in obesity and diabetic cardiomyopathy. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12568. [PMID: 38706718 PMCID: PMC11066298 DOI: 10.3389/jpps.2024.12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
Unhealthy sources of fats, ultra-processed foods with added sugars, and a sedentary lifestyle make humans more susceptible to developing overweight and obesity. While lipids constitute an integral component of the organism, excessive and abnormal lipid accumulation that exceeds the storage capacity of lipid droplets disrupts the intracellular composition of fatty acids and results in the release of deleterious lipid species, thereby giving rise to a pathological state termed lipotoxicity. This condition induces endoplasmic reticulum stress, mitochondrial dysfunction, inflammatory responses, and cell death. Recent advances in omics technologies and analytical methodologies and clinical research have provided novel insights into the mechanisms of lipotoxicity, including gut dysbiosis, epigenetic and epitranscriptomic modifications, dysfunction of lipid droplets, post-translational modifications, and altered membrane lipid composition. In this review, we discuss the recent knowledge on the mechanisms underlying the development of lipotoxicity and lipotoxic cardiometabolic disease in obesity, with a particular focus on lipotoxic and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, United States
| |
Collapse
|
15
|
Chen S, Chen C, Zheng L, Cheng W, Bu X, Liu Z. Assessment of new-onset heart failure prediction in a diabetic population using left ventricular global strain: a prospective cohort study based on UK Biobank. Front Endocrinol (Lausanne) 2024; 15:1365169. [PMID: 38628588 PMCID: PMC11018882 DOI: 10.3389/fendo.2024.1365169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Background Impaired glucose utilization influences myocardial contractile function. However, the prognostic importance of left ventricular global radial strain (LV-GRS), left ventricular global circumferential strain (LV-GCS), and left ventricular global longitudinal strain (LV-GLS) in predicting new-onset heart failure (HF) in a population with diabetes is unclear. Methods The study design is prospective cohort from the UK Biobank. Totally 37,899 participants had a complete data of cardiac magnetic resonance (CMR), of which 940 patients with diabetes were included, and all the participants completed follow-up. LV-GRS, LV-GCS, and LV-GLS were measured by completely automated CMR with tissue tagging. Cox proportional hazards regression analysis and C-index was performed to evaluate the association between the strain parameters and the new-onset HF in patients suffering from diabetes. Results The average age of the 940 participants was 57.67 ± 6.97 years, with males comprising 66.4% of the overall population. With an average follow-up period of 166.82 ± 15.26 months, 35 (3.72%) patients reached the endpoint (emergence of new-onset HF). Significant associations were found for the three strain parameters and the new-onset HF (LV-GRS-hazard ratio [HR]: 0.946, 95% CI: 0.916-0.976; LV-GCS-HR: 1.162, 95% CI: 1.086-1.244; LV-GCS-HR: 1.181, 95% CI: 1.082-1.289). LV-GRS, LV-GCS, and LV-GLS were closely related to the related indicators to HF, and showed a high relationship to new-onset HF in individuals with diabetes at 5 and 10 years: LV-GRS: 0.75 (95% CI, 0.41-0.94) and 0.76 (95% CI, 0.44-0.98), respectively; LV-GCS: 0.80 (95% CI, 0.50-0.96) and 0.75 (95% CI, 0.41-0.98), respectively; LV-GLS: 0.72 (95% CI, 0.40-0.93) and 0.76 (95% CI, 0.48-0.97), respectively. In addition, age, sex, body mass index (BMI), and presence of hypertension or coronary artery disease (CAD) made no impacts on the association between the global strain parameters and the incidence of HF. Conclusion LV-GRS, LV-GCS, and LV-GLS is significantly related to new-onset HF in patients with diabetes at 5 and 10 years.
Collapse
Affiliation(s)
- Siwei Chen
- Department of Cardiovascular Medicine, The Third Hospital of Nanchang, Jiangxi, China
| | - Cong Chen
- Department of Cardiology, Zaozhuang Municipal Hospital, Zaozhuang, China
| | - Longxuan Zheng
- Department of Cardiology, The Fifth People’s Hospital of Huai’an, The Affiliated Huai’an Hospital of Yangzhou University, Huai’an, China
| | - Wenke Cheng
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Xiancong Bu
- Department of Neurology, Zaozhuang Municipal Hospital, Zaozhuang, China
| | - Zhou Liu
- Department of Geriatric Medicine/Cardiology, The Fifth People’s Hospital of Huai’an, The Affiliated Huai’an Hospital of Yangzhou University, Huai’an, China
| |
Collapse
|
16
|
Zeng Y, Li Y, Jiang W, Hou N. Molecular mechanisms of metabolic dysregulation in diabetic cardiomyopathy. Front Cardiovasc Med 2024; 11:1375400. [PMID: 38596692 PMCID: PMC11003275 DOI: 10.3389/fcvm.2024.1375400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Diabetic cardiomyopathy (DCM), one of the most serious complications of diabetes mellitus, has become recognized as a cardiometabolic disease. In normoxic conditions, the majority of the ATP production (>95%) required for heart beating comes from mitochondrial oxidative phosphorylation of fatty acids (FAs) and glucose, with the remaining portion coming from a variety of sources, including fructose, lactate, ketone bodies (KB) and branched chain amino acids (BCAA). Increased FA intake and decreased utilization of glucose and lactic acid were observed in the diabetic hearts of animal models and diabetic patients. Moreover, the polyol pathway is activated, and fructose metabolism is enhanced. The use of ketones as energy sources in human diabetic hearts also increases significantly. Furthermore, elevated BCAA levels and impaired BCAA metabolism were observed in the hearts of diabetic mice and patients. The shift in energy substrate preference in diabetic hearts results in increased oxygen consumption and impaired oxidative phosphorylation, leading to diabetic cardiomyopathy. However, the precise mechanisms by which impaired myocardial metabolic alterations result in diabetes mellitus cardiac disease are not fully understood. Therefore, this review focuses on the molecular mechanisms involved in alterations of myocardial energy metabolism. It not only adds more molecular targets for the diagnosis and treatment, but also provides an experimental foundation for screening novel therapeutic agents for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yue Zeng
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Yilang Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Wenyue Jiang
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Ning Hou
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
17
|
Kotha S, Plein S, Greenwood JP, Levelt E. Role of epicardial adipose tissue in diabetic cardiomyopathy through the lens of cardiovascular magnetic resonance imaging - a narrative review. Ther Adv Endocrinol Metab 2024; 15:20420188241229540. [PMID: 38476217 PMCID: PMC10929063 DOI: 10.1177/20420188241229540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/14/2024] [Indexed: 03/14/2024] Open
Abstract
Accumulating evidence suggests that ectopic/visceral adiposity may play a key role in the pathogenesis of nonischaemic cardiovascular diseases associated with type 2 diabetes. Epicardial adipose tissue (EAT) is a complex visceral fat depot, covering 80% of the cardiac surface with anatomical and functional contiguity to the myocardium and coronary arteries. EAT interacts with the biology of the underlying myocardium by secreting a wide range of adipokines. Magnetic resonance imaging (MRI) is the reference modality for structural and functional imaging of the heart. The technique is now also emerging as the reference imaging modality for EAT quantification. With this narrative review, we (a) surveyed contemporary clinical studies that utilized cardiovascular MRI to characterize EAT (studies published 2010-2023); (b) listed the clinical trials monitoring the response to treatment in EAT size as well as myocardial functional and structural parameters and (c) discussed the potential pathophysiological role of EAT in the development of diabetic cardiomyopathy. We concluded that increased EAT quantity and its inflammatory phenotype correlate with early signs of left ventricle dysfunction and may have a role in the pathogenesis of cardiac disease in diabetes with and without coronary artery disease.
Collapse
Affiliation(s)
- Sindhoora Kotha
- Department of Biomedical Imaging Science, Multidisciplinary Cardiovascular Research Centre, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sven Plein
- Department of Biomedical Imaging Science, Multidisciplinary Cardiovascular Research Centre, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - John P. Greenwood
- Department of Biomedical Imaging Science, Multidisciplinary Cardiovascular Research Centre, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Eylem Levelt
- Department of Biomedical Imaging Science, Multidisciplinary Cardiovascular Research Centre, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK
| |
Collapse
|
18
|
Girona J, Soler O, Samino S, Junza A, Martínez-Micaelo N, García-Altares M, Ràfols P, Esteban Y, Yanes O, Correig X, Masana L, Rodríguez-Calvo R. Lipidomics Reveals Myocardial Lipid Composition in a Murine Model of Insulin Resistance Induced by a High-Fat Diet. Int J Mol Sci 2024; 25:2702. [PMID: 38473949 DOI: 10.3390/ijms25052702] [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: 01/29/2024] [Revised: 02/18/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Ectopic fat accumulation in non-adipose tissues is closely related to diabetes-related myocardial dysfunction. Nevertheless, the complete picture of the lipid metabolites involved in the metabolic-related myocardial alterations is not fully characterized. The aim of this study was to characterize the specific lipid profile in hearts in an animal model of obesity/insulin resistance induced by a high-fat diet (HFD). The cardiac lipidome profiles were assessed via liquid chromatography-mass spectrometry (LC-MS)/MS-MS and laser desorption/ionization-mass spectrometry (LDI-MS) tissue imaging in hearts from C57BL/6J mice fed with an HFD or standard-diet (STD) for 12 weeks. Targeted lipidome analysis identified a total of 63 lipids (i.e., 48 triacylglycerols (TG), 5 diacylglycerols (DG), 1 sphingomyelin (SM), 3 phosphatidylcholines (PC), 1 DihydroPC, and 5 carnitines) modified in hearts from HFD-fed mice compared to animals fed with STD. Whereas most of the TG were up-regulated in hearts from animals fed with an HFD, most of the carnitines were down-regulated, thereby suggesting a reduction in the mitochondrial β-oxidation. Roughly 30% of the identified metabolites were oxidated, pointing to an increase in lipid peroxidation. Cardiac lipidome was associated with a specific biochemical profile and a specific liver TG pattern. Overall, our study reveals a specific cardiac lipid fingerprint associated with metabolic alterations induced by HFD.
Collapse
Affiliation(s)
- Josefa Girona
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, "Sant Joan" University Hospital, Institut de Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, 43204 Reus, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Oria Soler
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, "Sant Joan" University Hospital, Institut de Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, 43204 Reus, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Sara Samino
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, 43002 Tarragona, Spain
| | - Alexandra Junza
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, 43002 Tarragona, Spain
| | - Neus Martínez-Micaelo
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, "Sant Joan" University Hospital, Institut de Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, 43204 Reus, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
| | - María García-Altares
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, 43002 Tarragona, Spain
| | - Pere Ràfols
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, 43002 Tarragona, Spain
| | - Yaiza Esteban
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, "Sant Joan" University Hospital, Institut de Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, 43204 Reus, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Oscar Yanes
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, 43002 Tarragona, Spain
| | - Xavier Correig
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, 43002 Tarragona, Spain
| | - Lluís Masana
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, "Sant Joan" University Hospital, Institut de Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, 43204 Reus, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Ricardo Rodríguez-Calvo
- Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, "Sant Joan" University Hospital, Institut de Investigació Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, 43204 Reus, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
19
|
Wang H, Shen M, Shu X, Guo B, Jia T, Feng J, Lu Z, Chen Y, Lin J, Liu Y, Zhang J, Zhang X, Sun D. Cardiac Metabolism, Reprogramming, and Diseases. J Cardiovasc Transl Res 2024; 17:71-84. [PMID: 37668897 DOI: 10.1007/s12265-023-10432-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
Cardiovascular diseases (CVD) account for the largest bulk of deaths worldwide, posing a massive burden on societies and the global healthcare system. Besides, the incidence and prevalence of these diseases are on the rise, demanding imminent action to revert this trend. Cardiovascular pathogenesis harbors a variety of molecular and cellular mechanisms among which dysregulated metabolism is of significant importance and may even proceed other mechanisms. The healthy heart metabolism primarily relies on fatty acids for the ultimate production of energy through oxidative phosphorylation in mitochondria. Other metabolites such as glucose, amino acids, and ketone bodies come next. Under pathological conditions, there is a shift in metabolic pathways and the preference of metabolites, termed metabolic remodeling or reprogramming. In this review, we aim to summarize cardiovascular metabolism and remodeling in different subsets of CVD to come up with a new paradigm for understanding and treatment of these diseases.
Collapse
Affiliation(s)
- Haichang Wang
- Heart Hospital, Xi'an International Medical Center, Xi'an, China
| | - Min Shen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Xiaofei Shu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Baolin Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Tengfei Jia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jiaxu Feng
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Zuocheng Lu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yanyan Chen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yue Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Xuan Zhang
- Institute for Hospital Management Research, Chinese PLA General Hospital, Beijing, China.
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
20
|
Oneglia AP, Szczepaniak LS, Zaha VG, Nelson MD. Myocardial steatosis across the spectrum of human health and disease. Exp Physiol 2024; 109:202-213. [PMID: 38063136 PMCID: PMC10841709 DOI: 10.1113/ep091566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/15/2023] [Indexed: 02/02/2024]
Abstract
Preclinical data strongly suggest that myocardial steatosis leads to adverse cardiac remodelling and left ventricular dysfunction. Using 1 H cardiac magnetic resonance spectroscopy, similar observations have been made across the spectrum of health and disease. The purpose of this brief review is to summarize these recent observations. We provide a brief overview of the determinants of myocardial triglyceride accumulation, summarize the current evidence that myocardial steatosis contributes to cardiac dysfunction, and identify opportunities for further research.
Collapse
Affiliation(s)
- Andrew P. Oneglia
- Applied Physiology and Advanced Imaging Laboratory, Department of Kinesiology, College of Nursing and Health InnovationUniversity of Texas at ArlingtonArlingtonTexasUSA
| | | | - Vlad G. Zaha
- Division of Cardiology, Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Advanced Imaging Research CenterUniversity of Texas Southwestern Medical CenterArlingtonTexasUSA
| | - Michael D. Nelson
- Applied Physiology and Advanced Imaging Laboratory, Department of Kinesiology, College of Nursing and Health InnovationUniversity of Texas at ArlingtonArlingtonTexasUSA
- Clinical Imaging Research CenterUniversity of Texas at ArlingtonArlingtonTexasUSA
- Center for Healthy Living and LongevityUniversity of Texas at ArlingtonArlingtonTexasUSA
| |
Collapse
|
21
|
Ying Q, Xu Y, Zhang Z, Cai L, Zhao Y, Jin L. Gestational diabetes mellitus and risk of long-term all-cause and cardiac mortality: a prospective cohort study. Cardiovasc Diabetol 2024; 23:47. [PMID: 38302966 PMCID: PMC10835835 DOI: 10.1186/s12933-024-02131-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/10/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND To investigate the association between gestational diabetes mellitus (GDM) without subsequent overt diabetes and long-term all-cause and cardiac mortality. METHODS This prospective cohort study included 10,327 women (weighted population: 132,332,187) with a pregnancy history from the National Health and Nutrition Examination Survey (2007 to 2018). Participants were divided into three groups (GDM alone, overt diabetes, and no diabetes). Mortality data was linked from the National Death Index up to December 31, 2019. Multivariable Cox regression analysis was performed to examine the association between GDM alone and overt diabetes with all-cause mortality and cardiac mortality. Data analysis was performed from October 2022 to April 2023. RESULTS Among the participants, 510 (weighted 5.3%) had GDM alone and 1862 (weighted 14.1%) had overt diabetes. Over a median follow-up period of 6.7 years (69,063 person-years), there were 758 deaths. The GDM group did not show an increased risk of all-cause mortality (hazard ratio [HR] 0.67; 95% CI, 0.25-1.84), while the overt diabetes group had a significantly higher risk (HR 1.95; 95% CI, 1.62-2.35). Similarly, the GDM group did not exhibit an elevated risk of cardiac mortality (HR 1.48; 95% CI, 0.50-4.39), whereas the overt diabetes group had a significantly higher risk (HR 2.37; 95% CI, 1.69-3.32). Furthermore, sensitivity analysis focusing on women aged 50 or above showed that the HR of GDM history for all-cause mortality was 1.14 (95% CI, 0.33-3.95) and the HR for cardiac mortality was 1.74 (95% CI, 0.49-6.20). CONCLUSIONS GDM alone was not associated with an increased risk of all-cause and cardiac mortality, while overt diabetes was significantly associated with both types of mortality.
Collapse
Affiliation(s)
- Qian Ying
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yao Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ziyi Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Luyi Cai
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yan Zhao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Liping Jin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China.
| |
Collapse
|
22
|
Dattani A, Singh A, McCann GP, Gulsin GS. Myocardial Calcium Handling in Type 2 Diabetes: A Novel Therapeutic Target. J Cardiovasc Dev Dis 2023; 11:12. [PMID: 38248882 PMCID: PMC10817027 DOI: 10.3390/jcdd11010012] [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: 11/06/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Type 2 diabetes (T2D) is a multisystem disease with rapidly increasing global prevalence. Heart failure has emerged as a major complication of T2D. Dysregulated myocardial calcium handling is evident in the failing heart and this may be a key driver of cardiomyopathy in T2D, but until recently this has only been demonstrated in animal models. In this review, we describe the physiological concepts behind calcium handling within the cardiomyocyte and the application of novel imaging techniques for the quantification of myocardial calcium uptake. We take an in-depth look at the evidence for the impairment of calcium handling in T2D using pre-clinical models as well as in vivo studies, following which we discuss potential novel therapeutic approaches targeting dysregulated myocardial calcium handling in T2D.
Collapse
Affiliation(s)
- Abhishek Dattani
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Leicester LE3 9QP, UK; (A.S.); (G.P.M.); (G.S.G.)
| | | | | | | |
Collapse
|
23
|
Ritterhoff J, Tian R. Metabolic mechanisms in physiological and pathological cardiac hypertrophy: new paradigms and challenges. Nat Rev Cardiol 2023; 20:812-829. [PMID: 37237146 DOI: 10.1038/s41569-023-00887-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 05/28/2023]
Abstract
Cardiac metabolism is vital for heart function. Given that cardiac contraction requires a continuous supply of ATP in large quantities, the role of fuel metabolism in the heart has been mostly considered from the perspective of energy production. However, the consequence of metabolic remodelling in the failing heart is not limited to a compromised energy supply. The rewired metabolic network generates metabolites that can directly regulate signalling cascades, protein function, gene transcription and epigenetic modifications, thereby affecting the overall stress response of the heart. In addition, metabolic changes in both cardiomyocytes and non-cardiomyocytes contribute to the development of cardiac pathologies. In this Review, we first summarize how energy metabolism is altered in cardiac hypertrophy and heart failure of different aetiologies, followed by a discussion of emerging concepts in cardiac metabolic remodelling, that is, the non-energy-generating function of metabolism. We highlight challenges and open questions in these areas and finish with a brief perspective on how mechanistic research can be translated into therapies for heart failure.
Collapse
Affiliation(s)
- Julia Ritterhoff
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany.
- Mitochondria and Metabolism Center, Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
24
|
Akhtar MS, Alavudeen SS, Raza A, Imam MT, Almalki ZS, Tabassum F, Iqbal MJ. Current understanding of structural and molecular changes in diabetic cardiomyopathy. Life Sci 2023; 332:122087. [PMID: 37714373 DOI: 10.1016/j.lfs.2023.122087] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Diabetic Mellitus has been characterized as the most prevalent disease throughout the globe associated with the serious morbidity and mortality of vital organs. Cardiomyopathy is the major leading complication of diabetes and within this, myocardial dysfunction or failure is the leading cause of the emergency hospital admission. The review is aimed to comprehend the perspectives associated with diabetes-induced cardiovascular complications. The data was collected from several electronic databases such as Google Scholar, Science Direct, ACS publication, PubMed, Springer, etc. using the keywords such as diabetes and its associated complication, the prevalence of diabetes, the anatomical and physiological mechanism of diabetes-induced cardiomyopathy, the molecular mechanism of diabetes-induced cardiomyopathy, oxidative stress, and inflammatory stress, etc. The collected scientific data was screened by different experts based on the inclusion and exclusion criteria of the study. This review findings revealed that diabetes is associated with inefficient substrate utilization, inability to increase glucose metabolism and advanced glycation end products within the diabetic heart resulting in mitochondrial uncoupling, glucotoxicity, lipotoxicity, and initially subclinical cardiac dysfunction and finally in overt heart failure. Furthermore, several factors such as hypertension, overexpression of renin angiotensin system, hypertrophic obesity, etc. have been seen as majorly associated with cardiomyopathy. The molecular examination showed biochemical disability and generation of the varieties of free radicals and inflammatory cytokines and becomes are the substantial causes of cardiomyopathy. This review provides a better understanding of the involved pathophysiology and offers an open platform for discussing and targeting therapy in alleviating diabetes-induced early heart failure or cardiomyopathy.
Collapse
Affiliation(s)
- Md Sayeed Akhtar
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Al-Fara, Abha 62223, Saudi Arabia.
| | - Sirajudeen S Alavudeen
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Al-Fara, Abha 62223, Saudi Arabia
| | - Asif Raza
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, Penn State Milton S. Hershey Medical Center, 500 University Drive, Hershey, PA 17033, USA
| | - Mohammad Tarique Imam
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 16273, Saudi Arabia
| | - Ziad Saeed Almalki
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 16273, Saudi Arabia
| | - Fauzia Tabassum
- Department of Pharmacology, College of Dentistry and Pharmacy, Buraydah Private College, Al Qassim 51418, Saudi Arabia; Department of Pharmacology, Vision College, Ishbilia, Riyadh 13226-3830, Saudi Arabia
| | - Mir Javid Iqbal
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
25
|
Scisciola L, Chianese U, Caponigro V, Basilicata MG, Salviati E, Altucci L, Campiglia P, Paolisso G, Barbieri M, Benedetti R, Sommella E. Multi-omics analysis reveals attenuation of cellular stress by empagliflozin in high glucose-treated human cardiomyocytes. J Transl Med 2023; 21:662. [PMID: 37742032 PMCID: PMC10518098 DOI: 10.1186/s12967-023-04537-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/16/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND Sodium-glucose cotransporter 2 (SGLT2) inhibitors constitute the gold standard treatment for type 2 diabetes mellitus (T2DM). Among them, empagliflozin (EMPA) has shown beneficial effects against heart failure. Because cardiovascular diseases (mainly diabetic cardiomyopathy) are the leading cause of death in diabetic patients, the use of EMPA could be, simultaneously, cardioprotective and antidiabetic, reducing the risk of death from cardiovascular causes and decreasing the risk of hospitalization for heart failure in T2DM patients. Interestingly, recent studies have shown that EMPA has positive benefits for people with and without diabetes. This finding broadens the scope of EMPA function beyond glucose regulation alone to include a more intricate metabolic process that is, in part, still unknown. Similarly, this significantly increases the number of people with heart diseases who may be eligible for EMPA treatment. METHODS This study aimed to clarify the metabolic effect of EMPA on the human myocardial cell model by using orthogonal metabolomics, lipidomics, and proteomics approaches. The untargeted and multivariate analysis mimicked the fasting blood sugar level of T2DM patients (hyperglycemia: HG) and in the average blood sugar range (normal glucose: NG), with and without the addition of EMPA. RESULTS Results highlighted that EMPA was able to modulate and partially restore the levels of multiple metabolites associated with cellular stress, which were dysregulated in the HG conditions, such as nicotinamide mononucleotide, glucose-6-phosphate, lactic acid, FA 22:6 as well as nucleotide sugars and purine/pyrimidines. Additionally, EMPA regulated the levels of several lipid sub-classes, in particular dihydroceramide and triacylglycerols, which tend to accumulate in HG conditions resulting in lipotoxicity. Finally, EMPA counteracted the dysregulation of endoplasmic reticulum-derived proteins involved in cellular stress management. CONCLUSIONS These results could suggest an effect of EMPA on different metabolic routes, tending to rescue cardiomyocyte metabolic status towards a healthy phenotype.
Collapse
Affiliation(s)
- Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Ugo Chianese
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vicky Caponigro
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | | | | | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- IEOS CNR, Naples, Italy
- Azienda Ospedaliera Universitaria "Luigi Vanvitelli", Medical Epigenetics Program, Naples, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- UniCamillus, International Medical University, Rome, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Azienda Ospedaliera Universitaria "Luigi Vanvitelli", Medical Epigenetics Program, Naples, Italy
| | - Eduardo Sommella
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| |
Collapse
|
26
|
Mukherjee AG, Renu K, Gopalakrishnan AV, Jayaraj R, Dey A, Vellingiri B, Ganesan R. Epicardial adipose tissue and cardiac lipotoxicity: A review. Life Sci 2023; 328:121913. [PMID: 37414140 DOI: 10.1016/j.lfs.2023.121913] [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: 03/29/2023] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Epicardial adipose tissue (EAT) has morphological and physiological contiguity with the myocardium and coronary arteries, making it a visceral fat deposit with some unique properties. Under normal circumstances, EAT exhibits biochemical, mechanical, and thermogenic cardioprotective characteristics. Under clinical processes, epicardial fat can directly impact the heart and coronary arteries by secreting proinflammatory cytokines via vasocrine or paracrine mechanisms. It is still not apparent what factors affect this equilibrium. Returning epicardial fat to its physiological purpose may be possible by enhanced local vascularization, weight loss, and focused pharmacological therapies. This review centers on EAT's developing physiological and pathophysiological dimensions and its various and pioneering clinical utilities.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India.
| | - Rama Jayaraj
- Jindal Institute of Behavioral Sciences (JIBS), Jindal Global Institution of Eminence Deemed to Be University, 28, Sonipat 131001, India; Director of Clinical Sciences, Northern Territory Institute of Research and Training, Darwin, NT 0909, Australia
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
27
|
Lewandowski ED. Metabolic flux in the driver's seat during cardiac health and disease. J Mol Cell Cardiol 2023; 182:15-24. [PMID: 37451081 PMCID: PMC10529670 DOI: 10.1016/j.yjmcc.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
Cardiac function is a dynamic process that must adjust efficiently to the immediate demands of physical state and activity. So too, the metabolic support of cardiac function is a dynamic process that must respond, in time, to the demands of cardiac function and viability. Flux through metabolic pathways provides chemical energy and generates signaling molecules that regulate activity among intracellular compartments to meet these demands. Thus, flux through metabolic pathways provides a dynamic mode of support of cardiomyocytes during physiological and pathophysiological challenges. Any inability of metabolic flux to keep pace with the demands of the cardiomyocyte results in progressive dysfunction that contributes to cardiac disease. Thus, the priority in maintaining and regulating flux through metabolic pathways in the cardiomyocyte cannot be understated. Great potential exists in current efforts to elucidate metabolic mechanisms as therapeutic targets for the diseased heart. As a consequence, detecting metabolic flux in the functioning myocardium of the heart, under normal and diseased conditions, is essential in elucidating the metabolic basis of contractile dysfunction. As a companion to the 2022 ISHR Research Achievement Award lecture, this review examines the use and applications of stable isotope kinetics to quantify metabolic flux through intermediary pathways and the exchange and transport of intermediates across the mitochondrial membrane and sarcolemma of intact functioning hearts in determining how these intracellular events are coordinated to support cardiac function and health. Finally, this work reviews recently demonstrated metabolic defects in diseased hearts and the potential for metabolic alleviation of heart disease.
Collapse
Affiliation(s)
- E Douglas Lewandowski
- Department of Internal Medicine and Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, United States of America.
| |
Collapse
|
28
|
Da Dalt L, Cabodevilla AG, Goldberg IJ, Norata GD. Cardiac lipid metabolism, mitochondrial function, and heart failure. Cardiovasc Res 2023; 119:1905-1914. [PMID: 37392421 PMCID: PMC10681665 DOI: 10.1093/cvr/cvad100] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/31/2023] [Accepted: 03/01/2023] [Indexed: 07/03/2023] Open
Abstract
A fine balance between uptake, storage, and the use of high energy fuels, like lipids, is crucial in the homeostasis of different metabolic tissues. Nowhere is this balance more important and more precarious than in the heart. This highly energy-demanding muscle normally oxidizes almost all the available substrates to generate energy, with fatty acids being the preferred source under physiological conditions. In patients with cardiomyopathies and heart failure, changes in the main energetic substrate are observed; these hearts often prefer to utilize glucose rather than oxidizing fatty acids. An imbalance between uptake and oxidation of fatty acid can result in cellular lipid accumulation and cytotoxicity. In this review, we will focus on the sources and uptake pathways used to direct fatty acids to cardiomyocytes. We will then discuss the intracellular machinery used to either store or oxidize these lipids and explain how disruptions in homeostasis can lead to mitochondrial dysfunction and heart failure. Moreover, we will also discuss the role of cholesterol accumulation in cardiomyocytes. Our discussion will attempt to weave in vitro experiments and in vivo data from mice and humans and use several human diseases to illustrate metabolism gone haywire as a cause of or accomplice to cardiac dysfunction.
Collapse
Affiliation(s)
- Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, Milan, Italy
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, 550 1st Ave., New York, NY, USA
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, 550 1st Ave., New York, NY, USA
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, Milan, Italy
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Via Massimo Gorki 50, Cinisello Balsamo, Italy
| |
Collapse
|
29
|
Gu Y, Avolio E, Alvino VV, Thomas AC, Herman A, Miller PJ, Sullivan N, Faulkner A, Madeddu P. The tyrosine kinase inhibitor Dasatinib reduces cardiac steatosis and fibrosis in obese, type 2 diabetic mice. Cardiovasc Diabetol 2023; 22:214. [PMID: 37592236 PMCID: PMC10436421 DOI: 10.1186/s12933-023-01955-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Cardiac steatosis is an early yet overlooked feature of diabetic cardiomyopathy. There is no available therapy to treat this condition. Tyrosine kinase inhibitors (TKIs) are used as first or second-line therapy in different types of cancer. In cancer patients with diabetes mellitus, TKIs reportedly improved glycemic control, allowing insulin discontinuation. They also reduced liver steatosis in a murine model of non-alcoholic fatty liver disease. The present study aimed to determine the therapeutic effect of the second-generation TKI Dasatinib on lipid accumulation and cardiac function in obese, type 2 diabetic mice. We also assessed if the drug impacts extra-cardiac fat tissue depots. METHODS Two studies on 21-week-old male obese leptin receptor mutant BKS.Cg-+Leprdb/+Leprdb/OlaHsd (db/db) mice compared the effect of Dasatinib (5 mg/kg) and vehicle (10% DMSO + 90% PEG-300) given via gavage once every three days for a week or once every week for four weeks. Functional and volumetric indices were studied using echocardiography. Post-mortem analyses included the assessment of fat deposits and fibrosis using histology, and senescence using immunohistochemistry and flow cytometry. The anti-adipogenic action of Dasatinib was investigated on human bone marrow (BM)-derived mesenchymal stem cells (MSCs). Unpaired parametric or non-parametric tests were used to compare two and multiple groups as appropriate. RESULTS Dasatinib reduced steatosis and fibrosis in the heart of diabetic mice. The drug also reduced BM adiposity but did not affect other fat depots. These structural changes were associated with improved diastolic indexes, specifically the E/A ratio and non-flow time. Moreover, Dasatinib-treated mice had lower levels of p16 in the heart compared with vehicle-treated controls, suggesting an inhibitory impact of the drug on the senescence signalling pathway. In vitro, Dasatinib inhibited human BM-MSC viability and adipogenesis commitment. CONCLUSIONS Our findings suggest that Dasatinib opposes heart and BM adiposity and cardiac fibrosis. In the heart, this was associated with favourable functional consequences, namely improvement in an index of diastolic function. Repurposing TKI for cardiac benefit could address the unmet need of diabetic cardiac steatosis.
Collapse
Affiliation(s)
- Yue Gu
- Bristol Heart Institute, Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Elisa Avolio
- Bristol Heart Institute, Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Valeria V Alvino
- Bristol Heart Institute, Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Anita C Thomas
- Bristol Heart Institute, Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
- School of Cellular and Molecular Medicine, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Andrew Herman
- School of Cellular and Molecular Medicine, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Poppy J Miller
- School of Cellular and Molecular Medicine, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | | | - Ashton Faulkner
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Paolo Madeddu
- Bristol Heart Institute, Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK.
| |
Collapse
|
30
|
Cesaro A, De Michele G, Fimiani F, Acerbo V, Scherillo G, Signore G, Rotolo FP, Scialla F, Raucci G, Panico D, Gragnano F, Moscarella E, Scudiero O, Mennitti C, Calabrò P. Visceral adipose tissue and residual cardiovascular risk: a pathological link and new therapeutic options. Front Cardiovasc Med 2023; 10:1187735. [PMID: 37576108 PMCID: PMC10421666 DOI: 10.3389/fcvm.2023.1187735] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Obesity is a heterogeneous disease that affects almost one-third of the global population. A clear association has been established between obesity and cardiovascular disease (CVD). However, CVD risk is known to be related more to the local distribution of fat than to total body fat. Visceral adipose tissue (VAT) in particular has a high impact on CVD risk. This manuscript reviews the role of VAT in residual CV risk and the available therapeutic strategies for decreasing residual CV risk related to VAT accumulation. Among the many pathways involved in residual CV risk, obesity and particularly VAT accumulation play a major role by generating low-grade systemic inflammation, which in turn has a high prognostic impact on all-cause mortality and myocardial infarction. In recent years, many therapeutic approaches have been developed to reduce body weight. Orlistat was shown to reduce both weight and VAT but has low tolerability and many drug-drug interactions. Naltrexone-bupropion combination lowers body weight but has frequent side effects and is contraindicated in patients with uncontrolled hypertension. Liraglutide and semaglutide, glucagon-like peptide 1 (GLP-1) agonists, are the latest drugs approved for the treatment of obesity, and both have been shown to induce significant body weight loss. Liraglutide, semaglutide and other GLP-1 agonists also showed a positive effect on CV outcomes in diabetic patients. In addition, liraglutide showed to specifically reduce VAT and inflammatory biomarkers in obese patients without diabetes. GLP-1 agonists are promising compounds to limit inflammation in human visceral adipocytes.
Collapse
Affiliation(s)
- Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Gianantonio De Michele
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Fabio Fimiani
- Unit of Inherited and Rare Cardiovascular Diseases, A.O.R.N. Dei Colli “V. Monaldi”, Naples, Italy
| | - Vincenzo Acerbo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Gianmaria Scherillo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Giovanni Signore
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Francesco Paolo Rotolo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Francesco Scialla
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Giuseppe Raucci
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Domenico Panico
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Elisabetta Moscarella
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Olga Scudiero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- Ceinge Biotecnologie Avanzate Franco Salvatore S. C. a R. L., Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Cristina Mennitti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| |
Collapse
|
31
|
Anker SD, Usman MS, Anker MS, Butler J, Böhm M, Abraham WT, Adamo M, Chopra VK, Cicoira M, Cosentino F, Filippatos G, Jankowska EA, Lund LH, Moura B, Mullens W, Pieske B, Ponikowski P, Gonzalez-Juanatey JR, Rakisheva A, Savarese G, Seferovic P, Teerlink JR, Tschöpe C, Volterrani M, von Haehling S, Zhang J, Zhang Y, Bauersachs J, Landmesser U, Zieroth S, Tsioufis K, Bayes-Genis A, Chioncel O, Andreotti F, Agabiti-Rosei E, Merino JL, Metra M, Coats AJS, Rosano GMC. Patient phenotype profiling in heart failure with preserved ejection fraction to guide therapeutic decision making. A scientific statement of the Heart Failure Association, the European Heart Rhythm Association of the European Society of Cardiology, and the European Society of Hypertension. Eur J Heart Fail 2023; 25:936-955. [PMID: 37461163 DOI: 10.1002/ejhf.2894] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 07/26/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents a highly heterogeneous clinical syndrome affected in its development and progression by many comorbidities. The left ventricular diastolic dysfunction may be a manifestation of various combinations of cardiovascular, metabolic, pulmonary, renal, and geriatric conditions. Thus, in addition to treatment with sodium-glucose cotransporter 2 inhibitors in all patients, the most effective method of improving clinical outcomes may be therapy tailored to each patient's clinical profile. To better outline a phenotype-based approach for the treatment of HFpEF, in this joint position paper, the Heart Failure Association of the European Society of Cardiology, the European Heart Rhythm Association and the European Hypertension Society, have developed an algorithm to identify the most common HFpEF phenotypes and identify the evidence-based treatment strategy for each, while taking into account the complexities of multiple comorbidities and polypharmacy.
Collapse
Affiliation(s)
- Stefan D Anker
- Department of Cardiology, Deutsches Herzzentrum der Charité (Campus CVK), Berlin Institute of Health Center for Regenerative Therapies (BCRT), and German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | | | - Markus S Anker
- Deutsches Herzzentrum der Charité, Klinik fär Kardiologie, Angiologie und Intensivmedizin (Campus CBF), Berlin Institute of Health Center for Regenerative Therapies (BCRT), and German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Javed Butler
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Baylor Scott and White Research Institute, Dallas, TX, USA
| | - Michael Böhm
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Saarland University, Homburg, Germany
| | | | - Marianna Adamo
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | | | | | - Francesco Cosentino
- Department of Medicine, Karolinska Institutet, and Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Gerasimos Filippatos
- National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Ewa A Jankowska
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Lars H Lund
- Department of Medicine, Karolinska Institutet, and Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Brenda Moura
- Centro de Investigação em Tecnologias e Serviços de Saúde, Porto, Portugal; Serviço de Cardiologia, Hospital das Forças Armadas-Pólo do Porto, Porto, Portugal
| | - Wilfried Mullens
- Department of Cardiology, Ziekenhuis Oost Limburg, Genk and Faculty of Medicine and Life Sciences, University Hasselt, Belgium
| | - Burkert Pieske
- Berlin-Brandenburgische Gesellschaft für Herz-Kreislauferkrankungen (BBGK), Berlin, Germany
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
- Cardiology Department, Wroclaw Medical University, Wroclaw, Poland
| | - Jose R Gonzalez-Juanatey
- Cardiology Department, Hospital Clínico Universitario, Santiago de Compostela, IDIS, CIBERCV, Santiago de Compostela, Spain
| | - Amina Rakisheva
- Department of Cardiology, Scientific Institution of Cardiology and Internal Diseases, Almaty, Kazakhstan
| | - Gianluigi Savarese
- Department of Medicine, Karolinska Institutet, and Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Petar Seferovic
- Department Faculty of Medicine, University of Belgrade, Belgrade & Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - John R Teerlink
- Section of Cardiology, San Francisco Veterans Affairs Medical Center and School of Medicine, University of California, San Francisco, CA, USA
| | - Carsten Tschöpe
- Department of Cardiology, Deutsches Herzzentrum der Charité (Campus CVK), Berlin Institute of Health Center for Regenerative Therapies (BCRT), and German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine (CVK), Charité Universitätsmedizin, Berlin, Germany
| | - Maurizio Volterrani
- Cardio-Pulmonary Department, San Raffaele Open University of Rome; Exercise Science and Medicine, IRCCS San Raffaele - Rome, Italy
| | | | - Jian Zhang
- Fuwai Hospital Chinese Academic of Medical Science, Beijing, China
| | - Yuhui Zhang
- Fuwai Hospital Chinese Academic of Medical Science, Beijing, China
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Klinik fär Kardiologie, Angiologie und Intensivmedizin (Campus CBF), Berlin Institute of Health Center for Regenerative Therapies (BCRT), and German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Berlin, Germany
| | - Shelley Zieroth
- Section of Cardiology, Max Rady College of Medicine, University of Manitoba Winnipeg, Winnipeg, Manitoba, Canada
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, CIBERCV, Barcelona, Spain
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases 'Prof. C.C. Iliescu', University of Medicine Carol Davila, Bucharest, Romania
| | - Felicita Andreotti
- Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Catholic University Medical School, Rome, Italy
| | - Enrico Agabiti-Rosei
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Jose L Merino
- Department of Cardiology, La Paz University Hospital, IdiPaz, Universidad Autonoma, Madrid, Spain
| | - Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | | | - Giuseppe M C Rosano
- Cardio-Pulmonary Department, San Raffaele Open University of Rome; Exercise Science and Medicine, IRCCS San Raffaele - Rome, Italy
| |
Collapse
|
32
|
Pandey A, Khan MS, Patel KV, Bhatt DL, Verma S. Predicting and preventing heart failure in type 2 diabetes. Lancet Diabetes Endocrinol 2023:S2213-8587(23)00128-6. [PMID: 37385290 DOI: 10.1016/s2213-8587(23)00128-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 04/25/2023] [Accepted: 05/04/2023] [Indexed: 07/01/2023]
Abstract
The burden of heart failure among people with type 2 diabetes is increasing globally. People with comorbid type 2 diabetes and heart failure often have worse outcomes than those with only one of these conditions-eg, higher hospitalisation and mortality rates. Therefore, it is essential to implement optimal heart failure prevention strategies for people with type 2 diabetes. A detailed understanding of the pathophysiology underlying the occurrence of heart failure in type 2 diabetes can aid clinicians in identifying relevant risk factors and lead to early interventions that can help prevent heart failure. In this Review, we discuss the pathophysiology and risk factors of heart failure in type 2 diabetes. We also review the risk assessment tools for predicting heart failure incidence in people with type 2 diabetes as well as the data from clinical trials that have assessed the efficacy of lifestyle and pharmacological interventions. Finally, we discuss the potential challenges in implementing new management approaches and offer pragmatic recommendations to help overcome these challenges.
Collapse
Affiliation(s)
- Ambarish Pandey
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Kershaw V Patel
- Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Health System, New York, NY, USA
| | - Subodh Verma
- Division of Cardiac Surgery, St Michael's Hospital, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
33
|
Soghomonian A, Dutour A, Kachenoura N, Thuny F, Lasbleiz A, Ancel P, Cristofari R, Jouve E, Simeoni U, Kober F, Bernard M, Gaborit B. Is increased myocardial triglyceride content associated with early changes in left ventricular function? A 1H-MRS and MRI strain study. Front Endocrinol (Lausanne) 2023; 14:1181452. [PMID: 37424866 PMCID: PMC10323751 DOI: 10.3389/fendo.2023.1181452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Background Type 2 diabetes (T2D) and obesity induce left ventricular (LV) dysfunction. The underlying pathophysiological mechanisms remain unclear, but myocardial triglyceride content (MTGC) could be involved. Objectives This study aimed to determine which clinical and biological factors are associated with increased MTGC and to establish whether MTGC is associated with early changes in LV function. Methods A retrospective study was conducted using five previous prospective cohorts, leading to 338 subjects studied, including 208 well-phenotyped healthy volunteers and 130 subjects living with T2D and/or obesity. All the subjects underwent proton magnetic resonance spectroscopy and feature tracking cardiac magnetic resonance imaging to measure myocardial strain. Results MTGC content increased with age, body mass index (BMI), waist circumference, T2D, obesity, hypertension, and dyslipidemia, but the only independent correlate found in multivariate analysis was BMI (p=0.01; R²=0.20). MTGC was correlated to LV diastolic dysfunction, notably with the global peak early diastolic circumferential strain rate (r=-0.17, p=0.003), the global peak late diastolic circumferential strain rate (r=0.40, p<0.0001) and global peak late diastolic longitudinal strain rate (r=0.24, p<0.0001). MTGC was also correlated to systolic dysfunction via end-systolic volume index (r=-0.34, p<0.0001) and stroke volume index (r=-0.31, p<0.0001), but not with longitudinal strain (r=0.009, p=0.88). Interestingly, the associations between MTGC and strain measures did not persist in multivariate analysis. Furthermore, MTGC was independently associated with LV end-systolic volume index (p=0.01, R²=0.29), LV end-diastolic volume index (p=0.04, R²=0.46), and LV mass (p=0.002, R²=0.58). Conclusions Predicting MTGC remains a challenge in routine clinical practice, as only BMI independently correlates with increased MTGC. MTGC may play a role in LV dysfunction but does not appear to be involved in the development of subclinical strain abnormalities.
Collapse
Affiliation(s)
- Astrid Soghomonian
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, Marseille, France
| | - Anne Dutour
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, Marseille, France
| | - Nadjia Kachenoura
- Sorbonne Université, INSERM, CNRS, Laboratoire d’Imagerie Biomédicale, Paris, France
| | - Franck Thuny
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Intensive Care Unit, Department of Cardiology, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Aix-Marseille University, Marseille, France
| | - Adele Lasbleiz
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, Marseille, France
| | - Patricia Ancel
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | | | - Elisabeth Jouve
- UPCET, Clinical Pharmacology, Assistance-Publique Hôpitaux de Marseille, Marseille, France
| | - Umberto Simeoni
- Division of Pediatrics & DOHaD Laboratory, CHUV University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Frank Kober
- Aix-Marseille Université, CNRS, CRMBM, Marseille, France
| | | | - Bénédicte Gaborit
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, Marseille, France
| |
Collapse
|
34
|
Shi YJ, Dong GJ, Guo M. Targeting epicardial adipose tissue: A potential therapeutic strategy for heart failure with preserved ejection fraction with type 2 diabetes mellitus. World J Diabetes 2023; 14:724-740. [PMID: 37383601 PMCID: PMC10294070 DOI: 10.4239/wjd.v14.i6.724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/10/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome with various comorbidities, multiple cardiac and extracardiac pathophysiologic abnormalities, and diverse phenotypic presentations. Since HFpEF is a heterogeneous disease with different phenotypes, individualized treatment is required. HFpEF with type 2 diabetes mellitus (T2DM) represents a specific phenotype of HFpEF, with about 45%-50% of HFpEF patients suffering from T2DM. Systemic inflammation associated with dysregulated glucose metabolism is a critical pathological mechanism of HFpEF with T2DM, which is intimately related to the expansion and dysfunction (inflammation and hypermetabolic activity) of epicardial adipose tissue (EAT). EAT is well established as a very active endocrine organ that can regulate the pathophysiological processes of HFpEF with T2DM through the paracrine and endocrine mechanisms. Therefore, suppressing abnormal EAT expansion may be a promising therapeutic strategy for HFpEF with T2DM. Although there is no treatment specifically for EAT, lifestyle management, bariatric surgery, and some pharmaceutical interventions (anti-cytokine drugs, statins, proprotein convertase subtilisin/kexin type 9 inhibitors, metformin, glucagon-like peptide-1 receptor agonists, and especially sodium-glucose cotransporter-2 inhibitors) have been shown to attenuate the inflammatory response or expansion of EAT. Importantly, these treatments may be beneficial in improving the clinical symptoms or prognosis of patients with HFpEF. Accordingly, well-designed randomized controlled trials are needed to validate the efficacy of current therapies. In addition, more novel and effective therapies targeting EAT are needed in the future.
Collapse
Affiliation(s)
- Yu-Jiao Shi
- Department of Cardiovascular Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Guo-Ju Dong
- Department of Cardiovascular Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Ming Guo
- Department of Cardiovascular Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing 100091, China
| |
Collapse
|
35
|
Aryee E, Ozkan B, Ndumele CE. Heart failure and obesity: The latest pandemic. Prog Cardiovasc Dis 2023:S0033-0620(23)00051-8. [PMID: 37236574 DOI: 10.1016/j.pcad.2023.05.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023]
Abstract
The marked rise in rates of obesity, which is most prominent among individuals from socio-economically disadvantaged circumstances, has been a powerful contributor to the rising prevalence of heart failure (HF). Obesity has indirect effects on HF through the development of several metabolic risk factors, but also direct adverse effects on the myocardium. Obesity contributes to myocardial dysfunction and HF risk through multiple mechanisms, including hemodynamic changes, neurohormonal activation, endocrine and paracrine effects of adipose tissue, ectopic fat deposition and lipotoxicity. These processes principally result in concentric left ventricular (LV) remodeling and predominant increase in the risk for HF with preserved LV ejection fraction (HFpEF). Despite the excess risk for HF associated with obesity, there is a well described obesity paradox in which individuals with overweight and grade I obesity have better survival than those with normal weight and overweight. Despite the obesity paradox among individuals with prevalent HF, intentional weight loss is associated with improvements in metabolic risk factors, myocardial dysfunction and quality of life, in a dose-response fashion. In matched observational studies of bariatric surgery patients, marked weight loss is associated with decreased risk for developing HF, as well as improved cardiovascular disease (CVD) outcomes in those with existing HF. Ongoing clinical trials using powerful new obesity pharmacotherapies in individuals in with obesity and CVD may provide definitive information regarding the cardiovascular impact of weight loss. Given the powerful contribution of rising obesity prevalence to rates of HF, addressing these intertwined epidemics is a clinical and public health priority.
Collapse
Affiliation(s)
- Ebenezer Aryee
- Division of Cardiology, Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Bige Ozkan
- Division of Cardiology, Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Chiadi E Ndumele
- Division of Cardiology, Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America.
| |
Collapse
|
36
|
Mavrogeni SI, Markousis-Mavrogenis G, Bacopoulou F, Chrousos GP. Cardiovascular Magnetic Resonance Imaging as an Adjunct to the Evaluation of Cardiovascular Involvement in Diabetes Mellitus. J Pers Med 2023; 13:724. [PMID: 37240894 PMCID: PMC10222166 DOI: 10.3390/jpm13050724] [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: 03/14/2023] [Revised: 04/13/2023] [Accepted: 04/22/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetes mellitus (DM) is a new epidemic which has presented an immense increase in recent decades, due to the rapid increase in obesity. Cardiovascular disease (CVD) significantly reduces life expectancy and is the main cause of death in type 2 diabetes mellitus (T2DM). Strict glycemic control is a well-established method to combat microvascular CVD of type 1 diabetes mellitus (T1DM); its role against CVD of the T2DM risk has not been well documented. Therefore, the most efficient prevention is multifactorial risk factor reduction. Recently, the European Society of Cardiology published its 2019 recommendations on CVD in DM. Although all clinical points were discussed in this document, only a few comments were presented about when and how we should recommend cardiovascular (CV) imaging. Currently, CV imaging is the "must" in CV noninvasive evaluation. Alterations in CV imaging parameters can lead to early recognition of various types of CVD. In this paper, we briefly discuss the role of noninvasive imaging modalities, emphasizing the benefits of including cardiovascular magnetic resonance (CMR) in the evaluation of DM. CMR, in the same examination, can provide an assessment of tissue characterization, perfusion and function, with excellent reproducibility and without radiation or limitations, due to the body habitus. Therefore, it can play a dominant role in the prevention and risk stratification of DM. The suggested protocol for DM evaluation should include routine annual echocardiographic evaluation of all DM patients and CMR assessment of those with poorly controlled DM, microalbuminuria, heart failure, arrhythmia and recent alterations in clinical or echocardiographic evaluation.
Collapse
Affiliation(s)
- Sophie I. Mavrogeni
- Onassis Cardiac Surgery Center, 17674 Athens, Greece
- University Research Institute of Maternal and Child Health and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece
| | | | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece
| | - George P. Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, 11527 Athens, Greece
| |
Collapse
|
37
|
Michaelidou M, Pappachan JM, Jeeyavudeen MS. Management of diabesity: Current concepts. World J Diabetes 2023; 14:396-411. [PMID: 37122433 PMCID: PMC10130896 DOI: 10.4239/wjd.v14.i4.396] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/15/2023] [Accepted: 03/20/2023] [Indexed: 04/12/2023] Open
Abstract
The global prevalence of obesity is increasing rapidly with an exponential rise in incidence of type 2 diabetes mellitus in recent years. 'Diabesity', the term coined to show the strong interlink between obesity and diabetes, is the direct cons-equence of the obesity pandemic, and poses significant challenges in the management of the disease. Without addressing the clinical and mechanistic complications of obesity such as metabolic-associated fatty liver disease and obstructive sleep apnoea, a rational management algorithm for diabesity cannot be developed. Several classes of anti-diabetic medications including insulins, sulphonylureas, thiazolidinediones and meglitinides are associated with the risk of weight gain and may potentially worsen diabesity. Therefore, appropriate selection of antidiabetic drug regimen is crucial in the medical management of diabesity. The role of non-pharmacological measures such as dietary adjustments, exercise interventions and bariatric procedures should also be emphasised. Unfortunately, the importance of appropriate and optimal management of diabesity is often overlooked by medical professionals when achieving adequate glycemic control which results in inappropriate management of the disease and its complications. This review provides a narrative clinical update on the evidence behind the management of diabesity.
Collapse
Affiliation(s)
- Maria Michaelidou
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
| | - Joseph M Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
- Faculty of Science, Manchester Metropolitan University, Manchester M15 6BH, United Kingdom
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Mohammad Sadiq Jeeyavudeen
- Department of Endocrinology & Metabolism, University Hospitals of Edinburgh, Edinburgh EH16 4SA, United Kingdom
| |
Collapse
|
38
|
Jung J, Park J, Kim M, Ha J, Cho H, Park SB. SB2301-mediated perturbation of membrane composition in lipid droplets induces lipophagy and lipid droplets ubiquitination. Commun Biol 2023; 6:300. [PMID: 36944894 PMCID: PMC10030462 DOI: 10.1038/s42003-023-04682-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/09/2023] [Indexed: 03/23/2023] Open
Abstract
Lipid droplets (LDs) are involved in various biological events in cells along with their primary role as a storage center for neutral lipids. Excessive accumulation of LDs is highly correlated with various diseases, including metabolic diseases. Therefore, a basic understanding of the molecular mechanism of LD degradation would be beneficial in both academic and industrial research. Lipophagy, a selective autophagy mechanism/LD degradation process, has gained increased attention in the research community. Herein, we sought to elucidate a novel lipophagy mechanism by utilizing the LD-degrading small molecule, SB2301, which activates ubiquitin-mediated lipophagy. Using a label-free target identification method, we revealed that ethanolamine-phosphate cytidylyltransferase 2 (PCYT2) is a potential target protein of SB2301. We also demonstrated that although SB2301 does not modulate PCYT2 function, it induces the cellular translocation of PCYT2 to the LD surface and spatially increases the phosphatidylethanolamine (PE)/phosphatidylcholine (PC) ratio of the LD membrane, causing LD coalescence, leading to the activation of lipophagy process to maintain energy homeostasis.
Collapse
Affiliation(s)
- Jinjoo Jung
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, South Korea
| | - Jongbeom Park
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, South Korea
| | - Mingi Kim
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, South Korea
| | - Jaeyoung Ha
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, South Korea
| | - Hana Cho
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, South Korea
| | - Seung Bum Park
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, South Korea.
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
39
|
Liu L, Sun K, Luo Y, Wang B, Yang Y, Chen L, Zheng S, Wu T, Xiao P. Myocardin-related transcription factor A, regulated by serum response factor, contributes to diabetic cardiomyopathy in mice. Life Sci 2023; 317:121470. [PMID: 36758668 DOI: 10.1016/j.lfs.2023.121470] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/10/2023]
Abstract
AIMS Diabetic cardiomyopathy is a significant contributor to the global pandemic of heart failure. In the present study we investigated the involvement of myocardin-related transcription factor A (MRTF-A), a transcriptional regulator, in this process. MATERIALS AND METHODS Diabetic cardiomyopathy was induced in mice by feeding with a high-fat diet (HFD) or streptozotocin (STZ) injection. KEY FINDINGS We report that MRTF-A was up-regulated in the hearts of mice with diabetic cardiomyopathy. MRTF-A expression was also up-regulated by treatment with palmitate in cultured cardiomyocytes in vitro. Mechanistically, serum response factor (SRF) bound to the MRTF-A gene promoter and activated MRTF-A transcription in response to pro-diabetic stimuli. Knockdown of SRF abrogated MRTF-A induction in cardiomyocytes treated with palmitate. When cardiomyocytes conditional MRTF-A knockout mice (MRTF-A CKO) and wild type (WT) mice were placed on an HFD to induce diabetic cardiomyopathy, it was found that the CKO mice and the WT mice displayed comparable metabolic parameters including body weight, blood insulin concentration, blood cholesterol concentration, and glucose tolerance. However, both systolic and diastolic cardiac function were exacerbated by MRTF-A deletion in the heart. SIGNIFICANCE These data suggest that MRTF-A up-regulation might serve as an important compensatory mechanism to safeguard the deterioration of cardiac function during diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Li Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China; Department of Cardiology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Ke Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yajun Luo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Bingshu Wang
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Medical Research Center of The First Affiliated Hospital, Hainan Women and Children Medical Center, Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou 571199, China; Department of Pathology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Yuyu Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Shaojiang Zheng
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Medical Research Center of The First Affiliated Hospital, Hainan Women and Children Medical Center, Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou 571199, China.
| | - Teng Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.
| | - Pingxi Xiao
- Department of Cardiology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
40
|
Oneglia AP, Szczepaniak LS, Jaffery MF, Cipher DJ, McDonald JG, Haykowsky MJ, Moreau KL, Clegg DJ, Zaha V, Nelson MD. Myocardial steatosis impairs left ventricular diastolic-systolic coupling in healthy humans. J Physiol 2023; 601:1371-1382. [PMID: 36891609 DOI: 10.1113/jp284272] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/02/2023] [Indexed: 03/10/2023] Open
Abstract
Mounting evidence suggests that myocardial steatosis contributes to left ventricular diastolic dysfunction, but definitive evidence in humans is lacking due to confounding comorbidities. As such, we utilized a 48-h food restriction model to acutely increase myocardial triglyceride (mTG) content - measured by 1 H magnetic resonance spectroscopy - in 27 young healthy volunteers (13 men/14 women). Forty-eight hours of fasting caused a more than 3-fold increase in mTG content (P < 0.001). Diastolic function - defined as early diastolic circumferential strain rate (CSRd) - was unchanged following the 48-h fasting intervention, but systolic circumferential strain rate was elevated (P < 0.001), indicative of systolic-diastolic uncoupling. Indeed, in a separate control experiment in 10 individuals, administration of low-dose dobutamine (2 μg/kg/min) caused a similar change in systolic circumferential strain rate as was found during 48 h of food restriction, along with a proportionate increase in CSRd, such that the two metrics remained coupled. Taken together, these data indicate that myocardial steatosis contributes to diastolic dysfunction by impairing diastolic-systolic coupling in healthy adults, and suggest that steatosis may contribute to the progression of heart disease. KEY POINTS: Preclinical evidence strongly suggests that myocardial lipid accumulation (termed steatosis) is an important mechanism driving heart disease. Definitive evidence in humans is limited due to the confounding influence of multiple underlying comorbidities. Using a 48-h food restriction model to acutely increase myocardial triglyceride content in young healthy volunteers, we demonstrate an association between myocardial steatosis and left ventricular diastolic dysfunction. These data advance the hypothesis that myocardial steatosis may contribute to diastolic dysfunction and suggest myocardial steatosis as a putative therapeutic target.
Collapse
Affiliation(s)
- Andrew P Oneglia
- College of Nursing and Health Innovation, Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| | | | - Manall F Jaffery
- College of Nursing and Health Innovation, Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| | - Daisha J Cipher
- College of Nursing and Health Innovation, Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| | - Jeffrey G McDonald
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mark J Haykowsky
- College of Health Sciences, Faculty of Nursing, University of Alberta, Edmonton, Alberta, Canada
| | - Kerrie L Moreau
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Vlad Zaha
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael D Nelson
- College of Nursing and Health Innovation, Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
41
|
Kashiwagi-Takayama R, Kozawa J, Hosokawa Y, Kato S, Kawata S, Ozawa H, Mineo R, Ishibashi C, Baden MY, Iwamoto R, Saisho K, Fujita Y, Tamba S, Sugiyama T, Nishizawa H, Maeda N, Yamamoto K, Higashi M, Yamada Y, Sakata Y, Matsuzawa Y, Shimomura I. Myocardial fat accumulation is associated with cardiac dysfunction in patients with type 2 diabetes, especially in elderly or female patients: a retrospective observational study. Cardiovasc Diabetol 2023; 22:48. [PMID: 36882731 PMCID: PMC9993532 DOI: 10.1186/s12933-023-01782-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Ectopic fat is fat that accumulates in or around specific organs or compartments of the body including myocardium. The clinical features of type 2 diabetes patients with high fat accumulation in the myocardium remain unknown. Moreover, little is known about the influence of myocardial fat accumulation in type 2 diabetes on coronary artery disease and cardiac dysfunction. We aimed to clarify the clinical features, including cardiac functions, of type 2 diabetes patients with myocardial fat accumulation. METHODS We retrospectively enrolled type 2 diabetes patients who underwent ECG-gated coronary computed tomography angiography (CCTA) and abdominal computed tomography (CT) scan examinations within 1 year of CCTA from January 2000 to March 2021. High fat accumulation in the myocardium was defined as the low mean myocardial CT value of three regions of interest, and the associations between CT values and clinical characteristics or cardiac functions were assessed. RESULTS In total, 124 patients were enrolled (72 males and 52 females). The mean age was 66.6 years, the mean BMI was 26.2 kg/m2, the mean ejection fraction (EF) was 67.6%, and the mean myocardial CT value was 47.7 Hounsfield unit. A significant positive correlation was found between myocardial CT value and EF (r = 0.3644, p = 0.0004). The multiple regression analyses also showed that myocardial CT value was independently associated with EF (estimate, 0.304; 95% confidence interval (CI) 0.092 to 0.517; p = 0.0056). Myocardial CT value showed significant negative correlations with BMI, visceral fat area and subcutaneous fat area (r = - 0.1923, - 0.2654, and -0.3569, respectively, p < 0.05). In patients who were ≥ 65 years or female, myocardial CT value showed significant positive correlations with not only EF (r = 0.3542 and 0.4085, respectively, p < 0.01) but also early lateral annular tissue Doppler velocity (Lat e') (r = 0.5148 and 0.5361, respectively, p < 0.05). The multiple regression analyses showed that myocardial CT value was independently associated with EF and Lat e' in these subgroups (p < 0.05). CONCLUSIONS Patients with type 2 diabetes, especially in elderly or female patients, who had more myocardial fat had more severe left ventricular systolic and diastolic dysfunctions. Reducing myocardial fat accumulation may be a therapeutic target for type 2 diabetes patients.
Collapse
Affiliation(s)
- Risa Kashiwagi-Takayama
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Junji Kozawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan. .,Department of Diabetes Care Medicine, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Yoshiya Hosokawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Sarasa Kato
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Satoshi Kawata
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Harutoshi Ozawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Lifestyle Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Ryohei Mineo
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Endocrinology and Metabolism, Sumitomo Hospital, Osaka, Japan
| | - Chisaki Ishibashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Megu Y Baden
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Lifestyle Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Ryuya Iwamoto
- Department of Endocrinology and Metabolism, Sumitomo Hospital, Osaka, Japan
| | - Kenji Saisho
- Department of Endocrinology and Metabolism, Sumitomo Hospital, Osaka, Japan
| | - Yukari Fujita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Sachiko Tamba
- Department of Endocrinology and Metabolism, Sumitomo Hospital, Osaka, Japan
| | - Takuya Sugiyama
- Department of Endocrinology and Metabolism, Sumitomo Hospital, Osaka, Japan
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Koji Yamamoto
- Department of Endocrinology and Metabolism, Sumitomo Hospital, Osaka, Japan
| | - Masahiro Higashi
- Department of Radiology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Yuya Yamada
- Department of Endocrinology and Metabolism, Sumitomo Hospital, Osaka, Japan
| | - Yasushi Sakata
- Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuji Matsuzawa
- Department of Endocrinology and Metabolism, Sumitomo Hospital, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
42
|
Rodríguez-Calvo R, Granado-Casas M, Pérez-Montes de Oca A, Julian MT, Domingo M, Codina P, Santiago-Vacas E, Cediel G, Julve J, Rossell J, Masana L, Mauricio D, Lupón J, Bayes-Genis A, Alonso N. Fatty Acid Binding Proteins 3 and 4 Predict Both All-Cause and Cardiovascular Mortality in Subjects with Chronic Heart Failure and Type 2 Diabetes Mellitus. Antioxidants (Basel) 2023; 12:antiox12030645. [PMID: 36978893 PMCID: PMC10044995 DOI: 10.3390/antiox12030645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Subjects with type 2 diabetes mellitus (T2D) are at increased risk for heart failure (HF). The cardiac-specific (FABP3) and adipose-tissue-specific (FABP4) types of the fatty acid binding proteins have been associated with both all-cause and cardiovascular (CV) mortality. The aim of this study was to explore the prognosis value of FABP3 and FABP4 in ambulatory subjects with chronic HF (CHF), with and without T2D. A prospective study involving 240 ambulatory CHF subjects was performed. Patients were followed-up for a mean of 5.78 ± 3.30 years and cause of death (if any) was recorded. Primary endpoints were defined as all-cause and CV death, and a composite endpoint that included CV death or hospitalization for HF was included as a secondary endpoint. Baseline serum samples were obtained and the serum FABP3 and FABP4 concentrations were assessed by sandwich enzyme-linked immunosorbent assay. Survival analysis was performed with multivariable Cox regressions, using Fine and Gray competing risks models when needed, to explore the prognostic value of FABP3 and FABP4 concentrations, adjusting for potential confounders. Type 2 diabetes mellitus was highly prevalent, accounting for 47.5% for total subjects with CHF. Subjects with T2D showed higher mortality rates (T2D: 69.30%; non-T2D: 50.79%, p = 0.004) and higher serum FABP3 (1829.3 (1104.9–3440.5) pg/mL vs. 1396.05 (820.3–2362.16) pg/mL, p = 0.007) and FABP4 (45.5 (27.6–79.8) ng/mL vs. 34.1 (24.09–55.3) ng/mL, p = 0.006) concentrations compared with non-T2D CHF subjects. In the whole study cohort, FABP3 was independently associated with all-cause death, and both FABP3 and FABP4 concentrations were associated with CV mortality. The predictive values of these two molecules for all-cause (FABP3: HR 1.25, 95% CI 1.09–1.44; p = 0.002. FABP4: HR 2.21, 95% CI 1.12–4.36; p = 0.023) and CV mortality (FABP3: HR 1.28, 95% CI 1.09–1.50; p = 0.002. FABP4: HR 4.19, 95% CI 2.21–7.95; p < 0.001) were only statistically significant in the subgroup of subjects with T2D. Notably, FABP4 (HR 2.07, 95% CI 1.11–3.87; p = 0.022), but not FABP3, also predicted the occurrence of the composite endpoint (death or hospitalization for HF) only in subjects with T2D. All these associations were not found in CHF subjects without T2D. Our findings support the usefulness of serum FABP3 and FABP4 concentrations as independent predictors for the occurrence of all-cause and CV mortality in ambulatory subjects with CHF with T2D.
Collapse
Affiliation(s)
- Ricardo Rodríguez-Calvo
- Vascular Medicine and Metabolism Unit, “Sant Joan” University Hospital, Institut de Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain
- Research Unit on Lipids and Atherosclerosis, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Minerva Granado-Casas
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Nursing and Physiotherapy, Health Sciences Faculty, University of Lleida, IRBLleida, 25198 Lleida, Spain
- DAP-Cat Group, Unitat de Suport a la Recerca Barcelona, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), 08041 Barcelona, Spain
| | | | - María Teresa Julian
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| | - Mar Domingo
- Heart Failure Clinic and Cardiology Service, University Hospital Germans Trias i Pujol, 08916 Badalona, Spain
| | - Pau Codina
- Heart Failure Clinic and Cardiology Service, University Hospital Germans Trias i Pujol, 08916 Badalona, Spain
| | - Evelyn Santiago-Vacas
- Heart Failure Clinic and Cardiology Service, University Hospital Germans Trias i Pujol, 08916 Badalona, Spain
| | - Germán Cediel
- Heart Failure Clinic and Cardiology Service, University Hospital Germans Trias i Pujol, 08916 Badalona, Spain
| | - Josep Julve
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Institut d’Investigació Biomèdica de l’Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Joana Rossell
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Lluís Masana
- Vascular Medicine and Metabolism Unit, “Sant Joan” University Hospital, Institut de Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain
- Research Unit on Lipids and Atherosclerosis, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), 43204 Reus, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Didac Mauricio
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain
- Faculty of Medicine, University of Vic & Central University of Catalonia, 08500 Vic, Spain
| | - Josep Lupón
- Heart Failure Clinic and Cardiology Service, University Hospital Germans Trias i Pujol, 08916 Badalona, Spain
- Department of Medicine, Universitat Autonoma de Barcelona, 08023 Barcelona, Spain
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Antoni Bayes-Genis
- Heart Failure Clinic and Cardiology Service, University Hospital Germans Trias i Pujol, 08916 Badalona, Spain
- Department of Medicine, Universitat Autonoma de Barcelona, 08023 Barcelona, Spain
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (A.B.-G.); (N.A.)
| | - Núria Alonso
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
- Department of Medicine, Universitat Autonoma de Barcelona, 08023 Barcelona, Spain
- Correspondence: (A.B.-G.); (N.A.)
| |
Collapse
|
43
|
Wu Y, Huang T, Li X, Shen C, Ren H, Wang H, Wu T, Fu X, Deng S, Feng Z, Xiong S, Li H, Gao S, Yang Z, Gao F, Dong L, Cheng J, Cai W. Retinol dehydrogenase 10 reduction mediated retinol metabolism disorder promotes diabetic cardiomyopathy in male mice. Nat Commun 2023; 14:1181. [PMID: 36864033 PMCID: PMC9981688 DOI: 10.1038/s41467-023-36837-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/20/2023] [Indexed: 03/04/2023] Open
Abstract
Diabetic cardiomyopathy is a primary myocardial injury induced by diabetes with complex pathogenesis. In this study, we identify disordered cardiac retinol metabolism in type 2 diabetic male mice and patients characterized by retinol overload, all-trans retinoic acid deficiency. By supplementing type 2 diabetic male mice with retinol or all-trans retinoic acid, we demonstrate that both cardiac retinol overload and all-trans retinoic acid deficiency promote diabetic cardiomyopathy. Mechanistically, by constructing cardiomyocyte-specific conditional retinol dehydrogenase 10-knockout male mice and overexpressing retinol dehydrogenase 10 in male type 2 diabetic mice via adeno-associated virus, we verify that the reduction in cardiac retinol dehydrogenase 10 is the initiating factor for cardiac retinol metabolism disorder and results in diabetic cardiomyopathy through lipotoxicity and ferroptosis. Therefore, we suggest that the reduction of cardiac retinol dehydrogenase 10 and its mediated disorder of cardiac retinol metabolism is a new mechanism underlying diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yandi Wu
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Tongsheng Huang
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Xinghui Li
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Conghui Shen
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Honglin Ren
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Haiping Wang
- Prenatal Diagnosis Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Teng Wu
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Xinlu Fu
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Shijie Deng
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Ziqi Feng
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Shijie Xiong
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Hui Li
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Saifei Gao
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Zhenyu Yang
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Fei Gao
- Durbrain Medical Laboratory, Hangzhou, 310000, Zhejiang, China
| | - Lele Dong
- Durbrain Medical Laboratory, Hangzhou, 310000, Zhejiang, China
| | - Jianding Cheng
- Department of Forensic Pathology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Weibin Cai
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China. .,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China. .,Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
44
|
Granieri MC, Rocca C, De Bartolo A, Nettore IC, Rago V, Romeo N, Ceramella J, Mariconda A, Macchia PE, Ungaro P, Sinicropi MS, Angelone T. Quercetin and Its Derivative Counteract Palmitate-Dependent Lipotoxicity by Inhibiting Oxidative Stress and Inflammation in Cardiomyocytes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3492. [PMID: 36834186 PMCID: PMC9958705 DOI: 10.3390/ijerph20043492] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Cardiac lipotoxicity plays an important role in the pathogenesis of obesity-related cardiovascular disease. The flavonoid quercetin (QUE), a nutraceutical compound that is abundant in the "Mediterranean diet", has been shown to be a potential therapeutic agent in cardiac and metabolic diseases. Here, we investigated the beneficial role of QUE and its derivative Q2, which demonstrates improved bioavailability and chemical stability, in cardiac lipotoxicity. To this end, H9c2 cardiomyocytes were pre-treated with QUE or Q2 and then exposed to palmitate (PA) to recapitulate the cardiac lipotoxicity occurring in obesity. Our results showed that both QUE and Q2 significantly attenuated PA-dependent cell death, although QUE was effective at a lower concentration (50 nM) when compared with Q2 (250 nM). QUE decreased the release of lactate dehydrogenase (LDH), an important indicator of cytotoxicity, and the accumulation of intracellular lipid droplets triggered by PA. On the other hand, QUE protected cardiomyocytes from PA-induced oxidative stress by counteracting the formation of malondialdehyde (MDA) and protein carbonyl groups (which are indicators of lipid peroxidation and protein oxidation, respectively) and intracellular ROS generation, and by improving the enzymatic activities of catalase and superoxide dismutase (SOD). Pre-treatment with QUE also significantly attenuated the inflammatory response induced by PA by reducing the release of key proinflammatory cytokines (IL-1β and TNF-α). Similar to QUE, Q2 (250 nM) also significantly counteracted the PA-provoked increase in intracellular lipid droplets, LDH, and MDA, improving SOD activity and decreasing the release of IL-1β and TNF-α. These results suggest that QUE and Q2 could be considered potential therapeutics for the treatment of the cardiac lipotoxicity that occurs in obesity and metabolic diseases.
Collapse
Affiliation(s)
- Maria Concetta Granieri
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Anna De Bartolo
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Immacolata Cristina Nettore
- Dipartimento di Medicina Clinica e Chirurgia, Scuola di Medicina, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Naomi Romeo
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Annaluisa Mariconda
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Paolo Emidio Macchia
- Dipartimento di Medicina Clinica e Chirurgia, Scuola di Medicina, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
| | - Paola Ungaro
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale (IEOS) “Gaetano Salvatore”, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
- National Institute of Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
45
|
Diabetes Mellitus and Heart Failure: Epidemiology, Pathophysiologic Mechanisms, and the Role of SGLT2 Inhibitors. Life (Basel) 2023; 13:life13020497. [PMID: 36836854 PMCID: PMC9968235 DOI: 10.3390/life13020497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Diabetes mellitus (DM) and heart failure (HF) are frequently encountered afflictions that are linked by a common pathophysiologic background. According to landmark studies, those conditions frequently coexist, and this interaction represents a poor prognostic indicator. Based on mechanistic studies, HF can be propagated by multiple pathophysiologic pathways, such as inflammation, oxidative stress, endothelial dysfunction, fibrosis, cardiac autonomic neuropathy, and alterations in substrate utilization. In this regard, DM may augment myocardial inflammation, fibrosis, autonomic dysfunction, and lipotoxicity. As the interaction between DM and HF appears critical, the new cornerstone in DM and HF treatment, sodium-glucose cotransporter-2 inhibitors (SGLT2i), may be able to revert the pathophysiology of those conditions and lead to beneficial HF outcomes. In this review, we aim to highlight the deleterious pathophysiologic interaction between DM and HF, as well as demonstrate the beneficial role of SGLT2i in this field.
Collapse
|
46
|
Shitole SG, Naveed M, Wang Z, Wang T, Kato Y, Ambale-Venkatesh B, Kaplan RC, Tien PC, Anastos K, Lazar JM, Lima JAC, Qi Q, Kizer JR. Metabolomic Profiling of Cardiac Fibrosis and Steatosis in Women With or at Risk for HIV. J Acquir Immune Defic Syndr 2023; 92:162-172. [PMID: 36215981 PMCID: PMC9839486 DOI: 10.1097/qai.0000000000003118] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/05/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Heart failure is a prevalent disorder whose prognosis remains poor despite advances in treatment. Women with or at risk for HIV may be particularly susceptible, yet the metabolic pathways that promote myocardial disease and heart failure in this context remain incompletely characterized. METHODS To evaluate the metabolomic signatures of cardiac magnetic resonance measured phenotypes, we used available plasma metabolomic measures from participants in the Women's Interagency HIV Study who underwent cardiac magnetic resonance imaging. Our primary outcomes were myocardial extracellular volume fraction (MECV) and intramyocardial triglyceride content (IMTG). We applied partial least squares and identified the top 10 lipid and polar metabolites associated with MECV and IMTG. We used multivariable linear regression to evaluate these metabolites' individual associations with each phenotype. RESULTS The mean age of participants (n = 153) was 53 ± 7, 93% were Black or Hispanic, and 74% were HIV positive. Phenylacetylglutamine, a microbial metabolite, was positively associated with MECV after full adjustment and false discovery rate correction. Three phosphatidylcholine species, N-acetylaspartic acid, and a lysophosphatidylcholine species were inversely associated with IMTG, while prolylglycine, methionine sulfoxide, sphingosine, taurine, and phosphorylcholine were positively associated with this phenotype. We found no evidence of interaction by HIV for the observed associations, but there was effect modification by hepatitis C virus of taurine's and phosphorylcholine's associations with IMTG. CONCLUSION Among women with or at risk for HIV, we related various lipid and polar metabolites to cardiac fibrosis or steatosis, of which phenylacetylglutamine, N-acetylaspartic acid, and prolylglycine are novel. These findings implicate plausible mechanisms that could be targetable for therapeutics.
Collapse
Affiliation(s)
- Sanyog G. Shitole
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
- University of California San Francisco, San Francisco, CA
| | - Mahim Naveed
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
- University of California San Francisco, San Francisco, CA
| | - Zheng Wang
- Albert Einstein College of Medicine, Bronx, NY
| | - Tao Wang
- Albert Einstein College of Medicine, Bronx, NY
| | - Yoko Kato
- Johns Hopkins University, Baltimore, MD
| | | | - Robert C. Kaplan
- Albert Einstein College of Medicine, Bronx, NY
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Phyllis C. Tien
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
- University of California San Francisco, San Francisco, CA
| | | | | | | | - Qibin Qi
- Albert Einstein College of Medicine, Bronx, NY
| | - Jorge R. Kizer
- San Francisco Veterans Affairs Health Care System, San Francisco, CA
- University of California San Francisco, San Francisco, CA
| |
Collapse
|
47
|
Fotaki A, Velasco C, Prieto C, Botnar RM. Quantitative MRI in cardiometabolic disease: From conventional cardiac and liver tissue mapping techniques to multi-parametric approaches. Front Cardiovasc Med 2023; 9:991383. [PMID: 36756640 PMCID: PMC9899858 DOI: 10.3389/fcvm.2022.991383] [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: 07/11/2022] [Accepted: 12/29/2022] [Indexed: 01/24/2023] Open
Abstract
Cardiometabolic disease refers to the spectrum of chronic conditions that include diabetes, hypertension, atheromatosis, non-alcoholic fatty liver disease, and their long-term impact on cardiovascular health. Histological studies have confirmed several modifications at the tissue level in cardiometabolic disease. Recently, quantitative MR methods have enabled non-invasive myocardial and liver tissue characterization. MR relaxation mapping techniques such as T1, T1ρ, T2 and T2* provide a pixel-by-pixel representation of the corresponding tissue specific relaxation times, which have been shown to correlate with fibrosis, altered tissue perfusion, oedema and iron levels. Proton density fat fraction mapping approaches allow measurement of lipid tissue in the organ of interest. Several studies have demonstrated their utility as early diagnostic biomarkers and their potential to bear prognostic implications. Conventionally, the quantification of these parameters by MRI relies on the acquisition of sequential scans, encoding and mapping only one parameter per scan. However, this methodology is time inefficient and suffers from the confounding effects of the relaxation parameters in each single map, limiting wider clinical and research applications. To address these limitations, several novel approaches have been proposed that encode multiple tissue parameters simultaneously, providing co-registered multiparametric information of the tissues of interest. This review aims to describe the multi-faceted myocardial and hepatic tissue alterations in cardiometabolic disease and to motivate the application of relaxometry and proton-density cardiac and liver tissue mapping techniques. Current approaches in myocardial and liver tissue characterization as well as latest technical developments in multiparametric quantitative MRI are included. Limitations and challenges of these novel approaches, and recommendations to facilitate clinical validation are also discussed.
Collapse
Affiliation(s)
- Anastasia Fotaki
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom,*Correspondence: Anastasia Fotaki,
| | - Carlos Velasco
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Claudia Prieto
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom,School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile,Institute for Biological and Medical Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile,Millennium Institute for Intelligent Healthcare Engineering, Santiago, Chile
| | - René M. Botnar
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom,School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile,Institute for Biological and Medical Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile,Millennium Institute for Intelligent Healthcare Engineering, Santiago, Chile
| |
Collapse
|
48
|
Genetic lineage tracing identifies cardiac mesenchymal-to-adipose transition in an arrhythmogenic cardiomyopathy model. SCIENCE CHINA. LIFE SCIENCES 2023; 66:51-66. [PMID: 36322324 DOI: 10.1007/s11427-022-2176-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/09/2022] [Indexed: 11/05/2022]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is one of the most common inherited cardiomyopathies, characterized by progressive fibrofatty replacement in the myocardium. However, the cellular origin of cardiac adipocytes in ACM remains largely unknown. Unraveling the cellular source of cardiac adipocytes in ACM would elucidate the underlying pathological process and provide a potential target for therapy. Herein, we generated an ACM mouse model by inactivating desmosomal gene desmoplakin in cardiomyocytes; and examined the adipogenic fates of several cell types in the disease model. The results showed that SOX9+, PDGFRa+, and PDGFRb+ mesenchymal cells, but not cardiomyocytes or smooth muscle cells, contribute to the intramyocardial adipocytes in the ACM model. Mechanistically, Bmp4 was highly expressed in the ACM mouse heart and functionally promoted cardiac mesenchymal-to-adipose transition in vitro.
Collapse
|
49
|
Dong X, Strudwick M, Wang WY, Borlaug BA, van der Geest RJ, Ng AC, Delgado V, Bax JJ, Ng AC. Impact of body mass index and diabetes on myocardial fat content, interstitial fibrosis and function. Int J Cardiovasc Imaging 2023; 39:379-390. [PMID: 36306044 PMCID: PMC9870836 DOI: 10.1007/s10554-022-02723-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/30/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE We hypothesize that both increased myocardial steatosis and interstitial fibrosis contributes to subclinical myocardial dysfunction in patients with increased body mass index and diabetes mellitus. BACKGROUND Increased body weight and diabetes mellitus are both individually associated with a higher incidence of heart failure with preserved ejection fraction. However, it is unclear how increased myocardial steatosis and interstitial fibrosis interact to influence myocardial composition and function. METHODS A total of 100 subjects (27 healthy lean volunteers, 21 healthy but overweight volunteers, and 52 asymptomatic overweight patients with diabetes) were prospectively recruited to measure left ventricular (LV) myocardial steatosis (LV-myoFat) and interstitial fibrosis (by extracellular volume [ECV]) using magnetic resonance imaging, and then used to determine their combined impact on LV global longitudinal strain (GLS) analysis by 2-dimensional (2D) speckle tracking echocardiography on the same day. RESULTS On multivariable analysis, both increased body mass index and diabetes were independently associated with increased LV-myoFat. In turn, increased LV-myoFat was independently associated with increased LV ECV. Both increased LV-myoFat and LV ECV were independently associated with impaired 2D LV GLS. CONCLUSION Patients with increased body weight and patients with diabetes display excessive myocardial steatosis, which is related to a greater burden of myocardial interstitial fibrosis. LV myocardial contractile function was determined by both the extent of myocardial steatosis and interstitial fibrosis, and was independent of increasing age. Further study is warranted to determine how weight loss and improved diabetes management can improve myocardial composition and function.
Collapse
Affiliation(s)
- Xin Dong
- School of Exercise and Nutrition Sciences, Queensland University of Technology, Brisbane, Australia
| | - Mark Strudwick
- Centre for Advanced Imaging, The University of Queensland, Queensland, Australia
| | - William Ys Wang
- Centre for Advanced Imaging, The University of Queensland, Queensland, Australia
- Department of Cardiology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Rob J van der Geest
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Austin Cc Ng
- Department of Cardiology, Concord Hospital, The University of Sydney, Concord, NSW, Australia
| | - Victoria Delgado
- Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jeroen J Bax
- Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands.
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Arnold Ct Ng
- Centre for Advanced Imaging, The University of Queensland, Queensland, Australia
- Department of Cardiology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
- Faculty of Medicine, South Western Sydney Clinical School, The University of New South Wales, Warwick Farm, Australia
| |
Collapse
|
50
|
Impact of Dysfunctional Adipose Tissue Depots on the Cardiovascular System. Int J Mol Sci 2022; 23:ijms232214296. [PMID: 36430774 PMCID: PMC9695168 DOI: 10.3390/ijms232214296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Obesity with its associated complications represents a social, economic and health problem of utmost importance worldwide. Specifically, obese patients carry a significantly higher risk of developing cardiovascular disease compared to nonobese individuals. Multiple molecular mechanisms contribute to the impaired biological activity of the distinct adipose tissue depots in obesity, including secretion of proinflammatory mediators and reactive oxygen species, ultimately leading to an unfavorable impact on the cardiovascular system. This review summarizes data relating to the contribution of the main adipose tissue depots, including both remote (i.e., intra-abdominal, hepatic, skeletal, pancreatic, renal, and mesenteric adipose fat), and cardiac (i.e., the epicardial fat) adipose locations, on the cardiovascular system. Finally, we discuss both pharmacological and non-pharmacological strategies aimed at reducing cardiovascular risk through acting on adipose tissues, with particular attention to the epicardial fat.
Collapse
|