1
|
Ding Y, Lin F, Liu Z, Zhou X, Liang X. Targeting Epicardial/Pericardial Adipose Tissue in Cardiovascular Diseases: A Novel Therapeutic Strategy. Rev Cardiovasc Med 2025; 26:26128. [PMID: 40160564 PMCID: PMC11951288 DOI: 10.31083/rcm26128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/01/2024] [Accepted: 11/14/2024] [Indexed: 04/02/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain a global health concern, prompting ongoing research into novel contributors to their pathogenesis. Due to the proximity of the coronary arteries and the myocardium in epicardial adipose tissue (EAT) and pericardial adipose tissue (PAT), these tissues have emerged as key areas of interest for their potential influence on cardiac function and vascular health. This review synthesizes current research on the physiological and biological characteristics of EAT and PAT, exploring their composition and clinical measurement approaches. The roles of EAT and PAT in coronary artery disease (CAD), atrial fibrillation, and heart failure are discussed, and the contributions of EAT and PAT to these cardiovascular conditions are highlighted alongside their potential as therapeutic targets.
Collapse
Affiliation(s)
- Yue Ding
- Department of Organ Transplantation, Changzheng Hospital, Second Military Medical University, 200003 Shanghai, China
| | - Fang Lin
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Zhongmin Liu
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Xiaohui Zhou
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Xiaoting Liang
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| |
Collapse
|
2
|
Gaborit B, Julla JB, Fournel J, Ancel P, Soghomonian A, Deprade C, Lasbleiz A, Houssays M, Ghattas B, Gascon P, Righini M, Matonti F, Venteclef N, Potier L, Gautier JF, Resseguier N, Bartoli A, Mourre F, Darmon P, Jacquier A, Dutour A. Fully automated epicardial adipose tissue volume quantification with deep learning and relationship with CAC score and micro/macrovascular complications in people living with type 2 diabetes: the multicenter EPIDIAB study. Cardiovasc Diabetol 2024; 23:328. [PMID: 39227844 PMCID: PMC11373274 DOI: 10.1186/s12933-024-02411-y] [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: 07/01/2024] [Accepted: 08/19/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND The aim of this study (EPIDIAB) was to assess the relationship between epicardial adipose tissue (EAT) and the micro and macrovascular complications (MVC) of type 2 diabetes (T2D). METHODS EPIDIAB is a post hoc analysis from the AngioSafe T2D study, which is a multicentric study aimed at determining the safety of antihyperglycemic drugs on retina and including patients with T2D screened for diabetic retinopathy (DR) (n = 7200) and deeply phenotyped for MVC. Patients included who had undergone cardiac CT for CAC (Coronary Artery Calcium) scoring after inclusion (n = 1253) were tested with a validated deep learning segmentation pipeline for EAT volume quantification. RESULTS Median age of the study population was 61 [54;67], with a majority of men (57%) a median duration of the disease 11 years [5;18] and a mean HbA1c of7.8 ± 1.4%. EAT was significantly associated with all traditional CV risk factors. EAT volume significantly increased with chronic kidney disease (CKD vs no CKD: 87.8 [63.5;118.6] vs 82.7 mL [58.8;110.8], p = 0.008), coronary artery disease (CAD vs no CAD: 112.2 [82.7;133.3] vs 83.8 mL [59.4;112.1], p = 0.0004, peripheral arterial disease (PAD vs no PAD: 107 [76.2;141] vs 84.6 mL[59.2; 114], p = 0.0005 and elevated CAC score (> 100 vs < 100 AU: 96.8 mL [69.1;130] vs 77.9 mL [53.8;107.7], p < 0.0001). By contrast, EAT volume was neither associated with DR, nor with peripheral neuropathy. We further evidenced a subgroup of patients with high EAT volume and a null CAC score. Interestingly, this group were more likely to be composed of young women with a high BMI, a lower duration of T2D, a lower prevalence of microvascular complications, and a higher inflammatory profile. CONCLUSIONS Fully-automated EAT volume quantification could provide useful information about the risk of both renal and macrovascular complications in T2D patients.
Collapse
Affiliation(s)
- Bénédicte Gaborit
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France.
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France.
| | - Jean Baptiste Julla
- IMMEDIAB Laboratory, Institut Necker Enfants Malades (INEM), CNRS UMR 8253, INSERM U1151, Université Paris Cité, 75015, Paris, France
- Diabetology and Endocrinology Department, Féderation de Diabétologie, Université Paris Cité, Lariboisière Hospital, APHP, 75015, Paris, France
| | | | - Patricia Ancel
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Astrid Soghomonian
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| | - Camille Deprade
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| | - Adèle Lasbleiz
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France
| | - Marie Houssays
- Medical Evaluation Department, Assistance-Publique Hôpitaux de Marseille, CIC-CPCET, 13005, Marseille, France
| | - Badih Ghattas
- Aix Marseille School of Economics, Aix Marseille University, CNRS, Marseille, France
| | - Pierre Gascon
- Centre Monticelli Paradis, 433 Bis Rue Paradis, 13008, Marseille, France
| | - Maud Righini
- Ophtalmology Department, Assistance-Publique Hôpitaux de Marseille, Aix-Marseille Univ, 13005, Marseille, France
| | - Frédéric Matonti
- Centre Monticelli Paradis, 433 Bis Rue Paradis, 13008, Marseille, France
- National Center for Scientific Research (CNRS), Timone Neuroscience Institute (INT), Aix Marseille Univ, 13008, Marseille, France
| | - Nicolas Venteclef
- IMMEDIAB Laboratory, Institut Necker Enfants Malades (INEM), CNRS UMR 8253, INSERM U1151, Université Paris Cité, 75015, Paris, France
| | - Louis Potier
- IMMEDIAB Laboratory, Institut Necker Enfants Malades (INEM), CNRS UMR 8253, INSERM U1151, Université Paris Cité, 75015, Paris, France
- Diabetology and Endocrinology Department, Fédération de Diabétologie, Bichat Hospital, Paris, France
| | - Jean François Gautier
- IMMEDIAB Laboratory, Institut Necker Enfants Malades (INEM), CNRS UMR 8253, INSERM U1151, Université Paris Cité, 75015, Paris, France
- Diabetology and Endocrinology Department, Féderation de Diabétologie, Université Paris Cité, Lariboisière Hospital, APHP, 75015, Paris, France
| | - Noémie Resseguier
- Support Unit for Clinical Research and Economic Evaluation, Assistance Publique-Hôpitaux de Marseille, 13385, Marseille, France
- Aix-Marseille Univ, EA 3279 CEReSS-Health Service Research and Quality of Life Center, Marseille, France
| | - Axel Bartoli
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France
- Department of Radiology, Hôpital de la TIMONE, AP-HM, Marseille, France
| | - Florian Mourre
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| | - Patrice Darmon
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| | - Alexis Jacquier
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France
- Department of Radiology, Hôpital de la TIMONE, AP-HM, Marseille, France
| | - Anne Dutour
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, Chemin des Bourrely, APHM, Hôpital Nord, 13915 Marseille Cedex 20, Marseille, France
| |
Collapse
|
3
|
Galeone A, Annicchiarico A, Buccoliero C, Barile B, Luciani GB, Onorati F, Nicchia GP, Brunetti G. Diabetic Cardiomyopathy: Role of Cell Death, Exosomes, Fibrosis and Epicardial Adipose Tissue. Int J Mol Sci 2024; 25:9481. [PMID: 39273428 PMCID: PMC11395197 DOI: 10.3390/ijms25179481] [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: 07/31/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) represents one of the typical complications associated with diabetes. It has been described as anomalies in heart function and structure, with consequent high morbidity and mortality. DCM development can be described by two stages; the first is characterized by left ventricular hypertrophy and diastolic dysfunction, and the second by heart failure (HF) with systolic dysfunction. The proposed mechanisms involve cardiac inflammation, advanced glycation end products (AGEs) and angiotensin II. Furthermore, different studies have focused their attention on cardiomyocyte death through the different mechanisms of programmed cell death, such as apoptosis, autophagy, necrosis, pyroptosis and ferroptosis. Exosome release, adipose epicardial tissue and aquaporins affect DCM development. This review will focus on the description of the mechanisms involved in DCM progression and development.
Collapse
Affiliation(s)
- Antonella Galeone
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Alessia Annicchiarico
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Cinzia Buccoliero
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Barbara Barile
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Giovanni Battista Luciani
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Francesco Onorati
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Division of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| |
Collapse
|
4
|
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
|
5
|
Dogan N, Ozuynuk-Ertugrul AS, Balkanay OO, Yildiz CE, Guclu-Geyik F, Kirsan CB, Coban N. Examining the effects of coronary artery disease- and mitochondrial biogenesis-related genes' and microRNAs' expression levels on metabolic disorders in epicardial adipose tissue. Gene 2024; 895:147988. [PMID: 37977322 DOI: 10.1016/j.gene.2023.147988] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND AND AIMS Epicardial adipose tissue (EAT) surrounds the heart and coronary arteries and is important for comprehending the pathogenesis of coronary artery disease (CAD). We aimed to evaluate the expressions of mitochondrial biogenesis- and CAD-related genes and miRNAs in EAT by comparing them to visceral adipose tissue (VAT) in CAD, diabetes, and obesity subgroups. METHODS In this study, a total of 93 individuals were recruited, and EAT samples (63 CAD; 30 non-CAD) and VAT samples from 65 individuals (46 CAD; 19 non-CAD) were collected. For further analysis, the study population was divided according to obesity and diabetes status. PRKAA1, PPARGC1A, SIRT1, RELA, TNFA, and miR-155-5p, let-7g-5p, miR-1247-5p, miR-326 expression levels were examined. RESULTS PRKAA1 and let-7g-5p were differentially expressed in EAT compared to VAT. TNFA expression was upregulated significantly in both tissues of CAD patients. In EAT, PRKAA1, PPARGC1A, and SIRT1 were downregulated with diabetes. Moreover, PPARGC1A expression is decreased under the condition of obesity in both tissues. EAT expressions of miR-1247-5p and miR-326 were downregulated with obesity, while miR-155-5p is decreased only in the VAT of obese. Also, miRNAs and genes were correlated with biochemical parameters and each other in EAT and VAT (p < 0.050). CONCLUSIONS The findings demonstrating distinct let-7g-5p and AMPKα1 mRNA expression between EAT and VAT underscores the importance of tissue-specific regulation in different clinical outcomes. In addition, the differential expressions of investigated genes and miRNAs highlight their responsiveness to obesity, DM, and CAD in adipose tissues.
Collapse
Affiliation(s)
- Nazli Dogan
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Istanbul University Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Aybike S Ozuynuk-Ertugrul
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Istanbul University Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Ozan O Balkanay
- Department of Cardiovascular Surgery, Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Cenk E Yildiz
- Department of Cardiovascular Surgery, Institute of Cardiology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Filiz Guclu-Geyik
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Cemre B Kirsan
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Istanbul University Institute of Graduate Studies in Health Sciences, Istanbul, Turkey
| | - Neslihan Coban
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
6
|
Liu Y, Han L, Zhu P, Song M, Zhang Y, Meng L, Zhang W, Zhang C, Zhong M. PTPN2 targets TAK1 for dephosphorylation to improve cellular senescence and promote adipose tissue browning in T2DM. Front Pharmacol 2023; 14:1124633. [PMID: 37251330 PMCID: PMC10213551 DOI: 10.3389/fphar.2023.1124633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/19/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction: The energy imbalance when energy intake exceeds expenditure acts as an essential factor in the development of insulin resistance (IR). The activity of brown adipose tissue, which is involved in the dissipation of energy via heat expenditure decreases under type 2 diabetic mellitus (T2DM) state when the number of pathological aging adipocytes increases. Protein tyrosine phosphatase non-receptor type 2 (PTPN2) regulates several biological processes by dephosphorylating several cellular substrates; however, whether PTPN2 regulates cellular senescence in adipocytes and the underlying mechanism has not been reported. Methods: We constructed a model of type 2 diabetic mice with PTPN2 overexpression to explore the role of PTPN2 in T2DM. Results: We revealed that PTPN2 facilitated adipose tissue browning by alleviating pathological senescence, thus improving glucose tolerance and IR in T2DM. Mechanistically, we are the first to report that PTPN2 could bind with transforming growth factor-activated kinase 1 (TAK1) directly for dephosphorylation to inhibit the downstream MAPK/NF-κB pathway in adipocytes and regulate cellular senescence and the browning process subsequently. Discussion: Our study revealed a critical mechanism of adipocytes browning progression and provided a potential target for the treatment of related diseases.
Collapse
Affiliation(s)
- Yapeng Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lu Han
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of General Practice, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ping Zhu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ming Song
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yaoyuan Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Linlin Meng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
7
|
Martín FM, Alzamendi A, Harnichar AE, Castrogiovanni D, Zubiría MG, Spinedi E, Giovambattista A. Role of glucocorticoid receptor (GR) in white adipose tissue beiging. Life Sci 2023; 322:121681. [PMID: 37040814 DOI: 10.1016/j.lfs.2023.121681] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/27/2023] [Accepted: 04/04/2023] [Indexed: 04/13/2023]
Abstract
AIM Glucocorticoids (GCs) play a crucial role in energy homeostasis including white adipose tissue function; however, chronic GC excess is detrimental to mammals' health. White hypertrophic adiposity is a main factor for neuroendocrine-metabolic dysfunctions in monosodium L-glutamate (MSG)-damaged hypercorticosteronemic rat. Nevertheless, little is known about the receptor path in endogenous GC impact on white adipose tissue-resident precursor cells to bring them into beige lineage. Thus, our aim was to explore whether transient/chronic endogenous hypercorticosteronemia affects browning capacity in white adipose tissue pads from MSG rats during development. MAIN METHODS Control and MSG male rats aged 30 and 90 days were 7-day exposed to cold conditions in order to stimulate wet white epidydimal adipose tissue (wEAT) beiging capacity. This procedure was also replicated in adrenalectomized rats. KEY FINDINGS Data indicated that whereas epidydimal white adipose tissue pads from prepubertal hypercorticosteronemic rats retained full expression of GR/MR genes resulting in a drastic reduction in wEAT beiging capacity, conversely, chronic hypercorticosteronemic adult MSG rats developed down-regulation of corticoid genes (and reduced GR cytosolic mediators) in wEAT pads and consequently partially restored local beiging capacity. Finally, wEAT pads from adrenalectomized rats revealed up-regulation of GR gene accompanied by full local beiging capacity. SIGNIFICANCE This study strongly supports a GR-dependent inhibitory effect of GC excess on white adipose tissue browning, an issue strongly supporting a key role of GR in the non-shivering thermogenic process. As a consequence, normalizing the GC milieu could be a relevant factor to handle dysmetabolism in white hyperadipose phenotypes.
Collapse
Affiliation(s)
- Florencia M Martín
- Centro de Endocrinología Experimental y Aplicada (CENEXA, UNLP-CONICET-CICPBA-CA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 1900 La Plata, Argentina; Laboratorio de Neuroendocrinología, Instituto Multidisciplinario de Biología Celular (IMBICE, CICPBA-CONICET-UNLP), 1900 La Plata, Argentina
| | - Ana Alzamendi
- Laboratorio de Neuroendocrinología, Instituto Multidisciplinario de Biología Celular (IMBICE, CICPBA-CONICET-UNLP), 1900 La Plata, Argentina
| | - Alejandro E Harnichar
- Laboratorio de Neuroendocrinología, Instituto Multidisciplinario de Biología Celular (IMBICE, CICPBA-CONICET-UNLP), 1900 La Plata, Argentina
| | - Daniel Castrogiovanni
- Laboratorio de Neuroendocrinología, Instituto Multidisciplinario de Biología Celular (IMBICE, CICPBA-CONICET-UNLP), 1900 La Plata, Argentina
| | - María Guillermina Zubiría
- Laboratorio de Neuroendocrinología, Instituto Multidisciplinario de Biología Celular (IMBICE, CICPBA-CONICET-UNLP), 1900 La Plata, Argentina
| | - Eduardo Spinedi
- Centro de Endocrinología Experimental y Aplicada (CENEXA, UNLP-CONICET-CICPBA-CA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 1900 La Plata, Argentina.
| | - Andrés Giovambattista
- Laboratorio de Neuroendocrinología, Instituto Multidisciplinario de Biología Celular (IMBICE, CICPBA-CONICET-UNLP), 1900 La Plata, Argentina
| |
Collapse
|
8
|
AlZaim I, Eid AH, Abd-Elrahman KS, El-Yazbi AF. Adipose Tissue Mitochondrial Dysfunction and Cardiometabolic Diseases: On the Search for Novel Molecular Targets. Biochem Pharmacol 2022; 206:115337. [DOI: 10.1016/j.bcp.2022.115337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
9
|
Krishnan A, Sharma H, Yuan D, Trollope AF, Chilton L. The Role of Epicardial Adipose Tissue in the Development of Atrial Fibrillation, Coronary Artery Disease and Chronic Heart Failure in the Context of Obesity and Type 2 Diabetes Mellitus: A Narrative Review. J Cardiovasc Dev Dis 2022; 9:jcdd9070217. [PMID: 35877579 PMCID: PMC9318726 DOI: 10.3390/jcdd9070217] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 12/07/2022] Open
Abstract
Cardiovascular diseases (CVDs) are a significant burden globally and are especially prevalent in obese and/or diabetic populations. Epicardial adipose tissue (EAT) surrounding the heart has been implicated in the development of CVDs as EAT can shift from a protective to a maladaptive phenotype in diseased states. In diabetic and obese patients, an elevated EAT mass both secretes pro-fibrotic/pro-inflammatory adipokines and forms intramyocardial fibrofatty infiltrates. This narrative review considers the proposed pathophysiological roles of EAT in CVDs. Diabetes is associated with a disordered energy utilization in the heart, which promotes intramyocardial fat and structural remodeling. Fibrofatty infiltrates are associated with abnormal cardiomyocyte calcium handling and repolarization, increasing the probability of afterdepolarizations. The inflammatory phenotype also promotes lateralization of connexin (Cx) proteins, undermining unidirectional conduction. These changes are associated with conduction heterogeneity, together creating a substrate for atrial fibrillation (AF). EAT is also strongly implicated in coronary artery disease (CAD); inflammatory adipokines from peri-vascular fat can modulate intra-luminal homeostasis through an “outside-to-inside” mechanism. EAT is also a significant source of sympathetic neurotransmitters, which promote progressive diastolic dysfunction with eventual cardiac failure. Further investigations on the behavior of EAT in diabetic/obese patients with CVD could help elucidate the pathogenesis and uncover potential therapeutic targets.
Collapse
Affiliation(s)
- Anirudh Krishnan
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; (A.K.); (H.S.); (D.Y.)
| | - Harman Sharma
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; (A.K.); (H.S.); (D.Y.)
| | - Daniel Yuan
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; (A.K.); (H.S.); (D.Y.)
| | - Alexandra F. Trollope
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia;
| | - Lisa Chilton
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD 4811, Australia
- Correspondence:
| |
Collapse
|
10
|
Mishra BK, Madhu SV, Aslam M, Agarwal V, Banerjee BD. Adipose tissue expression of UCP1 and PRDM16 genes and their association with postprandial triglyceride metabolism and glucose intolerance. Diabetes Res Clin Pract 2021; 182:109115. [PMID: 34718051 DOI: 10.1016/j.diabres.2021.109115] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 08/28/2021] [Accepted: 10/15/2021] [Indexed: 12/01/2022]
Abstract
AIMS UCP1 and PRDM16 genes, primarily involved in browning of adipose tissue that can affect lipid metabolism are also associated with diabetes risk. Therefore, we planned to study the adipose tissue expression of UCP1 and PRDM 16 genes in subjects with glucose intolerance to find out its association with postprandial triglyceride (PPTg) measures and T2DM. METHODS A total of 30 subjects were recruited in three groups i.e., NGT, prediabetes and T2DM (NDDM + known T2DM) who were matched for age, sex and BMI. An 8-hour standardized fat challenge test was performed to study lipemic responses. UCP1 and PRDM16 genes quantification in adipose tissue was performed by real-time PCR followed by SDS PAGE. RESULTS UCP1 gene expression in SAT was significantly lower in T2DM and prediabetes as compared to NGT group while PRDM16 gene expression was significantly lower in T2DM group as compared to NGT group. UCP1 gene expression correlated with PPTg measures as well as with glycaemic measures while PRDM16 gene expression correlated with glycaemic measures only. CONCLUSION This study found downregulation of PRDM16 and UCP1 gene expression in SAT in subjects with glucose intolerance. The association of UCP1 gene expression with PPTg dysmetabolism may contribute to greater predisposition to T2DM.
Collapse
Affiliation(s)
- B K Mishra
- Department of Endocrinology, University College of Medical Sciences & GTB Hospital, University of Delhi, India
| | - S V Madhu
- Department of Endocrinology, University College of Medical Sciences & GTB Hospital, University of Delhi, India.
| | - M Aslam
- Department of Endocrinology, University College of Medical Sciences & GTB Hospital, University of Delhi, India
| | - V Agarwal
- Department of Surgery, University College of Medical Sciences & GTB Hospital, University of Delhi, India
| | - B D Banerjee
- Department of Biochemistry, University College of Medical Sciences & GTB Hospital, University of Delhi, India
| |
Collapse
|
11
|
Omidifar A, Shirvani H, Taheri RA, Gorgani-Firouzjae S, Delfan M, Kalaki-Jouybari F, Khakdan S. Protective effects of HIIT vs. CET exercise training on high-fat-high-fructose diet-induced hyperglycemia, hyperlipidemia, and histopathology of liver in rats: regulation of SIRT1/PGC-1α. SPORT SCIENCES FOR HEALTH 2021. [DOI: 10.1007/s11332-021-00736-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
12
|
Rodriguez-Cuenca S, Lelliot CJ, Campbell M, Peddinti G, Martinez-Uña M, Ingvorsen C, Dias AR, Relat J, Mora S, Hyötyläinen T, Zorzano A, Orešič M, Bjursell M, Bohlooly-Y M, Lindén D, Vidal-Puig A. Allostatic hypermetabolic response in PGC1α/β heterozygote mouse despite mitochondrial defects. FASEB J 2021; 35:e21752. [PMID: 34369602 DOI: 10.1096/fj.202100262rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022]
Abstract
Aging, obesity, and insulin resistance are associated with low levels of PGC1α and PGC1β coactivators and defective mitochondrial function. We studied mice deficient for PGC1α and PGC1β [double heterozygous (DH)] to investigate their combined pathogenic contribution. Contrary to our hypothesis, DH mice were leaner, had increased energy dissipation, a pro-thermogenic profile in BAT and WAT, and improved carbohydrate metabolism compared to wild types. WAT showed upregulation of mitochondriogenesis/oxphos machinery upon allelic compensation of PGC1α4 from the remaining allele. However, DH mice had decreased mitochondrial OXPHOS and biogenesis transcriptomes in mitochondria-rich organs. Despite being metabolically healthy, mitochondrial defects in DH mice impaired muscle fiber remodeling and caused qualitative changes in the hepatic lipidome. Our data evidence first the existence of organ-specific compensatory allostatic mechanisms are robust enough to drive an unexpected phenotype. Second, optimization of adipose tissue bioenergetics is sufficient to maintain a healthy metabolic phenotype despite a broad severe mitochondrial dysfunction in other relevant metabolic organs. Third, the decrease in PGC1s in adipose tissue of obese and diabetic patients is in contrast with the robustness of the compensatory upregulation in the adipose of the DH mice.
Collapse
Affiliation(s)
| | | | - Mark Campbell
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Gopal Peddinti
- VTT, Technical Research Center of Finland, Espoo, Finland
| | - Maite Martinez-Uña
- Department of Physiology, University of the Basque Country UPV/EHU, Bilbao, Spain
| | - Camilla Ingvorsen
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Ana Rita Dias
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Joana Relat
- Department of Nutrition, Food Science and Gastronomy, School of Pharmacy and Food Science, Food and Nutrition Torribera Campus, University of Barcelona (UB), Santa Coloma de Gramenet, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Barcelona, Spain
| | - Silvia Mora
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, The University of Liverpool, Liverpool, UK
| | | | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Dept. Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Matej Orešič
- School of Science and Technology, Örebro University, Örebro, Sweden
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Mikael Bjursell
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Daniel Lindén
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Division of Endocrinology, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Antonio Vidal-Puig
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| |
Collapse
|
13
|
Sjögren RJO, Rizo-Roca D, Chibalin AV, Chorell E, Furrer R, Katayama S, Harada J, Karlsson HKR, Handschin C, Moritz T, Krook A, Näslund E, Zierath JR. Branched-chain amino acid metabolism is regulated by ERRα in primary human myotubes and is further impaired by glucose loading in type 2 diabetes. Diabetologia 2021; 64:2077-2091. [PMID: 34131782 PMCID: PMC8382616 DOI: 10.1007/s00125-021-05481-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/12/2021] [Indexed: 11/26/2022]
Abstract
AIMS/HYPOTHESIS Increased levels of branched-chain amino acids (BCAAs) are associated with type 2 diabetes pathogenesis. However, most metabolomic studies are limited to an analysis of plasma metabolites under fasting conditions, rather than the dynamic shift in response to a metabolic challenge. Moreover, metabolomic profiles of peripheral tissues involved in glucose homeostasis are scarce and the transcriptomic regulation of genes involved in BCAA catabolism is partially unknown. This study aimed to identify differences in circulating and skeletal muscle BCAA levels in response to an OGTT in individuals with normal glucose tolerance (NGT) or type 2 diabetes. Additionally, transcription factors involved in the regulation of the BCAA gene set were identified. METHODS Plasma and vastus lateralis muscle biopsies were obtained from individuals with NGT or type 2 diabetes before and after an OGTT. Plasma and quadriceps muscles were harvested from skeletal muscle-specific Ppargc1a knockout and transgenic mice. BCAA-related metabolites and genes were assessed by LC-MS/MS and quantitative RT-PCR, respectively. Small interfering RNA and adenovirus-mediated overexpression techniques were used in primary human skeletal muscle cells to study the role of PPARGC1A and ESRRA in the expression of the BCAA gene set. Radiolabelled leucine was used to analyse the impact of oestrogen-related receptor α (ERRα) knockdown on leucine oxidation. RESULTS Impairments in BCAA catabolism in people with type 2 diabetes under fasting conditions were exacerbated after a glucose load. Branched-chain keto acids were reduced 37-56% after an OGTT in the NGT group, whereas no changes were detected in individuals with type 2 diabetes. These changes were concomitant with a stronger correlation with glucose homeostasis biomarkers and downregulated expression of branched-chain amino acid transaminase 2, branched-chain keto acid dehydrogenase complex subunits and 69% of downstream BCAA-related genes in skeletal muscle. In primary human myotubes overexpressing peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α, encoded by PPARGC1A), 61% of the analysed BCAA genes were upregulated, while 67% were downregulated in the quadriceps of skeletal muscle-specific Ppargc1a knockout mice. ESRRA (encoding ERRα) silencing completely abrogated the PGC-1α-induced upregulation of BCAA-related genes in primary human myotubes. CONCLUSIONS/INTERPRETATION Metabolic inflexibility in type 2 diabetes impacts BCAA homeostasis and attenuates the decrease in circulating and skeletal muscle BCAA-related metabolites after a glucose challenge. Transcriptional regulation of BCAA genes in primary human myotubes via PGC-1α is ERRα-dependent.
Collapse
Affiliation(s)
- Rasmus J O Sjögren
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - David Rizo-Roca
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | | | - Shintaro Katayama
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Jun Harada
- Cardiovascular-Metabolics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Håkan K R Karlsson
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | | | - Thomas Moritz
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Erik Näslund
- Division of Surgery, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
14
|
Shi Z, Wang S, Deng J, Gong Z. PGC-1α attenuates the oxidative stress-induced impaired osteogenesis and angiogenesis regulation effects of mesenchymal stem cells in the presence of diabetic serum. Biochem Biophys Rep 2021; 27:101070. [PMID: 34286110 PMCID: PMC8278528 DOI: 10.1016/j.bbrep.2021.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 11/30/2022] Open
Abstract
Oxidative stress is believed to induce dysfunction of the bone remodeling process and be associated with progressive loss of bone mass. The peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) is a master controller during mitochondrial biogenesis and the antioxidant response. We postulated that PGC-1α could function as a cyto-protective effector in mesenchymal stem cells (MSCs) under oxidative stress conditions. In this study, diabetic serum was firstly used to treat MSCs to induce oxidative damage. The anti-oxidative protective effects of PGC-1α overexpression on MSCs, as well as MSCs' osteogenesis and angiogenic regulation effects were investigated in vitro. Results showed that diabetic conditions induced significantly increase of intracellular oxidative damage and mitochondrial permeability transition pore (mPTP) opening activity, decrease of cellular viability, and osteogenic differentiation and pro-angiogenic regulation effects of MSCs. However, the diabetic conditions induced oxidative impair on MSCs were significantly alleviated via PGC-1α overexpression under diabetic conditions. Taken together, this study indicates the anti-oxidative treatment potential of PGC-1α regulation as a promising strategy to promote coupling pro-osteogenesis and pro-angiogenesis effects of MSCs.
Collapse
Affiliation(s)
- Zongxin Shi
- Department of Orthopedic Surgery, Liangxiang Hospital of Beijing Fangshan District, and Liangxiang Teaching Hospital of Capital Medical University, No.45, Gongchen Ave., Liangxiang, Fangshan Dist., Beijing, 102488, China
| | - Shikun Wang
- Department of Orthopedic Surgery, Liangxiang Hospital of Beijing Fangshan District, and Liangxiang Teaching Hospital of Capital Medical University, No.45, Gongchen Ave., Liangxiang, Fangshan Dist., Beijing, 102488, China
| | - Jiechao Deng
- Department of Orthopedic Surgery, Liangxiang Hospital of Beijing Fangshan District, and Liangxiang Teaching Hospital of Capital Medical University, No.45, Gongchen Ave., Liangxiang, Fangshan Dist., Beijing, 102488, China
| | - Zishun Gong
- Department of Orthopedic Surgery, Liangxiang Hospital of Beijing Fangshan District, and Liangxiang Teaching Hospital of Capital Medical University, No.45, Gongchen Ave., Liangxiang, Fangshan Dist., Beijing, 102488, China
| |
Collapse
|
15
|
Role of PGC-1α in the Mitochondrial NAD + Pool in Metabolic Diseases. Int J Mol Sci 2021; 22:ijms22094558. [PMID: 33925372 PMCID: PMC8123861 DOI: 10.3390/ijms22094558] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondria play vital roles, including ATP generation, regulation of cellular metabolism, and cell survival. Mitochondria contain the majority of cellular nicotinamide adenine dinucleotide (NAD+), which an essential cofactor that regulates metabolic function. A decrease in both mitochondria biogenesis and NAD+ is a characteristic of metabolic diseases, and peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) orchestrates mitochondrial biogenesis and is involved in mitochondrial NAD+ pool. Here we discuss how PGC-1α is involved in the NAD+ synthesis pathway and metabolism, as well as the strategy for increasing the NAD+ pool in the metabolic disease state.
Collapse
|
16
|
Sasson A, Kristoferson E, Batista R, McClung JA, Abraham NG, Peterson SJ. The pivotal role of heme Oxygenase-1 in reversing the pathophysiology and systemic complications of NAFLD. Arch Biochem Biophys 2020; 697:108679. [PMID: 33248947 DOI: 10.1016/j.abb.2020.108679] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/03/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023]
Abstract
The pathogenesis and molecular pathways involved in non-alcoholic fatty liver disease (NAFLD) are reviewed, as well as what is known about mitochondrial dysfunction that leads to heart disease and the progression to steatohepatitis and hepatic fibrosis. We focused our discussion on the role of the antioxidant gene heme oxygenase-1 (HO-1) and its nuclear coactivator, peroxisome proliferator-activated receptor-gamma coactivator (PGC1-α) in the regulation of mitochondrial biogenesis and function and potential therapeutic benefit for cardiac disease, NAFLD as well as the pharmacological effect they have on the chronic inflammatory state of obesity. The result is increased mitochondrial function and the conversion of white adipocyte tissue to beige adipose tissue ("browning of white adipose tissue") that leads to an improvement in signaling pathways and overall liver function. Improved mitochondrial biogenesis and function is essential to preventing the progression of hepatic steatosis to NASH and cirrhosis as well as preventing cardiovascular complications.
Collapse
Affiliation(s)
- Ariel Sasson
- Department of Medicine, New York Medical College, Valhalla, NY, 10595, USA; Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Eva Kristoferson
- Department of Medicine, New York Medical College, Valhalla, NY, 10595, USA
| | - Rogerio Batista
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - John A McClung
- Department of Medicine, New York Medical College, Valhalla, NY, 10595, USA
| | - Nader G Abraham
- Department of Medicine, New York Medical College, Valhalla, NY, 10595, USA; Department of Pharmacology, New York Medical College, Valhalla, NY, 10595, USA; Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25701, USA
| | - Stephen J Peterson
- Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA; New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY, 11215, USA.
| |
Collapse
|
17
|
Uddin SJ, Afroz M, Zihad SMNK, Rahman MS, Akter S, Khan IN, Al-Rabbi SMS, Rouf R, Islam MT, Shilpi JA, Nahar L, Tiralongo E, Sarker SD. A Systematic Review on Anti-diabetic and Cardioprotective Potential of Gallic Acid: A Widespread Dietary Phytoconstituent. FOOD REVIEWS INTERNATIONAL 2020. [DOI: 10.1080/87559129.2020.1734609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Shaikh Jamal Uddin
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Mohasana Afroz
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | | | - Md. Shamim Rahman
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Sanzida Akter
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Ishaq N. Khan
- PK-Neurooncology Research Group, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, 25000, Pakistan
| | | | - Razina Rouf
- Department of Pharmacy, Faculty of Life Science, Bangabandhu Sheikh Mujibur Rahman Science & Technology University, Gopalganj, Bangladesh
| | - Muhammad Torequl Islam
- Department of Pharmacy, Faculty of Life Science, Bangabandhu Sheikh Mujibur Rahman Science & Technology University, Gopalganj, Bangladesh
| | - Jamil A. Shilpi
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, Bangladesh
| | - Lutfun Nahar
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Byrom Street, LiverpoolL3 3AF, UK
| | - Evelin Tiralongo
- School of Pharmacy and Pharmacology & Menzies Health Institute Queensland, Griffith University, Southport, Qld, Australia
| | - Satyajit D. Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Byrom Street, LiverpoolL3 3AF, UK
| |
Collapse
|
18
|
Wang SY, Zhu S, Wu J, Zhang M, Xu Y, Xu W, Cui J, Yu B, Cao W, Liu J. Exercise enhances cardiac function by improving mitochondrial dysfunction and maintaining energy homoeostasis in the development of diabetic cardiomyopathy. J Mol Med (Berl) 2020; 98:245-261. [PMID: 31897508 DOI: 10.1007/s00109-019-01861-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022]
Abstract
Diabetic cardiomyopathy (DCM) is a major cause of morbidity and mortality in diabetic patients. Reactive oxygen species (ROS) produced by oxidative stress play an important role in the development of DCM. DCM involves abnormal energy metabolism, thereby reducing energy production. Exercise has been reported to be effective in protecting the heart against ROS accumulation during the development of DCM. We hypothesize that the AMPK/PGC-1α axis may play a crucial role in exercise-induced bioenergetic metabolism and aerobic respiration on oxidative stress parameters in the development of diabetic cardiomyopathy. Using a streptozotocin/high-fat diet mouse to generate a diabetic model, our aim was to evaluate the effects of exercise on the cardiac function, mitochondrial oxidative capacity, mitochondrial function, and cardiac expression of PGC-1α. Mice fed a high-fat diet were given MO-siPGC-1α or treated with AMPK inhibitor. Mitochondrial structure and effects of switching between the Warburg effect and aerobic respiration were analysed. Exercise improved blood pressure and systolic dysfunction in diabetic mouse hearts. The beneficial effects of exercise were also observed in a mitochondrial function study, as reflected by an enhanced oxidative phosphorylation level, increased membrane potential, and decreased ROS level and oxygen consumption. On the other hand, depletion of PGC-1α attenuated the effects of exercise on the enhancement of mitochondrial function. In addition, PGC-1α may be responsible for reversing the Warburg effect to aerobic respiration, thus enhancing mitochondrial metabolism and energy homoeostasis. In this study, we demonstrate the protective effects of exercise on shifting energy metabolism from fatty acid oxidation to glucose oxidation in an established diabetic stage. These data suggest that exercise is effective at ameliorating diabetic cardiomyopathy by improving mitochondrial function and reducing metabolic disturbances.
Collapse
Affiliation(s)
- Shawn Yongshun Wang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China.,Department of Biomedical Science, University of Hong Kong, Pokfulam, Hong Kong
| | - Siyu Zhu
- Department of Medical Imaging, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Jian Wu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Maomao Zhang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Yousheng Xu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Wei Xu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Jinjin Cui
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Bo Yu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China
| | - Wei Cao
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China. .,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China.
| | - Jingjin Liu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China. .,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, 150086, Heilongjiang, China. .,Department of Anesthesiology, University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
19
|
Christensen RH, von Scholten BJ, Lehrskov LL, Rossing P, Jørgensen PG. Epicardial adipose tissue: an emerging biomarker of cardiovascular complications in type 2 diabetes? Ther Adv Endocrinol Metab 2020; 11:2042018820928824. [PMID: 32518616 PMCID: PMC7252363 DOI: 10.1177/2042018820928824] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/03/2020] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes (T2D) is associated with an increased risk of cardiovascular disease and heart failure, which highlights the need for improved understanding of factors contributing to the pathophysiology of these complications as they are the leading cause of mortality in T2D. Patients with T2D have high levels of epicardial adipose tissue (EAT). EAT is known to secrete inflammatory factors, lipid metabolites, and has been proposed to apply mechanical stress on the cardiac muscle that may accelerate atherosclerosis, cardiac remodeling, and heart failure. High levels of EAT in patients with T2D have been associated with atherosclerosis, diastolic dysfunction, and incident cardiovascular events, and this fat depot has been suggested as an important link coupling diabetes, obesity, and cardiovascular disease. Despite this, the predictive potential of EAT in general, and in patients with diabetes, is yet to be established, and, up until now, the clinical relevance of EAT is therefore limited. Should this link be established, importantly, studies show that this fat depot can be modified both by pharmacological and lifestyle interventions. In this review, we first introduce the role of adipose tissue in T2D and present mechanisms involved in the pathophysiology of EAT and pericardial adipose tissue (PAT) in general, and in patients with T2D. Next, we summarize the evidence that these fat depots are elevated in patients with T2D, and discuss whether they might drive the high cardiometabolic risk in patients with T2D. Finally, we discuss the clinical potential of cardiac adipose tissues, address means to target this depot, and briefly touch upon underlying mechanisms and future research questions.
Collapse
Affiliation(s)
| | | | - Louise Lang Lehrskov
- Center for Inflammation and Metabolism/Center for Physical Activity Research, Rigshospitalet, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
20
|
The Expression/Methylation Profile of Adipogenic and Inflammatory Transcription Factors in Adipose Tissue Are Linked to Obesity-Related Colorectal Cancer. Cancers (Basel) 2019; 11:cancers11111629. [PMID: 31652933 PMCID: PMC6893417 DOI: 10.3390/cancers11111629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/04/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is well accepted as crucial risk factor that plays a critical role in the initiation and progression of colorectal cancer (CRC). More specifically, visceral adipose tissue (VAT) in people with obesity could produce chronic inflammation and an altered profile expression of key transcription factors that promote a favorable microenvironment to colorectal carcinogenesis. For this, the aim of this study was to explore the relationship between adipogenic and inflammatory transcription factors in VAT from nonobese, obese, and/or CRC patients. To test this idea, we studied the expression and methylation of CCAAT-enhancer binding protein type alpha (C/EBP-α), peroxisome proliferator-activated receptor gamma (PPAR-γ), peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α) and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) in VAT from non-obese control, non-obese CRC subjects, overweight/obese control, and overweight/obese CRC patients and their correlation with anthropometric and biochemical variables. We found decreased expression of C/EBP-α in overweight/obese CRC patients in comparison with overweight/obese control subjects. PGC-1α and NF-κB were overexpressed in CRC patients independently of the BMI. NF-κB promoter was hypomethylated in overweight/obese CRC patients when compared to overweight/obese control individuals. In addition, multiple significant correlations between expression, methylation, and biochemical parameters were found. Finally, linear regression analysis showed that the expression of C/EBP-α and NF-κB and that NF-κB methylation were associated with CRC and able to explain up to 55% of CRC variability. Our results suggest that visceral adipose tissue may be a key factor in tumor development and inflammatory state. We propose C/EBP-α, PGC-1α and NF-κB to be interesting candidates as potential biomarkers in adipose tissue for CRC patients.
Collapse
|
21
|
Role of epicardial adipose tissue NPR-C in acute coronary syndrome. Atherosclerosis 2019; 286:79-87. [DOI: 10.1016/j.atherosclerosis.2019.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/28/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022]
|
22
|
Caporarello N, Meridew JA, Jones DL, Tan Q, Haak AJ, Choi KM, Manlove LJ, Prakash YS, Tschumperlin DJ, Ligresti G. PGC1α repression in IPF fibroblasts drives a pathologic metabolic, secretory and fibrogenic state. Thorax 2019; 74:749-760. [PMID: 31182654 DOI: 10.1136/thoraxjnl-2019-213064] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/16/2019] [Accepted: 05/18/2019] [Indexed: 12/11/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal ageing-related disease linked to mitochondrial dysfunction. The present study aimed to determine whether peroxisome proliferator activated receptor gamma co-activator 1-alpha (PPARGC1A, encoding PGC1α), a master regulator of mitochondrial biogenesis, is diminished in IPF and controls pathologic fibroblast activation. Primary human IPF, control lung fibroblasts and fibroblasts sorted from bleomycin-injured mice were used to evaluate the expression and function of PGC1α. In vitro PGC1α manipulation was performed by small interfering RNA knockdown or overexpression. Fibroblast activation was assessed by quantitative PCR, Western blotting, matrix deposition, secreted cytokine array, immunofluorescence and traction force microscopy. Mitochondrial function was assessed by Seahorse analyzer and mitochondria mass and number by flow cytometry, mitochondrial DNA quantification and transmission electron microscopy (TEM). We found that PGC1α levels are stably repressed in IPF fibroblasts. After bleomycin injury in young mice, PGC1α expression drops transiently but then increases prior to fibrosis resolution. In contrast, PGC1α expression fails to recover in aged mice with persistent fibrosis. PGC1α knockdown alone in normal human lung fibroblasts reduces mitochondrial mass and function while enhancing contractile and matrix synthetic fibroblast activation, senescence-related gene expression and soluble profibrotic and prosenescence signalling. Re-expression of PGC1α in IPF fibroblasts ameliorates all of these pathological cellular functions. Pharmacological treatment of IPF fibroblasts with rosiglitazone, but not thyroid hormone, elevated PGC1α expression and attenuated fibroblast activation. The sustained repression of PGC1α and beneficial effects of its rescue in IPF fibroblasts identifies PGC1α as an important regulator of the fibroblast's pathological state in IPF.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Jeffrey A Meridew
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Dakota L Jones
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Qi Tan
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Andrew J Haak
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Kyoung M Choi
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Logan J Manlove
- Anesthesiology and Perioperative Medicine, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Y S Prakash
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA.,Anesthesiology and Perioperative Medicine, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Daniel J Tschumperlin
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Giovanni Ligresti
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| |
Collapse
|
23
|
Santiago-Fernández C, Pérez-Belmonte LM, Millán-Gómez M, Moreno-Santos I, Carrasco-Chinchilla F, Ruiz-Salas A, Morcillo-Hidalgo L, Melero JM, Garrido-Sánchez L, Jiménez-Navarro M. Overexpression of scavenger receptor and infiltration of macrophage in epicardial adipose tissue of patients with ischemic heart disease and diabetes. J Transl Med 2019; 17:95. [PMID: 30894181 PMCID: PMC6425581 DOI: 10.1186/s12967-019-1842-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/11/2019] [Indexed: 12/20/2022] Open
Abstract
Background Oxidized low-density lipoproteins and scavenger receptors (SRs) play an important role in the formation and development of atherosclerotic plaques. However, little is known about their presence in epicardial adipose tissue (EAT). The objective of the study was to evaluate the mRNA expression of different SRs in EAT of patients with ischemic heart disease (IHD), stratifying by diabetes status and its association with clinical and biochemical variables. Methods We analyzed the mRNA expression of SRs (LOX-1, MSR1, CXCL16, CD36 and CL-P1) and macrophage markers (CD68, CD11c and CD206) in EAT from 45 patients with IHD (23 with type 2 diabetes mellitus (T2DM) and 22 without T2DM) and 23 controls without IHD or T2DM. Results LOX-1, CL-P1, CD68 and CD11c mRNA expression were significantly higher in diabetic patients with IHD when compared with those without T2DM and control patients. MSR1, CXCL16, CD36 and CD206 showed no significant differences. In IHD patients, LOX-1 (OR 2.9; 95% CI 1.6–6.7; P = 0.019) and CD68 mRNA expression (OR 1.7; 95% CI 0.98–4.5; P = 0.049) were identified as independent risk factors associated with T2DM. Glucose and glycated hemoglobin were also shown to be risk factors. Conclusions SRs mRNA expression is found in EAT. LOX-1 and CD68 and were higher in IHD patients with T2DM and were identified as a cardiovascular risk factor of T2DM. This study suggests the importance of EAT in coronary atherosclerosis among patients with T2DM.
Collapse
Affiliation(s)
- Concepción Santiago-Fernández
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital (IBIMA), Malaga University, Campus de Teatinos s/n, 29010, Malaga, Spain.,Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Malaga, Spain
| | - Luis M Pérez-Belmonte
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Campus Universitario de Teatinos, s/n., Malaga, Spain. .,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Malaga, Spain.
| | - Mercedes Millán-Gómez
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Campus Universitario de Teatinos, s/n., Malaga, Spain.,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Malaga, Spain
| | - Inmaculada Moreno-Santos
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Campus Universitario de Teatinos, s/n., Malaga, Spain.,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Malaga, Spain
| | - Fernando Carrasco-Chinchilla
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Campus Universitario de Teatinos, s/n., Malaga, Spain.,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Malaga, Spain
| | - Amalio Ruiz-Salas
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Campus Universitario de Teatinos, s/n., Malaga, Spain.,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Malaga, Spain
| | - Luis Morcillo-Hidalgo
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Campus Universitario de Teatinos, s/n., Malaga, Spain.,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Malaga, Spain
| | - José M Melero
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Campus Universitario de Teatinos, s/n., Malaga, Spain.,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Malaga, Spain
| | - Lourdes Garrido-Sánchez
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital (IBIMA), Malaga University, Campus de Teatinos s/n, 29010, Malaga, Spain. .,Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Malaga, Spain.
| | - Manuel Jiménez-Navarro
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), Campus Universitario de Teatinos, s/n., Malaga, Spain.,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Malaga, Spain
| |
Collapse
|
24
|
Xu W, Wang Y, Guo Y, Liu J, Ma L, Cao W, Yu B, Zhou Y. Fibroblast growth factor 19 improves cardiac function and mitochondrial energy homoeostasis in the diabetic heart. Biochem Biophys Res Commun 2018; 505:242-248. [PMID: 30243718 DOI: 10.1016/j.bbrc.2018.09.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 09/08/2018] [Indexed: 11/29/2022]
Abstract
In diabetic cardiomyopathy, mitochondrial fatty acid oxidation dominates over mitochondrial glucose oxidation, leading to metabolic disturbances. Fibroblast growth factor 19 (FGF19) acts as a metabolic regulator and may have a cardioprotective role on diabetic cardiomyopathy. In this study, we investigated the effects of FGF19 on energy metabolism. FGF19 treatment of diabetic hearts exhibited higher glucose uptake and lower lipid profiles, suggesting changes in energy metabolism. The protective effects of FGF19 prevented ventricular dysfunction in diabetic hearts and improved mitochondrial function by the upregulation of PGC-1α expression. On the other side, knockdown of PGC-1α by siRNA attenuated the effects of FGF19 on the enhancement of mitochondrial function and energy efficiency. Taken together, these results show that FGF19 exhibited improved mitochondrial efficiency, which might be associated with higher cardiac contractility in diabetic hearts. It is also of note that modulation of PGC-1α, which is responsible for the activation by FGF19, may be a therapeutic target for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Wei Xu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang Province, China
| | - Yongshun Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang Province, China
| | - Yibo Guo
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jingjin Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang Province, China
| | - Lijia Ma
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang Province, China
| | - Wei Cao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang Province, China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang Province, China
| | - Yuhong Zhou
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, China.
| |
Collapse
|
25
|
Shirkhani S, Marandi SM, Kazeminasab F, Esmaeili M, Ghaedi K, Esfarjani F, Shiralian-Esfahani H, Nasr-Esfahani MH. Comparative studies on the effects of high-fat diet, endurance training and obesity on Ucp1 expression in male C57BL/6 mice. Gene 2018; 676:16-21. [PMID: 30201103 DOI: 10.1016/j.gene.2018.07.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/04/2018] [Accepted: 07/05/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Obesity triggers a variety of severe conditions, therefore deteriorates metabolism rate of adipose tissues and muscles. Uncoupling proteins which are highly stimulated by fatty acids are potential targets for anti-obesity agents through breaking the electron gradient in the mitochondrial matrix and creating imbalances in the electron transport chain, thereby increasing the amount of substrate used to produce energy. Therefore, the aim of present study is assessment of exercise and high fat diet on expression level of Ucp1 subcutaneous white and brown adipose tissues (scWAT & BAT) respectively. METHODS To perform experiments, 48 male C57BL/6 mice were divided to two major groups and fed with high fat diet (HFD) or low fat diet (LFD) during a period of 12 weeks. After the first intervention, each groups was divided into four groups randomly as (HF-EX), (HF-SED), (LF-EX), (LF-SED) [EX: exercise; SED: sedentary] in form of treadmill running for 45 min/day, 5 days/week during 8 weeks. One day after the last practice session, mice were sacrificed and Ucp1 expression was assessed on scWAT & BAT. RESULTS Data indicated a down-regulation in scWAT Ucp1 in obese mice similar to what observed for the expression of Pgc1α. Both, BAT Ucp1 and Pgc1α mRNA decreased significantly in response to obesity and physical activity. Moreover, exercise caused significant decrease in scWAT mitochondrial proteins contradictory to BAT. CONCLUSION Taken together, exercise exerted controversial effects compared with HFD and obesity on expression of Ucp1 and Pgc1α in scWAT dissimilar to BAT tissues, concluding that obesity may cause a resistance to exercise in terms of metabolic demands for scWAT tissue.
Collapse
Affiliation(s)
- Samaneh Shirkhani
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Isfahan, Iran
| | - Sayed Mohammad Marandi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Isfahan, Iran.
| | - Fatemeh Kazeminasab
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Isfahan, Iran
| | - Maryam Esmaeili
- Department of Cellular Biotechnology, Cell Sciences Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran; Department of Cellular Biotechnology, Cell Sciences Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Fahimeh Esfarjani
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Isfahan, Iran
| | - Hanieh Shiralian-Esfahani
- Department of Cellular Biotechnology, Cell Sciences Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Sciences Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
26
|
Bombassaro B, Ignacio-Souza LM, Nunez CE, Razolli DS, Pedro RM, Coope A, Araujo EP, Chaim EA, Velloso LA. A20 deubiquitinase controls PGC-1α expression in the adipose tissue. Lipids Health Dis 2018; 17:90. [PMID: 29678181 PMCID: PMC5909260 DOI: 10.1186/s12944-018-0740-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 04/09/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Peroxisome proliferator-activated receptor γ coactivator- 1alpha (PGC-1α) plays an important role in whole body metabolism and, particularly in glucose homeostasis. Its expression is highly regulated and, small variations in tissue levels can have a major impact in a number of physiological and pathological conditions. Recent studies have shown that the ubiquitin/proteasome system plays a role in the control of PGC-1α degradation. METHODS Here we evaluated the interaction of PGC-1α with the protein A20, which plays a dual-role in the control of the ubiquitin/proteasome system acting as a deubiquitinase and as an E3 ligase. We employed immunoprecipitation, quantitative real-time PCR and immunofluorescence staining to evaluate PGC-1α, A20, PPARγ and ubiquitin in the adipose tissue of humans and mice. RESULTS In distinct sites of the adipose tissue, A20 binds to PGC-1α. At least in the subcutaneous fat of humans and mice the levels of PGC-1α decrease during obesity, while its physical association with A20 increases. The inhibition of A20 leads to a reduction of PGC-1α and PPARγ expression, suggesting that A20 acts as a protective factor against PGC-1α disposal. CONCLUSION We provide evidence that mechanisms regulating PGC-1α ubiquitination are potentially involved in the control of the function of this transcriptional co-activator.
Collapse
Affiliation(s)
- Bruna Bombassaro
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center University of Campinas, Campinas, Brazil
| | - Leticia M Ignacio-Souza
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center University of Campinas, Campinas, Brazil
| | - Carla E Nunez
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center University of Campinas, Campinas, Brazil
| | - Daniela S Razolli
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center University of Campinas, Campinas, Brazil
| | - Rafael M Pedro
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center University of Campinas, Campinas, Brazil
| | - Andressa Coope
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center University of Campinas, Campinas, Brazil
| | - Eliana P Araujo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center University of Campinas, Campinas, Brazil
| | - Elinton A Chaim
- Department of Surgery, University of Campinas, Campinas, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center University of Campinas, Campinas, Brazil. .,Laboratory of Cell Signaling, Faculdade de Ciencias Medicas da Universidade Estadual de Campinas, Campinas, SP, 13084 970, Brazil.
| |
Collapse
|
27
|
Alexandar SP, Dhinakaran I, Ravi V, Parthasarathy N, Ganesan S, Bhaskaran M, Arun Kumar GP. Meta-Analysis of Association of Mitochondrial DNA Mutations with Type 2 Diabetes and Gestational Diabetes Mellitus. INT J HUM GENET 2018. [DOI: 10.1080/09723757.2018.1430110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Soundarya Priya Alexandar
- Human Genomics Laboratory, School of Chemical & Biotechnology, SASTRA University Thanjavur, Thanjavur 613 401, Tamil Nadu, India
| | - Indhumathi Dhinakaran
- Human Genomics Laboratory, School of Chemical & Biotechnology, SASTRA University Thanjavur, Thanjavur 613 401, Tamil Nadu, India
| | - Vidhya Ravi
- K.A.P. Viswanatham Govt. Medical College, Trichy, 620 001, Tamil Nadu, India
| | - Nandhini Parthasarathy
- Human Genomics Laboratory, School of Chemical & Biotechnology, SASTRA University Thanjavur, Thanjavur 613 401, Tamil Nadu, India
| | - Somasundari Ganesan
- Human Genomics Laboratory, School of Chemical & Biotechnology, SASTRA University Thanjavur, Thanjavur 613 401, Tamil Nadu, India
| | - Muthumeenakshi Bhaskaran
- Human Genomics Laboratory, School of Chemical & Biotechnology, SASTRA University Thanjavur, Thanjavur 613 401, Tamil Nadu, India
| | - Ganesh Prasad Arun Kumar
- Human Genomics Laboratory, School of Chemical & Biotechnology, SASTRA University Thanjavur, Thanjavur 613 401, Tamil Nadu, India
| |
Collapse
|
28
|
Resistance exercise improves cardiac function and mitochondrial efficiency in diabetic rat hearts. Pflugers Arch 2017; 470:263-275. [PMID: 29032504 DOI: 10.1007/s00424-017-2076-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/22/2017] [Accepted: 10/02/2017] [Indexed: 12/25/2022]
Abstract
Metabolic disturbance and mitochondrial dysfunction are a hallmark of diabetic cardiomyopathy (DC). Resistance exercise (RE) not only enhances the condition of healthy individuals but could also improve the status of those with disease. However, the beneficial effects of RE in the prevention of DC and mitochondrial dysfunction are uncertain. Therefore, this study investigated whether RE attenuates DC by improving mitochondrial function using an in vivo rat model of diabetes. Fourteen Otsuka Long-Evans Tokushima Fatty rats were assigned to sedentary control (SC, n = 7) and RE (n = 7) groups at 28 weeks of age. Long-Evans Tokushima Otsuka rats were used as the non-diabetic control. The RE rats were trained by 20 repetitions of climbing a ladder 5 days per week. RE rats exhibited higher glucose uptake and lower lipid profiles, indicating changes in energy metabolism. RE rats significantly increased the ejection fraction and fractional shortening compared with the SC rats. Isolated mitochondria in RE rats showed increase in mitochondrial numbers, which were accompanied by higher expression of mitochondrial biogenesis proteins such as proliferator-activated receptor-γ coactivator-1α and TFAM. Moreover, RE rats reduced proton leakage and reactive oxygen species production, with higher membrane potential. These results were accompanied by higher superoxide dismutase 2 and lower uncoupling protein 2 (UCP2) and UCP3 levels in RE rats. These data suggest that RE is effective at ameliorating DC by improving mitochondrial function, which may contribute to the maintenance of diabetic cardiac contractility.
Collapse
|
29
|
Gaborit B, Dutour A. Looking beyond ectopic fat amount: A SMART method to quantify epicardial adipose tissue density. Eur J Prev Cardiol 2017; 24:657-659. [DOI: 10.1177/2047487317689976] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Bénédicte Gaborit
- Faculté de Médecine, Aix Marseille University, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Centre Hospitalier Universitaire Nord, France
| | - Anne Dutour
- Faculté de Médecine, Aix Marseille University, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Centre Hospitalier Universitaire Nord, France
| |
Collapse
|
30
|
Pérez-Belmonte LM, Moreno-Santos I, Gómez-Doblas JJ, García-Pinilla JM, Morcillo-Hidalgo L, Garrido-Sánchez L, Santiago-Fernández C, Crespo-Leiro MG, Carrasco-Chinchilla F, Sánchez-Fernández PL, de Teresa-Galván E, Jiménez-Navarro M. Expression of epicardial adipose tissue thermogenic genes in patients with reduced and preserved ejection fraction heart failure. Int J Med Sci 2017; 14:891-895. [PMID: 28824327 PMCID: PMC5562197 DOI: 10.7150/ijms.19854] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/25/2017] [Indexed: 12/02/2022] Open
Abstract
Epicardial adipose tissue has been proposed to participate in the pathogenesis of heart failure. The aim of our study was to assess the expression of thermogenic genes (Uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), and PR-domain-missing 16 (PRDM16) in epicardial adipose tissue in patients with heart failure, stablishing the difference according to left ventricular ejection fraction (reduced or preserved). Among the 75 patients in our study, 42.7% (n=32) had reduced left ventricular ejection fraction. UCP1, PGC1α and PRDM16 mRNA in EAT were significantly lower in patients with reduced left ventricular ejection fraction. Multiple regression analysis showed that age, male gender, body max index, presence of obesity, type-2-diabetes mellitus, hypertension and coronary artery disease and left ventricular ejection fraction were associated with the expression levels of UCP1, PGC1α and PRDM16 mRNA. Thermogenic genes expressions in epicardial adipose tissue (UCP1: OR 0.617, 95%CI 0.103-0.989, p=0.042; PGC1α: OR 0.416, 95%CI 0.171-0.912, p=0.031; PRDM16: OR 0.643, 95%CI 0.116-0.997, p=0.044) were showed as protective factors against the presence of heart failure with reduced left ventricular ejection fraction, and age (OR 1.643, 95%CI 1.001-3.143, p=0.026), presence of coronary artery disease (OR 6.743, 95%CI 1.932-15.301, p<0.001) and type-2-diabetes mellitus (OR 4.031, 95%CI 1.099-7.231, p<0.001) were associated as risk factors. The adequate expression of thermogenic genes has been shown as possible protective factors against heart failure with reduced ejection fraction, suggesting that a loss of functional epicardial adipose tissue brown-like features would participate in a deleterious manner on heart metabolism. Thermogenic genes could represent a future novel therapeutic target in heart failure.
Collapse
Affiliation(s)
- Luis M Pérez-Belmonte
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Inmaculada Moreno-Santos
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Juan J Gómez-Doblas
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - José M García-Pinilla
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Luis Morcillo-Hidalgo
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Lourdes Garrido-Sánchez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Spain
| | - Concepción Santiago-Fernández
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Spain
| | - María G Crespo-Leiro
- Servicio de Cardiología, Instituto de Investigación Biomédica A Coruña (INIBIC), Complejo Hospitalario Universitario A Coruña, CIBERCV Enfermedades Cardiovasculares, A Coruña. Spain
| | - Fernando Carrasco-Chinchilla
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Pedro L Sánchez-Fernández
- Servicio de Cardiología, Instituto de Investigación Biomédica de Salamanca (IBISAL), Hospital Universitario de Salamanca, Universidad de Salamanca (USAL), CIBERCV Enfermedades Cardiovasculares, Salamanca, Spain
| | - Eduardo de Teresa-Galván
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Manuel Jiménez-Navarro
- Unidad de Gestión Clínica Área del Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga (UMA), CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| |
Collapse
|