1
|
Sun Q, Karwi QG, Wong N, Lopaschuk GD. Advances in myocardial energy metabolism: metabolic remodelling in heart failure and beyond. Cardiovasc Res 2024; 120:1996-2016. [PMID: 39453987 PMCID: PMC11646102 DOI: 10.1093/cvr/cvae231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/28/2024] [Accepted: 07/03/2024] [Indexed: 10/27/2024] Open
Abstract
The very high energy demand of the heart is primarily met by adenosine triphosphate (ATP) production from mitochondrial oxidative phosphorylation, with glycolysis providing a smaller amount of ATP production. This ATP production is markedly altered in heart failure, primarily due to a decrease in mitochondrial oxidative metabolism. Although an increase in glycolytic ATP production partly compensates for the decrease in mitochondrial ATP production, the failing heart faces an energy deficit that contributes to the severity of contractile dysfunction. The relative contribution of the different fuels for mitochondrial ATP production dramatically changes in the failing heart, which depends to a large extent on the type of heart failure. A common metabolic defect in all forms of heart failure [including heart failure with reduced ejection fraction (HFrEF), heart failure with preserved EF (HFpEF), and diabetic cardiomyopathies] is a decrease in mitochondrial oxidation of pyruvate originating from glucose (i.e. glucose oxidation). This decrease in glucose oxidation occurs regardless of whether glycolysis is increased, resulting in an uncoupling of glycolysis from glucose oxidation that can decrease cardiac efficiency. The mitochondrial oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in HFpEF and diabetic cardiomyopathies myocardial fatty acid oxidation increases, while in HFrEF myocardial fatty acid oxidation either decreases or remains unchanged. The oxidation of ketones (which provides the failing heart with an important energy source) also differs depending on the type of heart failure, being increased in HFrEF, and decreased in HFpEF and diabetic cardiomyopathies. The alterations in mitochondrial oxidative metabolism and glycolysis in the failing heart are due to transcriptional changes in key enzymes involved in the metabolic pathways, as well as alterations in redox state, metabolic signalling and post-translational epigenetic changes in energy metabolic enzymes. Of importance, targeting the mitochondrial energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac function and cardiac efficiency in the failing heart.
Collapse
Affiliation(s)
- Qiuyu Sun
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John’s, NL A1B 3V6, Canada
| | - Nathan Wong
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
2
|
Bornstein MR, Tian R, Arany Z. Human cardiac metabolism. Cell Metab 2024; 36:1456-1481. [PMID: 38959861 PMCID: PMC11290709 DOI: 10.1016/j.cmet.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/12/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024]
Abstract
The heart is the most metabolically active organ in the human body, and cardiac metabolism has been studied for decades. However, the bulk of studies have focused on animal models. The objective of this review is to summarize specifically what is known about cardiac metabolism in humans. Techniques available to study human cardiac metabolism are first discussed, followed by a review of human cardiac metabolism in health and in heart failure. Mechanistic insights, where available, are reviewed, and the evidence for the contribution of metabolic insufficiency to heart failure, as well as past and current attempts at metabolism-based therapies, is also discussed.
Collapse
Affiliation(s)
- Marc R Bornstein
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Zoltan Arany
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Divya KP, Kanwar N, Anuranjana PV, Kumar G, Beegum F, George KT, Kumar N, Nandakumar K, Kanwal A. SIRT6 in Regulation of Mitochondrial Damage and Associated Cardiac Dysfunctions: A Possible Therapeutic Target for CVDs. Cardiovasc Toxicol 2024; 24:598-621. [PMID: 38689163 DOI: 10.1007/s12012-024-09858-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Cardiovascular diseases (CVDs) can be described as a global health emergency imploring possible prevention strategies. Although the pathogenesis of CVDs has been extensively studied, the role of mitochondrial dysfunction in CVD development has yet to be investigated. Diabetic cardiomyopathy, ischemic-reperfusion injury, and heart failure are some of the CVDs resulting from mitochondrial dysfunction Recent evidence from the research states that any dysfunction of mitochondria has an impact on metabolic alteration, eventually causes the death of a healthy cell and therefore, progressively directing to the predisposition of disease. Cardiovascular research investigating the targets that both protect and treat mitochondrial damage will help reduce the risk and increase the quality of life of patients suffering from various CVDs. One such target, i.e., nuclear sirtuin SIRT6 is strongly associated with cardiac function. However, the link between mitochondrial dysfunction and SIRT6 concerning cardiovascular pathologies remains poorly understood. Although the Role of SIRT6 in skeletal muscles and cardiomyocytes through mitochondrial regulation has been well understood, its specific role in mitochondrial maintenance in cardiomyocytes is poorly determined. The review aims to explore the domain-specific function of SIRT6 in cardiomyocytes and is an effort to know how SIRT6, mitochondria, and CVDs are related.
Collapse
Affiliation(s)
- K P Divya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Navjot Kanwar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab, Technical University, Bathinda, Punjab, 151005, India
| | - P V Anuranjana
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Gautam Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
- School of Pharmacy, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Krupa Thankam George
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology, National Institute of Pharmaceutical Educations and Research, Hajipur, Bihar, 844102, India
| | - K Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India.
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, Punjab, 151005, India.
| |
Collapse
|
4
|
Rubio-Tomás T, Soler-Botija C, Martínez-Estrada O, Villena JA. Transcriptional control of cardiac energy metabolism in health and disease: Lessons from animal models. Biochem Pharmacol 2024; 224:116185. [PMID: 38561091 DOI: 10.1016/j.bcp.2024.116185] [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: 12/11/2023] [Revised: 02/27/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Cardiac ATP production is tightly regulated in order to satisfy the evolving energetic requirements imposed by different cues during health and pathological conditions. In order to sustain high ATP production rates, cardiac cells are endowed with a vast mitochondrial network that is essentially acquired during the perinatal period. Nevertheless, adult cardiac cells also adapt their mitochondrial mass and oxidative function to changes in energy demand and substrate availability by fine-tuning the pathways and mitochondrial machinery involved in energy production. The reliance of cardiac cells on mitochondrial metabolism makes them particularly sensitive to alterations in proper mitochondrial function, so that deficiency in energy production underlies or precipitates the development of heart diseases. Mitochondrial biogenesis is a complex process fundamentally controlled at the transcriptional level by a network of transcription factors and co-regulators, sometimes with partially redundant functions, that ensure adequate energy supply to the working heart. Novel uncovered regulators, such as RIP140, PERM1, MED1 or BRD4 have been recently shown to modulate or facilitate the transcriptional activity of the PGC-1s/ERRs/PPARs regulatory axis, allowing cardiomyocytes to adapt to a variety of physiological or pathological situations requiring different energy provision. In this review, we summarize the current knowledge on the mechanisms that regulate cardiac mitochondrial biogenesis, highlighting the recent discoveries of new transcriptional regulators and describing the experimental models that have provided solid evidence of the relevant contribution of these factors to cardiac function in health and disease.
Collapse
Affiliation(s)
- Teresa Rubio-Tomás
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion GR-70013, Crete, Greece
| | - Carolina Soler-Botija
- ICREC (Heart Failure and Cardiac Regeneration) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBER on Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Josep A Villena
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; CIBER on Diabetes and Associated Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain.
| |
Collapse
|
5
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
6
|
Qian L, Zhu Y, Deng C, Liang Z, Chen J, Chen Y, Wang X, Liu Y, Tian Y, Yang Y. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases. Signal Transduct Target Ther 2024; 9:50. [PMID: 38424050 PMCID: PMC10904817 DOI: 10.1038/s41392-024-01756-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family (PGC-1s), consisting of three members encompassing PGC-1α, PGC-1β, and PGC-1-related coactivator (PRC), was discovered more than a quarter-century ago. PGC-1s are essential coordinators of many vital cellular events, including mitochondrial functions, oxidative stress, endoplasmic reticulum homeostasis, and inflammation. Accumulating evidence has shown that PGC-1s are implicated in many diseases, such as cancers, cardiac diseases and cardiovascular diseases, neurological disorders, kidney diseases, motor system diseases, and metabolic disorders. Examining the upstream modulators and co-activated partners of PGC-1s and identifying critical biological events modulated by downstream effectors of PGC-1s contribute to the presentation of the elaborate network of PGC-1s. Furthermore, discussing the correlation between PGC-1s and diseases as well as summarizing the therapy targeting PGC-1s helps make individualized and precise intervention methods. In this review, we summarize basic knowledge regarding the PGC-1s family as well as the molecular regulatory network, discuss the physio-pathological roles of PGC-1s in human diseases, review the application of PGC-1s, including the diagnostic and prognostic value of PGC-1s and several therapies in pre-clinical studies, and suggest several directions for future investigations. This review presents the immense potential of targeting PGC-1s in the treatment of diseases and hopefully facilitates the promotion of PGC-1s as new therapeutic targets.
Collapse
Affiliation(s)
- Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China
| | - Junmin Chen
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xue Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yanqing Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China.
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
7
|
Oka SI, Sreedevi K, Shankar TS, Yedla S, Arowa S, James A, Stone KG, Olmos K, Sabry AD, Horiuchi A, Cawley KM, O’very SA, Tong M, Byun J, Xu X, Kashyap S, Mourad Y, Vehra O, Calder D, Lunde T, Liu T, Li H, Mashchek JA, Cox J, Saijoh Y, Drakos SG, Warren JS. PERM1 regulates energy metabolism in the heart via ERRα/PGC-1α axis. Front Cardiovasc Med 2022; 9:1033457. [PMID: 36419485 PMCID: PMC9676655 DOI: 10.3389/fcvm.2022.1033457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022] Open
Abstract
Aims PERM1 is a striated muscle-specific regulator of mitochondrial bioenergetics. We previously demonstrated that PERM1 is downregulated in the failing heart and that PERM1 positively regulates metabolic genes known as targets of the transcription factor ERRα and its coactivator PGC-1α in cultured cardiomyocytes. The aims of this study were to determine the effect of loss of PERM1 on cardiac function and energetics using newly generated Perm1-knockout (Perm1 -/-) mice and to investigate the molecular mechanisms of its transcriptional control. Methods and results Echocardiography showed that ejection fraction and fractional shortening were lower in Perm1 -/- mice than in wild-type mice (both p < 0.05), and the phosphocreatine-to-ATP ratio was decreased in Perm1 -/- hearts (p < 0.05), indicating reduced contractile function and energy reserves of the heart. Integrated proteomic and metabolomic analyses revealed downregulation of oxidative phosphorylation and upregulation of glycolysis and polyol pathways in Perm1 -/- hearts. To examine whether PERM1 regulates energy metabolism through ERRα, we performed co-immunoprecipitation assays, which showed that PERM1 bound to ERRα in cardiomyocytes and the mouse heart. DNA binding and reporter gene assays showed that PERM1 was localized to and activated the ERR target promoters partially through ERRα. Mass spectrometry-based screening in cardiomyocytes identified BAG6 and KANK2 as potential PERM1's binding partners in transcriptional regulation. Mammalian one-hybrid assay, in which PERM1 was fused to Gal4 DNA binding domain, showed that the recruitment of PERM1 to a gene promoter was sufficient to activate transcription, which was blunted by silencing of either PGC-1α, BAG6, or KANK2. Conclusion This study demonstrates that PERM1 is an essential regulator of cardiac energetics and function and that PERM1 is a novel transcriptional coactivator in the ERRα/PGC-1α axis that functionally interacts with BAG6 and KANK2.
Collapse
Affiliation(s)
- Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Karthi Sreedevi
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Thirupura S. Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Shreya Yedla
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Sumaita Arowa
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Amina James
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Kathryn G. Stone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Katia Olmos
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Amira D. Sabry
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Amanda Horiuchi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Keiko M. Cawley
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Sean A. O’very
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Mingming Tong
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Youssef Mourad
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Omair Vehra
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Dallen Calder
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Ty Lunde
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Tong Liu
- Department of Microbiology, Biochemistry, and Molecular Genetics, Center for Advanced Proteomics Research, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, NJ, United States
| | - Hong Li
- Department of Microbiology, Biochemistry, and Molecular Genetics, Center for Advanced Proteomics Research, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, NJ, United States
| | - J. Alan Mashchek
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, United States
| | - James Cox
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, United States
- Department of Biochemistry, University of Utah, Salt Lake City, UT, United States
| | - Yukio Saijoh
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Junco S. Warren
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
- Department of Human Nutrition, Food and Exercise, Virginia Tech, Blacksburg, VA, United States
- Division of Developmental Genetics, Institute of Resource Developmental and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
8
|
Huerta-Delgado AS, Roffe-Vazquez DN, Luna-Ceron E, Gonzalez-Gil AM, Casillas-Fikentscher A, Villarreal-Calderon JR, Enriquez C, de la Peña-Almaguer E, Castillo EC, Silva-Platas C, Garcia-Rivas G, Elizondo-Montemayor L. Association of irisin levels with cardiac magnetic resonance, inflammatory, and biochemical parameters in patients with chronic heart failure versus controls. Magn Reson Imaging 2022; 93:62-72. [PMID: 35842196 DOI: 10.1016/j.mri.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/15/2022] [Accepted: 07/11/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Chronic heart failure (CHF) represents a significant cause of morbidity and mortality globally. Metabolic maladaptation has proven to be critical in the progression of this condition. Preclinical studies have shown that irisin, an adipomyokine involved in metabolic regulations, can induce positive cardioprotective effects by improving cardiac remodeling, cardiomyocyte viability, calcium delivery, and reducing inflammatory mediators. However, data on clinical studies identifying the associations between irisin levels and functional imaging parameters are scarce in CHF patients. The objective of this study was to determine the association of irisin levels with cardiac imaging measurements through cardiac magnetic resonance, inflammatory markers, and biochemical parameters in patients with CHF compared with control subjects. METHODS AND RESULTS Thirty-two subjects diagnosed with CHF and thirty-two healthy controls were evaluated in a cross-sectional study. Serum irisin levels were significantly lower in patients with CHF than in controls. This is the first study to report a significant positive correlation between irisin levels and cardiac magnetic resonance parameters such as left ventricular ejection fraction, fraction shortening, and global radial strain. A negative correlation was demonstrated between irisin levels and brain natriuretic peptide, insulin levels, and Homeostatic model assessment for insulin resistance index. We did not observe significant correlations between irisin levels and inflammatory cytokines. CONCLUSIONS Given the importance of fraction shortening and global radial strain as accurate markers of ventricular wall motion, these results support the hypothesis that irisin may play an essential role in maintaining an adequate myocardial wall architecture, deformation, and thickness.
Collapse
Affiliation(s)
- Anna S Huerta-Delgado
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Daniel N Roffe-Vazquez
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Eder Luna-Ceron
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Adrian M Gonzalez-Gil
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Andrea Casillas-Fikentscher
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - José R Villarreal-Calderon
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico
| | - Cecilio Enriquez
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Erasmo de la Peña-Almaguer
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Elena C Castillo
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Christian Silva-Platas
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Gerardo Garcia-Rivas
- Tecnologico de Monterrey, Centro de Investigacion Biomedica, Hospital Zambrano Hellion, 66278 San Pedro Garza-Garcia, N.L., Mexico; Tecnologico de Monterrey, Cardiovascular Medicine and Metabolomics Research Group, Escuela de Medicina, 66278 San Pedro Garza-Garcia, N.L., Mexico
| | - Leticia Elizondo-Montemayor
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Escuela de Medicina, 64710 Monterrey, N.L., Mexico; Tecnologico de Monterrey, Cardiovascular Medicine and Metabolomics Research Group, Escuela de Medicina, 66278 San Pedro Garza-Garcia, N.L., Mexico.
| |
Collapse
|
9
|
Chen L, Qin Y, Liu B, Gao M, Li A, Li X, Gong G. PGC-1 α-Mediated Mitochondrial Quality Control: Molecular Mechanisms and Implications for Heart Failure. Front Cell Dev Biol 2022; 10:871357. [PMID: 35721484 PMCID: PMC9199988 DOI: 10.3389/fcell.2022.871357] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/27/2022] [Indexed: 12/26/2022] Open
Abstract
Mitochondria with structural and functional integrity are essential for maintaining mitochondrial function and cardiac homeostasis. It is involved in the pathogenesis of many diseases. Peroxisome proliferator-activated receptor γ coactivator 1 α (PGC-1α), acted as a transcriptional cofactor, is abundant in the heart, which modulates mitochondrial biogenesis and mitochondrial dynamics and mitophagy to sustain a steady-state of mitochondria. Cumulative evidence suggests that dysregulation of PGC-1α is closely related to the onset and progression of heart failure. PGC-1α deficient-mice can lead to worse cardiac function under pressure overload compared to sham. Here, this review mainly focuses on what is known about its regulation in mitochondrial functions, as well as its crucial role in heart failure.
Collapse
Affiliation(s)
- Lei Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuan Qin
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Bilin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Meng Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Anqi Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xue Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Guohua Gong
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
10
|
Gul R, Alsalman N, Bazighifan A, Alfadda AA. Comparative beneficial effects of nebivolol and nebivolol/valsartan combination against mitochondrial dysfunction in angiotensin II-induced pathology in H9c2 cardiomyoblasts. J Pharm Pharmacol 2021; 73:1520-1529. [PMID: 34453839 DOI: 10.1093/jpp/rgab124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 08/03/2021] [Indexed: 11/14/2022]
Abstract
OBJECTIVES Considering the complementary nature of signalling mechanisms and the therapeutic effects of nebivolol, a β1-adrenoreceptor antagonist, and valsartan, an angiotensin receptor blocker (ARB), here we aimed to investigate whether nebivolol/valsartan combination would complement the cardioprotective effects of nebivolol on angiotensin II (ANG II)-induced pathology in H9c2 cardiomyoblasts. METHODS H9c2 cardiomyoblasts were used to investigate the protective effects of nebivolol and nebivolol and valsartan combination against ANG II-induced pathology. Reactive oxygen species (ROS) generation was determined by 2',7'-dichlorofluorescein diacetate (DCFDA) and MitoSOX Red staining. Real-time PCR and immunoblotting were employed to quantify the changes in mRNA and protein expression levels, respectively. KEY FINDINGS Our data revealed that pretreatment with nebivolol and nebivolol/valsartan combination significantly reduced ANG II-induced oxidative stress and mTORC1 signalling. Concurrently, ANG II-induced activation of inflammatory cytokines and fetal gene expressions were significantly suppressed by nebivolol and nebivolol/valsartan combination. Pretreatment with nebivolol and nebivolol/valsartan combination alleviated ANG II-induced impairment of mitochondrial biogenesis by restoring the gene expression levels of PGC-1α, TFAM, NRF-1 and SIRT3. Our data further show that nebivolol and nebivolol/valsartan combination mediated up-regulation in mitochondrial biogenesis is accompanied by decrease in ANG II-stimulated mitochondrial ROS generation as well as increase in expression of mitochondrial fusion genes MFN2 and OPA1, indicative of improved mitochondrial dynamics. SUMMARY These findings suggest that both nebivolol and nebivolol/valsartan combination exert protective effects on ANG II-induced mitochondrial dysfunction by alleviating its biogenesis and dynamics. Moreover, addition of valsartan to nebivolol do not produce any additive effects compared with nebivolol alone on ANG II-induced cardiac pathology.
Collapse
Affiliation(s)
- Rukhsana Gul
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Nouf Alsalman
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Arwa Bazighifan
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Assim A Alfadda
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia.,Department of Medicine, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia.,Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
11
|
Feliciano RDS, Atum ALB, Ruiz ÉGDS, Serra AJ, Antônio EL, Manchini MT, Silva JMA, Tucci PJF, Nathanson L, Morris M, Chavantes MC, Silva Júnior JA. Photobiomodulation Therapy on Myocardial Infarction in Rats: Transcriptional and Posttranscriptional Implications to Cardiac Remodeling. Lasers Surg Med 2021; 53:1247-1257. [PMID: 33846991 DOI: 10.1002/lsm.23407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND OBJECTIVES Induction of myocardial infarction (MI) in rats by occlusion of the left anterior descending coronary artery is an experimental model used in research to elucidate functional, structural, and molecular modifications associated with ischemic heart disease. Photobiomodulation therapy (PBMT) has become a therapeutic alternative by modulating various biological processes eliciting several effects, including anti-inflammatory and pro-proliferative actions. The main objective of this work was to evaluate the effect of PBMT in the modulation of transcriptional and post-transcriptional changes that occurred in myocardium signal transduction pathways after MI. STUDY DESIGN/MATERIALS AND METHODS Continuous wave (CW) non-thermal laser parameters were: 660 nm wavelength, power 15 mW, with a total energy of 0.9 J, fluence of 1.15 J/cm2 , spot size of 0.785 cm2 , and time of 60 seconds. Using in silico analysis, we selected and then, quantified the expression of messenger RNA (mRNA) of 47 genes of 9 signaling pathways associated with MI (angiogenesis, cell survival, hypertrophy, oxidative stress, apoptosis, extracellular matrix, calcium kinetics, cell metabolism, and inflammation). Messenger RNA expression quantification was performed in myocardial samples by polymerase chain reaction real-time array using TaqMan customized plates. RESULTS Our results evidenced that MI modified mRNA expression of several well-known biomarkers related to detrimental cardiac activity in almost all signaling pathways analyzed. However, PBMT reverted most of these transcriptional changes. More expressively, PBMT provoked a robust decrease in mRNA expression of molecules that participate in post-MI inflammation and ECM composition, such as IL-6, TNF receptor, TGFb1, and collagen I and III. Global microRNA (miRNA) expression analysis revealed that PBMT decreased miR-221, miR-34c, and miR-93 expressions post-MI, which are related to deleterious effects in cardiac remodeling. CONCLUSION Thus, the identification of transcriptional and post-transcriptional changes induced by PBMT may be used to interfere in the molecular dynamics of cardiac remodeling post-MI.
Collapse
Affiliation(s)
| | | | | | - Andrey Jorge Serra
- Universidade Federal de São Paulo, Rua Pedro de Toledo 709, Vila Clementino, São Paulo, SP, 04039-001, Brazil
| | - Ednei Luiz Antônio
- Universidade Federal de São Paulo, Rua Pedro de Toledo 709, Vila Clementino, São Paulo, SP, 04039-001, Brazil
| | | | | | - Paulo José Ferreira Tucci
- Universidade Federal de São Paulo, Rua Pedro de Toledo 709, Vila Clementino, São Paulo, SP, 04039-001, Brazil
| | - Lubov Nathanson
- Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, Florida, 33314
| | - Mariana Morris
- Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, Florida, 33314
| | | | | |
Collapse
|
12
|
Ikeda G, Santoso MR, Tada Y, Li AM, Vaskova E, Jung JH, O’Brien C, Egan E, Ye J, Yang P. Mitochondria-Rich Extracellular Vesicles From Autologous Stem Cell-Derived Cardiomyocytes Restore Energetics of Ischemic Myocardium. J Am Coll Cardiol 2021; 77:1073-1088. [PMID: 33632482 PMCID: PMC8626617 DOI: 10.1016/j.jacc.2020.12.060] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/07/2020] [Accepted: 12/23/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Mitochondrial dysfunction results in an imbalance between energy supply and demand in a failing heart. An innovative therapy that targets the intracellular bioenergetics directly through mitochondria transfer may be necessary. OBJECTIVES The purpose of this study was to establish a preclinical proof-of-concept that extracellular vesicle (EV)-mediated transfer of autologous mitochondria and their related energy source enhance cardiac function through restoration of myocardial bioenergetics. METHODS Human-induced pluripotent stem cell-derived cardiomyocytes (iCMs) were employed. iCM-conditioned medium was ultracentrifuged to collect mitochondria-rich EVs (M-EVs). Therapeutic effects of M-EVs were investigated using in vivo murine myocardial infarction (MI) model. RESULTS Electron microscopy revealed healthy-shaped mitochondria inside M-EVs. Confocal microscopy showed that M-EV-derived mitochondria were transferred into the recipient iCMs and fused with their endogenous mitochondrial networks. Treatment with 1.0 × 108/ml M-EVs significantly restored the intracellular adenosine triphosphate production and improved contractile profiles of hypoxia-injured iCMs as early as 3 h after treatment. In contrast, isolated mitochondria that contained 300× more mitochondrial proteins than 1.0 × 108/ml M-EVs showed no effect after 24 h. M-EVs contained mitochondrial biogenesis-related messenger ribonucleic acids, including proliferator-activated receptor γ coactivator-1α, which on transfer activated mitochondrial biogenesis in the recipient iCMs at 24 h after treatment. Finally, intramyocardial injection of 1.0 × 108 M-EVs demonstrated significantly improved post-MI cardiac function through restoration of bioenergetics and mitochondrial biogenesis. CONCLUSIONS M-EVs facilitated immediate transfer of their mitochondrial and nonmitochondrial cargos, contributing to improved intracellular energetics in vitro. Intramyocardial injection of M-EVs enhanced post-MI cardiac function in vivo. This therapy can be developed as a novel, precision therapeutic for mitochondria-related diseases including heart failure.
Collapse
Affiliation(s)
- Gentaro Ikeda
- Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Michelle R. Santoso
- Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Yuko Tada
- Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Albert M. Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Evgeniya Vaskova
- Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Ji Hye Jung
- Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Connor O’Brien
- Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Elizabeth Egan
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Phillip Yang
- Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
13
|
Christiansen LB, Reimann MJ, Schou-Pedersen AMV, Larsen S, Lykkesfeldt J, Olsen LH. Depleted Myocardial Coenzyme Q10 in Cavalier King Charles Spaniels with Congestive Heart Failure Due to Myxomatous Mitral Valve Disease. Antioxidants (Basel) 2021; 10:antiox10020161. [PMID: 33499156 PMCID: PMC7911325 DOI: 10.3390/antiox10020161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/21/2022] Open
Abstract
Congestive heart failure (CHF) has been associated with depleted myocardial coenzyme Q10 (Q10) concentrations in human patients. The aim of this study was to investigate associations between myocardial Q10 concentrations and myxomatous mitral valve disease (MMVD) severity in dogs. Furthermore, citrate synthase (CS) activity was analysed to determine if a reduction in myocardial Q10 was associated with mitochondrial depletion in the myocardium. Thirty Cavalier King Charles spaniels (CKCS) in MMVD stages B1 (n = 11), B2 (n = 5) and C (n = 14) according to the American College of Veterinary Internal Medicine (ACVIM) guidelines and 10 control (CON) dogs of other breeds were included. Myocardial Q10 concentration was analysed in left ventricular tissue samples using HPLC-ECD. CKCS with congestive heart failure (CHF; group C) had significantly reduced Q10 concentrations (median, 1.54 µg/mg; IQR, 1.36–1.94), compared to B1 (2.76 µg/mg; 2.10–4.81, p < 0.0018), B2 (3.85 µg/mg; 3.13–4.46, p < 0.0054) and CON dogs (2.8 µg/mg; 1.64–4.88, p < 0.0089). CS activity was comparable between disease groups. In conclusion, dogs with CHF due to MMVD had reduced myocardial Q10 concentrations. Studies evaluating antioxidant defense mechanisms as a therapeutic target for treatment of CHF in dogs are warranted.
Collapse
Affiliation(s)
- Liselotte B. Christiansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg C, Denmark; (L.B.C.); (M.J.R.); (A.M.V.S.-P.); (J.L.)
| | - Maria J. Reimann
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg C, Denmark; (L.B.C.); (M.J.R.); (A.M.V.S.-P.); (J.L.)
| | - Anne Marie V. Schou-Pedersen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg C, Denmark; (L.B.C.); (M.J.R.); (A.M.V.S.-P.); (J.L.)
| | - Steen Larsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark;
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Jens Lykkesfeldt
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg C, Denmark; (L.B.C.); (M.J.R.); (A.M.V.S.-P.); (J.L.)
| | - Lisbeth H. Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg C, Denmark; (L.B.C.); (M.J.R.); (A.M.V.S.-P.); (J.L.)
- Correspondence:
| |
Collapse
|
14
|
Activation of Cannabinoid Receptors Attenuates Endothelin-1-Induced Mitochondrial Dysfunction in Rat Ventricular Myocytes. J Cardiovasc Pharmacol 2020; 75:54-63. [PMID: 31815823 PMCID: PMC6964873 DOI: 10.1097/fjc.0000000000000758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Supplemental Digital Content is Available in the Text. Evidence suggests that the activation of the endocannabinoid system offers cardioprotection. Aberrant energy production by impaired mitochondria purportedly contributes to various aspects of cardiovascular disease. We investigated whether cannabinoid (CB) receptor activation would attenuate mitochondrial dysfunction induced by endothelin-1 (ET1). Acute exposure to ET1 (4 hours) in the presence of palmitate as primary energy substrate induced mitochondrial membrane depolarization and decreased mitochondrial bioenergetics and expression of genes related to fatty acid oxidation (ie, peroxisome proliferator–activated receptor-gamma coactivator-1α, a driver of mitochondrial biogenesis, and carnitine palmitoyltransferase-1β, facilitator of fatty acid uptake). A CB1/CB2 dual agonist with limited brain penetration, CB-13, corrected these parameters. AMP-activated protein kinase (AMPK), an important regulator of energy homeostasis, mediated the ability of CB-13 to rescue mitochondrial function. In fact, the ability of CB-13 to rescue fatty acid oxidation–related bioenergetics, as well as expression of proliferator-activated receptor-gamma coactivator-1α and carnitine palmitoyltransferase-1β, was abolished by pharmacological inhibition of AMPK using compound C and shRNA knockdown of AMPKα1/α2, respectively. Interventions that target CB/AMPK signaling might represent a novel therapeutic approach to address the multifactorial problem of cardiovascular disease.
Collapse
|
15
|
Peterzan MA, Lewis AJM, Neubauer S, Rider OJ. Non-invasive investigation of myocardial energetics in cardiac disease using 31P magnetic resonance spectroscopy. Cardiovasc Diagn Ther 2020; 10:625-635. [PMID: 32695642 DOI: 10.21037/cdt-20-275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cardiac metabolism and function are intrinsically linked. High-energy phosphates occupy a central and obligate position in cardiac metabolism, coupling oxygen and substrate fuel delivery to the myocardium with external work. This insight underlies the widespread clinical use of ischaemia testing. However, other deficits in high-energy phosphate metabolism (not secondary to supply-demand mismatch of oxygen and substrate fuels) may also be documented, and are of particular interest when found in the context of structural heart disease. This review introduces the scope of deficits in high-energy phosphate metabolism that may be observed in the myocardium, how to assess for them, and how they might be interpreted.
Collapse
Affiliation(s)
- Mark A Peterzan
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew J M Lewis
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Oliver J Rider
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Heyne E, Schrepper A, Doenst T, Schenkl C, Kreuzer K, Schwarzer M. High-fat diet affects skeletal muscle mitochondria comparable to pressure overload-induced heart failure. J Cell Mol Med 2020; 24:6741-6749. [PMID: 32363733 PMCID: PMC7299710 DOI: 10.1111/jcmm.15325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 01/01/2023] Open
Abstract
In heart failure, high-fat diet (HFD) may exert beneficial effects on cardiac mitochondria and contractility. Skeletal muscle mitochondrial dysfunction in heart failure is associated with myopathy. However, it is not clear if HFD affects skeletal muscle mitochondria in heart failure as well. To induce heart failure, we used pressure overload (PO) in rats fed normal chow or HFD. Interfibrillar mitochondria (IFM) and subsarcolemmal mitochondria (SSM) from gastrocnemius were isolated and functionally characterized. With PO heart failure, maximal respiratory capacity was impaired in IFM but increased in SSM of gastrocnemius. Unexpectedly, HFD affected mitochondria comparably to PO. In combination, PO and HFD showed additive effects on mitochondrial subpopulations which were reflected by isolated complex activities. While PO impaired diastolic as well as systolic cardiac function and increased glucose tolerance, HFD did not affect cardiac function but decreased glucose tolerance. We conclude that HFD and PO heart failure have comparable effects leading to more severe impairment of IFM. Glucose tolerance seems not causally related to skeletal muscle mitochondrial dysfunction. The additive effects of HFD and PO may suggest accelerated skeletal muscle mitochondrial dysfunction when heart failure is accompanied with a diet containing high fat.
Collapse
Affiliation(s)
- Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Katrin Kreuzer
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| | - Michael Schwarzer
- Department of Cardiothoracic Surgery, Jena University Hospital - Friedrich Schiller University of Jena, Jena, Germany
| |
Collapse
|
17
|
Oka SI, Sabry AD, Cawley KM, Warren JS. Multiple Levels of PGC-1α Dysregulation in Heart Failure. Front Cardiovasc Med 2020; 7:2. [PMID: 32083094 PMCID: PMC7002390 DOI: 10.3389/fcvm.2020.00002] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022] Open
Abstract
Metabolic adaption is crucial for the heart to sustain its contractile activity under various physiological and pathological conditions. At the molecular level, the changes in energy demand impinge on the expression of genes encoding for metabolic enzymes. Among the major components of an intricate transcriptional circuitry, peroxisome proliferator-activated receptor γ coactivator 1 alpha (PGC-1α) plays a critical role as a metabolic sensor, which is responsible for the fine-tuning of transcriptional responses to a plethora of stimuli. Cumulative evidence suggests that energetic impairment in heart failure is largely attributed to the dysregulation of PGC-1α. In this review, we summarize recent studies revealing how PGC-1α is regulated by a multitude of mechanisms, operating at different regulatory levels, which include epigenetic regulation, the expression of variants, post-transcriptional inhibition, and post-translational modifications. We further discuss how the PGC-1α regulatory cascade can be impaired in the failing heart.
Collapse
Affiliation(s)
- Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Amira D Sabry
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Keiko M Cawley
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Junco S Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States.,Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
18
|
Whitehead N, Gill JF, Brink M, Handschin C. Moderate Modulation of Cardiac PGC-1α Expression Partially Affects Age-Associated Transcriptional Remodeling of the Heart. Front Physiol 2018; 9:242. [PMID: 29618980 PMCID: PMC5871735 DOI: 10.3389/fphys.2018.00242] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/06/2018] [Indexed: 01/09/2023] Open
Abstract
Aging is associated with a decline in cardiac function due to a decreased myocardial reserve. This adverse cardiac remodeling comprises of a variety of changes, including a reduction in mitochondrial function and a decline in the expression of the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a central regulator of mitochondrial biogenesis and metabolic adaptation in the myocardium. To study the etiological involvement of PGC-1α in cardiac aging, we used mouse models mimicking the modest down- and upregulation of this coactivator in the old and the exercised heart, respectively. Young mice with reduced cardiac expression of PGC-1α recapitulated part of the age-related impairment in mitochondrial gene expression, but otherwise did not aggravate the aging process. Inversely however, moderate overexpression of PGC-1α counteracts numerous key age-related remodeling changes, e.g., by improving blood pressure, age-associated apoptosis, and collagen accumulation, as well as in the expression of many, but not all cardiac genes involved in mitochondrial biogenesis, dynamics, metabolism, calcium handling and contractility. Thus, while the reduction of PGC-1α in the heart is insufficient to cause an aging phenotype, moderate overexpression reduces pathological remodeling of older hearts and could thereby contribute to the beneficial effects of exercise on cardiac function in aging.
Collapse
Affiliation(s)
| | | | - Marijke Brink
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | | |
Collapse
|
19
|
Abstract
Mitochondria play a crucial role in a variety of cellular processes ranging from energy metabolism, generation of reactive oxygen species (ROS) and Ca(2+) handling to stress responses, cell survival and death. Malfunction of the organelle may contribute to the pathogenesis of neuromuscular, cancer, premature aging and cardiovascular diseases (CVD), including myocardial ischemia, cardiomyopathy and heart failure (HF). Mitochondria contain their own genome organized into DNA-protein complexes, called "mitochondrial nucleoids," along with multiprotein machineries, which promote mitochondrial DNA (mtDNA) replication, transcription and repair. Although the mammalian organelle possesses almost all known nuclear DNA repair pathways, including base excision repair, mismatch repair and recombinational repair, the proximity of mtDNA to the main sites of ROS production and the lack of protective histones may result in increased susceptibility to various types of mtDNA damage. These include accumulation of mtDNA point mutations and/or deletions and decreased mtDNA copy number, which will impair mitochondrial function and finally, may lead to CVD including HF.
Collapse
Affiliation(s)
- José Marín-García
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Avenue, Highland Park, NJ, 08904, USA.
| |
Collapse
|
20
|
Diguet N, Trammell SAJ, Tannous C, Deloux R, Piquereau J, Mougenot N, Gouge A, Gressette M, Manoury B, Blanc J, Breton M, Decaux JF, Lavery GG, Baczkó I, Zoll J, Garnier A, Li Z, Brenner C, Mericskay M. Nicotinamide Riboside Preserves Cardiac Function in a Mouse Model of Dilated Cardiomyopathy. Circulation 2017; 137:2256-2273. [PMID: 29217642 DOI: 10.1161/circulationaha.116.026099] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 11/06/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Myocardial metabolic impairment is a major feature in chronic heart failure. As the major coenzyme in fuel oxidation and oxidative phosphorylation and a substrate for enzymes signaling energy stress and oxidative stress response, nicotinamide adenine dinucleotide (NAD+) is emerging as a metabolic target in a number of diseases including heart failure. Little is known on the mechanisms regulating homeostasis of NAD+ in the failing heart. METHODS To explore possible alterations of NAD+ homeostasis in the failing heart, we quantified the expression of NAD+ biosynthetic enzymes in the human failing heart and in the heart of a mouse model of dilated cardiomyopathy (DCM) triggered by Serum Response Factor transcription factor depletion in the heart (SRFHKO) or of cardiac hypertrophy triggered by transverse aorta constriction. We studied the impact of NAD+ precursor supplementation on cardiac function in both mouse models. RESULTS We observed a 30% loss in levels of NAD+ in the murine failing heart of both DCM and transverse aorta constriction mice that was accompanied by a decrease in expression of the nicotinamide phosphoribosyltransferase enzyme that recycles the nicotinamide precursor, whereas the nicotinamide riboside kinase 2 (NMRK2) that phosphorylates the nicotinamide riboside precursor is increased, to a higher level in the DCM (40-fold) than in transverse aorta constriction (4-fold). This shift was also observed in human failing heart biopsies in comparison with nonfailing controls. We show that the Nmrk2 gene is an AMP-activated protein kinase and peroxisome proliferator-activated receptor α responsive gene that is activated by energy stress and NAD+ depletion in isolated rat cardiomyocytes. Nicotinamide riboside efficiently rescues NAD+ synthesis in response to FK866-mediated inhibition of nicotinamide phosphoribosyltransferase and stimulates glycolysis in cardiomyocytes. Accordingly, we show that nicotinamide riboside supplementation in food attenuates the development of heart failure in mice, more robustly in DCM, and partially after transverse aorta constriction, by stabilizing myocardial NAD+ levels in the failing heart. Nicotinamide riboside treatment also robustly increases the myocardial levels of 3 metabolites, nicotinic acid adenine dinucleotide, methylnicotinamide, and N1-methyl-4-pyridone-5-carboxamide, that can be used as validation biomarkers for the treatment. CONCLUSIONS The data show that nicotinamide riboside, the most energy-efficient among NAD precursors, could be useful for treatment of heart failure, notably in the context of DCM, a disease with few therapeutic options.
Collapse
Affiliation(s)
- Nicolas Diguet
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Samuel A J Trammell
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City (S.A.J.T., C.B.)
| | - Cynthia Tannous
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.).,Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Robin Deloux
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.).,Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | | | - Nathalie Mougenot
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Plateforme PECMV, UMS28, Paris, France (N.M.)
| | - Anne Gouge
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Mélanie Gressette
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Boris Manoury
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Jocelyne Blanc
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.).,Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Marie Breton
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Jean-François Decaux
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, United Kingdom (G.G.L.)
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Hungary (I.B.)
| | - Joffrey Zoll
- Physiology Department, Faculty of Medicine and EA3072, Université de Strasbourg, France (J.Z.)
| | - Anne Garnier
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.)
| | - Zhenlin Li
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, Department of Biology of Adaptation and Ageing, CNRS UMR8256, INSERM U1164, Institute of Biology Paris-Seine, DHU FAST, France (N.D., C.T., R.D., A. Gouge, J.B., J.-F.D., Z.L.)
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City (S.A.J.T., C.B.)
| | - Mathias Mericskay
- Signalling and Cardiovascular Pathophysiology, UMR-S 1180, University Paris-Sud, INSERM, Université Paris- Saclay, Châtenay-Malabry, France (C.T., R.D., J.P., M.G., B.M., M.B., A. Garnier, M.M.).
| |
Collapse
|
21
|
Peterzan MA, Lygate CA, Neubauer S, Rider OJ. Metabolic remodeling in hypertrophied and failing myocardium: a review. Am J Physiol Heart Circ Physiol 2017. [PMID: 28646030 DOI: 10.1152/ajpheart.00731.2016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The energy starvation hypothesis proposes that maladaptive metabolic remodeling antedates, initiates, and maintains adverse contractile dysfunction in heart failure (HF). Better understanding of the cardiac metabolic phenotype and metabolic signaling could help identify the role metabolic remodeling plays within HF and the conditions known to transition toward HF, including "pathological" hypertrophy. In this review, we discuss metabolic phenotype and metabolic signaling in the contexts of pathological hypertrophy and HF. We discuss the significance of alterations in energy supply (substrate utilization, oxidative capacity, and phosphotransfer) and energy sensing using observations from human and animal disease models and models of manipulated energy supply/sensing. We aim to provide ways of thinking about metabolic remodeling that center around metabolic flexibility, capacity (reserve), and efficiency rather than around particular substrate preferences or transcriptomic profiles. We show that maladaptive metabolic remodeling takes multiple forms across multiple energy-handling domains. We suggest that lack of metabolic flexibility and reserve (substrate, oxidative, and phosphotransfer) represents a final common denominator ultimately compromising efficiency and contractile reserve in stressful contexts.
Collapse
Affiliation(s)
- Mark A Peterzan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Oliver J Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
22
|
Gupta A, Houston B. A comprehensive review of the bioenergetics of fatty acid and glucose metabolism in the healthy and failing heart in nondiabetic condition. Heart Fail Rev 2017; 22:825-842. [DOI: 10.1007/s10741-017-9623-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Pena MJ, de Zeeuw D, Andress D, Brennan JJ, Correa-Rotter R, Coll B, Kohan DE, Makino H, Perkovic V, Remuzzi G, Tobe SW, Toto R, Parving HH, Sharma S, Corringham T, Sharma K, Heerspink HJL. The effects of atrasentan on urinary metabolites in patients with type 2 diabetes and nephropathy. Diabetes Obes Metab 2017; 19:749-753. [PMID: 28019071 DOI: 10.1111/dom.12864] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/08/2016] [Accepted: 12/21/2016] [Indexed: 01/23/2023]
Abstract
We assessed the effect of atrasentan therapy on a pre-specified panel of 13 urinary metabolites known to reflect mitochondrial function in patients with diabetic kidney disease. This post-hoc analysis was performed using urine samples collected during the RADAR study which was a randomized, double-blind, placebo-controlled trial that tested the effects of atrasentan on albuminuria reduction in patients with type 2 diabetes and nephropathy. At baseline, 4 of the 13 metabolites, quantified by gas-chromatography mass spectrometry, were below detectable levels, and 6 were reduced in patients with eGFR < 60 mL/min/1.73 m2 . After 12 weeks of atrasentan treatment in patients with eGFR < 60 mL/min/1.73 m2 , a single-value index of the metabolites changed by -0.31 (95%CI -0.60 to -0.02; P = .035), -0.08 (-12 to 0.29; P = .43) and 0.01 (-0.21 to 0.19; P = .913) in placebo, atrasentan 0.75 and 1.25 mg/d, respectively. The metabolite index difference compared to placebo was 0.13 (-0.17 to 0.43; P = .40) and 0.35 (0.05-0.65; P = .02) for atrasentan 0.75 and 1.25 mg/d, respectively. These data corroborate previous findings of mitochondrial dysfunction in patients with type 2 diabetes, nephropathy and eGFR < 60 mL/min/1.73 m2 , suggesting that atrasentan may prevent the progression of mitochondrial dysfunction common to this specific patient population. Future studies of longer treatment duration with atrasentan are indicated.
Collapse
Affiliation(s)
- Michelle J Pena
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dick de Zeeuw
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dennis Andress
- Renal Clinical Development, AbbVie, North Chicago, Illinois
| | - John J Brennan
- Renal Clinical Development, AbbVie, North Chicago, Illinois
| | - Ricardo Correa-Rotter
- Department of Nephrology and Mineral Metabolism, National Medical Science and Nutrition Institute Salvador Zubirán, Mexico City, Mexico
| | - Blai Coll
- Clinical Research Cardiovascular, Amgen Inc., Thousand Oaks, California
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Hirofumi Makino
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Vlado Perkovic
- George Institute for Global Health, University of Sydney, Sydney, Australia
| | - Giuseppe Remuzzi
- Unit of Nephrology and Dialysis, Azienda Ospedaliera Papa Giovanni XXIII, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Sheldon W Tobe
- Department of Hypertension and Nephrology, Sunnybrook Health Sciences Center, Toronto, Ontario, Canada
| | - Robert Toto
- Department of Clinical Science, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hans-Henrik Parving
- Department of Medical Endocrinology, Rigshospitalet University Hospital of Copenhagen, Copenhagen, Denmark
| | | | | | - Kumar Sharma
- Institute of Metabolomic Medicine, Center for Renal Translational Medicine, Division of Nephrology-Hypertension, Division of Medical Genetics, Department of Medicine, University of California San Diego, San Diego, California
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
24
|
Melenovsky V, Petrak J, Mracek T, Benes J, Borlaug BA, Nuskova H, Pluhacek T, Spatenka J, Kovalcikova J, Drahota Z, Kautzner J, Pirk J, Houstek J. Myocardial iron content and mitochondrial function in human heart failure: a direct tissue analysis. Eur J Heart Fail 2016; 19:522-530. [PMID: 27647766 DOI: 10.1002/ejhf.640] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/13/2016] [Accepted: 07/29/2016] [Indexed: 11/08/2022] Open
Abstract
AIMS Iron replacement improves clinical status in iron-deficient patients with heart failure (HF), but the pathophysiology is poorly understood. Iron is essential not only for erythropoiesis, but also for cellular bioenergetics. The impact of myocardial iron deficiency (MID) on mitochondrial function, measured directly in the failing human heart, is unknown. METHODS AND RESULTS Left ventricular samples were obtained from 91 consecutive HF patients undergoing transplantation and 38 HF-free organ donors (controls). Total myocardial iron content, mitochondrial respiration, citric acid cycle and respiratory chain enzyme activities, respiratory chain components (complex I-V), and protein content of reactive oxygen species (ROS)-protective enzymes were measured in tissue homogenates to quantify mitochondrial function. Myocardial iron content was lower in HF compared with controls (156 ± 41 vs. 200 ± 38 µg·g-1 dry weight, P < 0.001), independently of anaemia. MID (the lowest iron tercile in HF) was associated with more extensive coronary disease and less beta-blocker usage compared with non-MID HF patients. Compared with controls, HF patients displayed reduced myocardial oxygen2 respiration and reduced activity of all examined mitochondrial enzymes (all P < 0.001). MID in HF was associated with preserved activity of respiratory chain enzymes but reduced activity of aconitase and citrate synthase (by -26% and -15%, P < 0.05) and reduced expression of catalase, glutathione peroxidase, and superoxide dismutase 2. CONCLUSION Myocardial iron content is decreased and mitochondrial functions are impaired in advanced HF. MID in HF is associated with diminished citric acid cycle enzyme activities and decreased ROS-protecting enzymes. MID may contribute to altered myocardial substrate use and to worsening of mitochondrial dysfunction that exists in HF.
Collapse
Affiliation(s)
- Vojtech Melenovsky
- Department of Cardiology and Cardiac Surgery, Institute of Clinical and Experimental Medicine-IKEM, Prague, Czech Republic
| | - Jiri Petrak
- Department of Pathological Physiology and BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomas Mracek
- Department of Bioenergetics, Institute of Physiology, Academy of Sciences, Prague, Czech Republic
| | - Jan Benes
- Department of Cardiology and Cardiac Surgery, Institute of Clinical and Experimental Medicine-IKEM, Prague, Czech Republic
| | - Barry A Borlaug
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Hana Nuskova
- Department of Bioenergetics, Institute of Physiology, Academy of Sciences, Prague, Czech Republic
| | - Tomas Pluhacek
- Department of Analytical Chemistry and RCPTM, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Jaroslav Spatenka
- Department of Transplantation and Tissue Bank, University Hospital in Motol, Prague, Czech Republic
| | - Jana Kovalcikova
- Department of Bioenergetics, Institute of Physiology, Academy of Sciences, Prague, Czech Republic
| | - Zdenek Drahota
- Department of Bioenergetics, Institute of Physiology, Academy of Sciences, Prague, Czech Republic
| | - Josef Kautzner
- Department of Cardiology and Cardiac Surgery, Institute of Clinical and Experimental Medicine-IKEM, Prague, Czech Republic
| | - Jan Pirk
- Department of Cardiology and Cardiac Surgery, Institute of Clinical and Experimental Medicine-IKEM, Prague, Czech Republic
| | - Josef Houstek
- Department of Bioenergetics, Institute of Physiology, Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
25
|
Schlagowski AI, Isner-Horobeti ME, Dufour SP, Rasseneur L, Enache I, Lonsdorfer-Wolf E, Doutreleau S, Charloux A, Goupilleau F, Bentz I, Charles AL, Kouassi BY, Zoll J, Geny B, Favret F. Mitochondrial function following downhill and/or uphill exercise training in rats. Muscle Nerve 2016; 54:925-935. [PMID: 27064266 DOI: 10.1002/mus.25144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2016] [Indexed: 01/02/2023]
Abstract
INTRODUCTION The goal of this study was to compare the effects of downhill (DH), uphill (UH), and UH-DH exercise training, at the same metabolic rate, on exercise capacity and skeletal muscle mitochondrial function. METHODS Thirty-two Wistar rats were separated into a control and 3 trained groups. The trained groups exercised for 4 weeks, 5 times per week at the same metabolic rate, either in UH, DH, or combined UH-DH. Twenty-four hours after the last training session, the soleus, gastrocnemius, and vastus intermedius muscles were removed for assessment of mitochondrial respiration. RESULTS Exercise training, at the same metabolic rate, improved maximal running speed without specificity for exercise modalities. Maximal fiber respiration was enhanced in soleus and vastus intermedius in the UH group only. CONCLUSIONS Exercise training, performed at the same metabolic rate, improved exercise capacity, but only UH-trained rats enhanced mitochondrial function in both soleus and vastus intermedius skeletal muscle. Muscle Nerve 54: 925-935, 2016.
Collapse
Affiliation(s)
- Anna-Isabel Schlagowski
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,CHRU of Strasbourg, Physiology and Functional Explorations Department, New Civil Hospital, Strasbourg, France
| | - Marie-Eve Isner-Horobeti
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,Department of Physical and Rehabilitation Medicine, University Institute of Rehabilitation Clémenceau, Strasbourg, France
| | - Stéphane P Dufour
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,Strasbourg University, Faculty of Sport Sciences, Strasbourg, France
| | - Laurence Rasseneur
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,Strasbourg University, Faculty of Sport Sciences, Strasbourg, France
| | - Irina Enache
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,CHRU of Strasbourg, Physiology and Functional Explorations Department, New Civil Hospital, Strasbourg, France
| | - Evelyne Lonsdorfer-Wolf
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,CHRU of Strasbourg, Physiology and Functional Explorations Department, New Civil Hospital, Strasbourg, France
| | - Stéphane Doutreleau
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,CHRU of Strasbourg, Physiology and Functional Explorations Department, New Civil Hospital, Strasbourg, France
| | - Anne Charloux
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,CHRU of Strasbourg, Physiology and Functional Explorations Department, New Civil Hospital, Strasbourg, France
| | - Fabienne Goupilleau
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France
| | - Isabelle Bentz
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France
| | - Anne Laure Charles
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France
| | - Blah Y Kouassi
- Strasbourg University, Faculty of Sport Sciences, Strasbourg, France
| | - Joffrey Zoll
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,CHRU of Strasbourg, Physiology and Functional Explorations Department, New Civil Hospital, Strasbourg, France
| | - Bernard Geny
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France.,CHRU of Strasbourg, Physiology and Functional Explorations Department, New Civil Hospital, Strasbourg, France
| | - Fabrice Favret
- Strasbourg University, Faculty of Medicine, Fédération de Médecine Translationnelle de Strasbourg, EA3072 "Mitochondria, Oxidative Stress and Muscular Protection," 4 rue Kirschleger, 67085, Strasbourg, France. .,Strasbourg University, Faculty of Sport Sciences, Strasbourg, France.
| |
Collapse
|
26
|
Abstract
Mitochondrial dynamics, fission and fusion, were first identified in yeast with investigation in heart cells beginning only in the last 5 to 7 years. In the ensuing time, it has become evident that these processes are not only required for healthy mitochondria, but also, that derangement of these processes contributes to disease. The fission and fusion proteins have a number of functions beyond the mitochondrial dynamics. Many of these functions are related to their membrane activities, such as apoptosis. However, other functions involve other areas of the mitochondria, such as OPA1's role in maintaining cristae structure and preventing cytochrome c leak, and its essential (at least a 10 kDa fragment of OPA1) role in mtDNA replication. In heart disease, changes in expression of these important proteins can have detrimental effects on mitochondrial and cellular function.
Collapse
Affiliation(s)
- A A Knowlton
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| | - T T Liu
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| |
Collapse
|
27
|
Liu XP, Gao H, Huang XY, Chen YF, Feng XJ, He YH, Li ZM, Liu PQ. Peroxisome proliferator-activated receptor gamma coactivator 1 alpha protects cardiomyocytes from hypertrophy by suppressing calcineurin-nuclear factor of activated T cells c4 signaling pathway. Transl Res 2015; 166:459-473.e3. [PMID: 26118953 DOI: 10.1016/j.trsl.2015.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/21/2015] [Accepted: 06/02/2015] [Indexed: 01/11/2023]
Abstract
Peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) is a crucial coregulator interacting with multiple transcriptional factors in the regulation of cardiac hypertrophy. The present study revealed that PGC-1α protected cardiomyocytes from hypertrophy by suppressing calcineurin-nuclear factor of activated T cells c4 (NFATc4) signaling pathway. Overexpression of PGC-1α by adenovirus infection prevented the increased protein and messenger RNA expression of NFATc4 in phenylephrine (PE)-treated hypertrophic cardiomyocytes, whereas knockdown of PGC-1α by RNA silencing augmented the expression of NFATc4. An interaction between PGC-1α and NFATc4 was observed in both the cytoplasm and nucleus of neonatal rat cardiomyocytes. Adenovirus PGC-1α prevented the nuclear import of NFATc4 and increased its phosphorylation level of NFATc4, probably through repressing the expression and activity of calcineurin and interfering with the interaction between calcineurin and NFATc4. On the contrary, PGC-1α silencing aggravated PE-induced calcineurin activation, NFATc4 dephosphorylation, and nuclear translocation. Moreover, the binding activity and transcription activity of NFATc4 to DNA promoter of brain natriuretic peptide were abrogated by PGC-1α overexpression but were enhanced by PGC-1α knockdown. The effect of PGC-1α on suppressing the calcinuerin-NFATc4 signaling pathway might at least partially contribute to the protective effect of PGC-1α on cardiomyocyte hypertrophy. These findings provide novel insights into the role of PGC-1α in regulation of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xue-Ping Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China
| | - Hui Gao
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China; Department of Pharmacology, School of Medicine, Jishou University, Jishou, PR China
| | - Xiao-Yang Huang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China
| | - Yan-Fang Chen
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Xiao-Jun Feng
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China
| | - Yan-Hong He
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China
| | - Zhuo-Ming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China.
| | - Pei-Qing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China.
| |
Collapse
|
28
|
Akhmedov AT, Marín-García J. Mitochondrial DNA maintenance: an appraisal. Mol Cell Biochem 2015; 409:283-305. [DOI: 10.1007/s11010-015-2532-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/06/2015] [Indexed: 12/13/2022]
|
29
|
Abstract
The ultrastructure of the cardiac myocyte is remarkable for the high density of mitochondria tightly packed between sarcomeres. This structural organization is designed to provide energy in the form of ATP to fuel normal pump function of the heart. A complex system comprised of regulatory factors and energy metabolic machinery, encoded by both mitochondrial and nuclear genomes, is required for the coordinate control of cardiac mitochondrial biogenesis, maturation, and high-capacity function. This process involves the action of a transcriptional regulatory network that builds and maintains the mitochondrial genome and drives the expression of the energy transduction machinery. This finely tuned system is responsive to developmental and physiological cues, as well as changes in fuel substrate availability. Deficiency of components critical for mitochondrial energy production frequently manifests as a cardiomyopathic phenotype, underscoring the requirement to maintain high respiration rates in the heart. Although a precise causative role is not clear, there is increasing evidence that perturbations in this regulatory system occur in the hypertrophied and failing heart. This review summarizes current knowledge and highlights recent advances in our understanding of the transcriptional regulatory factors and signaling networks that serve to regulate mitochondrial biogenesis and function in the mammalian heart.
Collapse
Affiliation(s)
- Rick B Vega
- From the Diabetes and Obesity Research Center, Cardiovascular Pathobiology Program, Sanford-Burnham Medical Research Institute, Orlando, FL
| | - Julie L Horton
- From the Diabetes and Obesity Research Center, Cardiovascular Pathobiology Program, Sanford-Burnham Medical Research Institute, Orlando, FL
| | - Daniel P Kelly
- From the Diabetes and Obesity Research Center, Cardiovascular Pathobiology Program, Sanford-Burnham Medical Research Institute, Orlando, FL.
| |
Collapse
|
30
|
Tetrahydrocannabinol induces brain mitochondrial respiratory chain dysfunction and increases oxidative stress: a potential mechanism involved in cannabis-related stroke. BIOMED RESEARCH INTERNATIONAL 2015; 2015:323706. [PMID: 25654095 PMCID: PMC4310259 DOI: 10.1155/2015/323706] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/15/2014] [Accepted: 09/22/2014] [Indexed: 12/02/2022]
Abstract
Cannabis has potential therapeutic use but tetrahydrocannabinol (THC), its main psychoactive component, appears as a risk factor for ischemic stroke in young adults. We therefore evaluate the effects of THC on brain mitochondrial function and oxidative stress, key factors involved in stroke. Maximal oxidative capacities Vmax (complexes I, III, and IV activities), Vsucc (complexes II, III, and IV activities), Vtmpd (complex IV activity), together with mitochondrial coupling (Vmax/V0), were determined in control conditions and after exposure to THC in isolated mitochondria extracted from rat brain, using differential centrifugations. Oxidative stress was also assessed through hydrogen peroxide (H2O2) production, measured with Amplex Red. THC significantly decreased Vmax (−71%; P < 0.0001), Vsucc (−65%; P < 0.0001), and Vtmpd (−3.5%; P < 0.001). Mitochondrial coupling (Vmax/V0) was also significantly decreased after THC exposure (1.8±0.2 versus 6.3±0.7; P < 0.001). Furthermore, THC significantly enhanced H2O2 production by cerebral mitochondria (+171%; P < 0.05) and mitochondrial free radical leak was increased from 0.01±0.01 to 0.10±0.01% (P < 0.001). Thus, THC increases oxidative stress and induces cerebral mitochondrial dysfunction. This mechanism may be involved in young cannabis users who develop ischemic stroke since THC might increase patient's vulnerability to stroke.
Collapse
|
31
|
Villena JA. New insights into PGC-1 coactivators: redefining their role in the regulation of mitochondrial function and beyond. FEBS J 2015; 282:647-72. [DOI: 10.1111/febs.13175] [Citation(s) in RCA: 252] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/31/2014] [Accepted: 12/10/2014] [Indexed: 12/25/2022]
Affiliation(s)
- Josep A. Villena
- Laboratory of Metabolism and Obesity; Vall d'Hebron-Institut de Recerca; Universitat Autònoma de Barcelona; Spain
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabólicas Asociadas); Instituto de Salud Carlos III; Barcelona Spain
| |
Collapse
|
32
|
Moulin M, Piquereau J, Mateo P, Fortin D, Rucker-Martin C, Gressette M, Lefebvre F, Gresikova M, Solgadi A, Veksler V, Garnier A, Ventura-Clapier R. Sexual Dimorphism of Doxorubicin-Mediated Cardiotoxicity. Circ Heart Fail 2015; 8:98-108. [DOI: 10.1161/circheartfailure.114.001180] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background—
Cardiovascular diseases are the major cause of mortality among both men and women with a lower incidence in women before menopause. The clinical use of doxorubicin, widely used as an antineoplastic agent, is markedly hampered by severe cardiotoxicity. Even if there is a significant sex difference in incidence of cardiovascular disease at the adult stage, it is not known whether a difference in doxorubicin-related cardiotoxicity between men and women also exists. The objective of this work was to explore the cardiac side effects of doxorubicin in adult rats and decipher whether signaling pathways involved in cardiac toxicity differ between sexes.
Methods and Results—
After 7 weeks of doxorubicin (2 mg/kg per week), males developed major signs of cardiomyopathy with cardiac atrophy, reduced left ventricular ejection fraction and 50% mortality. In contrast, no female died and their left ventricular ejection fraction was only moderately affected. Surprisingly, neither global oxidation levels nor the antioxidant response nor the apoptosis signaling pathways were altered by doxorubicin. However, the level of total adenosine monophosphate–activated protein kinase was severely decreased only in males. Moreover, markers of mitochondrial biogenesis and cardiolipin content were strongly reduced only in males. To analyze the onset of the pathology, maximal oxygen consumption rate of left ventricular permeabilized fibers after 4 weeks of treatment was reduced only in doxorubicin-treated males.
Conclusions—
Altogether, these results clearly evidence sex differences in doxorubicin toxicity. Cardiac mitochondrial dysfunction and adenosine monophosphate–activated protein kinase seem as critical sites of sex differences in cardiotoxicity as evidenced by significant statistical interactions between sex and treatment effects.
Collapse
Affiliation(s)
- Maryline Moulin
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Jérôme Piquereau
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Philippe Mateo
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Dominique Fortin
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Catherine Rucker-Martin
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Mélanie Gressette
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Florence Lefebvre
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Milada Gresikova
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Audrey Solgadi
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Vladimir Veksler
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Anne Garnier
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| | - Renée Ventura-Clapier
- From the INSERM UMR-S 769, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., M.G., F.L., M.G., V.V., A.G., R.V.-C.); IPSIT-IFR141 Université de Paris-Sud, Châtenay-Malabry, France (M.M., J.P., P.M., D.F., C.R.-M., M.G., F.L., M.G., A.S., V.V., A.G., R.V.-C.); INSERM UMR-S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (C.R.-M.); and IPSIT-IFR141 Service d’Analyse des Médicaments et Métabolites, Châtenay-Malabry, France (A.S.)
| |
Collapse
|
33
|
Thibault R, Meyer P, Cano N. Activité physique, nutrition, et insuffisance cardiaque chronique. NUTR CLIN METAB 2014. [DOI: 10.1016/j.nupar.2014.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Heart failure and mitochondrial dysfunction: the role of mitochondrial fission/fusion abnormalities and new therapeutic strategies. J Cardiovasc Pharmacol 2014; 63:196-206. [PMID: 23884159 DOI: 10.1097/01.fjc.0000432861.55968.a6] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The treatment of heart failure (HF) has evolved during the past 30 years with the recognition of neurohormonal activation and the effectiveness of its inhibition in improving the quality of life and survival. Over the past 20 years, there has been a revolution in the investigation of the mitochondrion with the development of new techniques and the finding that mitochondria are connected in networks and undergo constant division (fission) and fusion, even in cardiac myocytes. This has led to new molecular and cellular discoveries in HF, which offer the potential for the development of new molecular-based therapies. Reactive oxygen species are an important cause of mitochondrial and cellular injury in HF, but there are other abnormalities, such as depressed mitochondrial fusion, that may eventually become the targets of at least episodic treatment. The overall need for mitochondrial fission/fusion balance may preclude sustained change in either fission or fusion. In this review, we will discuss the current HF therapy and its impact on the mitochondria. In addition, we will review some of the new drug targets under development. There is potential for effective, novel therapies for HF to arise from new molecular understanding.
Collapse
|
35
|
Abstract
The concept of skeletal muscle myopathy as a main determinant of exercise intolerance in chronic heart failure (HF) is gaining acceptance. Symptoms that typify HF patients, including shortness of breath and fatigue, are often directly related to the abnormalities of the skeletal muscle in HF. Besides muscular wasting, alterations in skeletal muscle energy metabolism, including insulin resistance, have been implicated in HF. Adiponectin, an adipocytokine with insulin-sensitizing properties, receives increasing interest in HF. Circulating adiponectin levels are elevated in HF patients, but high levels are paradoxically associated with poor outcome. Previous analysis of m. vastus lateralis biopsies in HF patients highlighted a striking functional adiponectin resistance. Together with increased circulating adiponectin levels, adiponectin expression within the skeletal muscle is elevated in HF patients, whereas the expression of the main adiponectin receptor and genes involved in the downstream pathway of lipid and glucose metabolism is downregulated. In addition, the adiponectin-related metabolic disturbances strongly correlate with aerobic capacity (VO2 peak), sub-maximal exercise performance and muscle strength. These observations strengthen our hypothesis that adiponectin and its receptors play a key role in the development and progression of the "heart failure myopathy". The question whether adiponectin exerts beneficial rather than detrimental effects in HF is still left unanswered. This current research overview will elucidate the emerging role of adiponectin in HF and suggests potential therapeutic targets to tackle energy wasting in these patients.
Collapse
|
36
|
Gupte AA, Hamilton DJ, Cordero-Reyes AM, Youker KA, Yin Z, Estep JD, Stevens RD, Wenner B, Ilkayeva O, Loebe M, Peterson LE, Lyon CJ, Wong STC, Newgard CB, Torre-Amione G, Taegtmeyer H, Hsueh WA. Mechanical unloading promotes myocardial energy recovery in human heart failure. ACTA ACUST UNITED AC 2014; 7:266-76. [PMID: 24825877 DOI: 10.1161/circgenetics.113.000404] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Impaired bioenergetics is a prominent feature of the failing heart, but the underlying metabolic perturbations are poorly understood. METHODS AND RESULTS We compared metabolomic, gene transcript, and protein data from 6 paired samples of failing human left ventricular tissue obtained during left ventricular assist device insertion (heart failure samples) and at heart transplant (post-left ventricular assist device samples). Nonfailing left ventricular wall samples procured from explanted hearts of patients with right heart failure served as novel comparison samples. Metabolomic analyses uncovered a distinct pattern in heart failure tissue: 2.6-fold increased pyruvate concentrations coupled with reduced Krebs cycle intermediates and short-chain acylcarnitines, suggesting a global reduction in substrate oxidation. These findings were associated with decreased transcript levels for enzymes that catalyze fatty acid oxidation and pyruvate metabolism and for key transcriptional regulators of mitochondrial metabolism and biogenesis, peroxisome proliferator-activated receptor γ coactivator 1α (PGC1A, 1.3-fold) and estrogen-related receptor α (ERRA, 1.2-fold) and γ (ERRG, 2.2-fold). Thus, parallel decreases in key transcription factors and their target metabolic enzyme genes can explain the decreases in associated metabolic intermediates. Mechanical support with left ventricular assist device improved all of these metabolic and transcriptional defects. CONCLUSIONS These observations underscore an important pathophysiologic role for severely defective metabolism in heart failure, while the reversibility of these defects by left ventricular assist device suggests metabolic resilience of the human heart.
Collapse
Affiliation(s)
- Anisha A Gupte
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Dale J Hamilton
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Andrea M Cordero-Reyes
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Keith A Youker
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Zheng Yin
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Jerry D Estep
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Robert D Stevens
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Brett Wenner
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Olga Ilkayeva
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Matthias Loebe
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Leif E Peterson
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Christopher J Lyon
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Stephen T C Wong
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Christopher B Newgard
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Guillermo Torre-Amione
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Heinrich Taegtmeyer
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.)
| | - Willa A Hsueh
- From the Methodist Diabetes and Metabolism Institute, Houston Methodist Research Institute, Houston, TX (A.A.G., D.J.H., C.J.L., W.A.H.); Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX (Z.Y., S.T.C.W.); Center for Biostatistics, Houston Methodist Research Institute, Houston, TX (L.E.P.); Department of Medicine, Houston Methodist Hospital, Houston, TX (D.J.H., W.A.H.), Department of Radiology, Houston Methodist Hospital, Houston, TX (S.T.C.W.); Methodist DeBakey Heart and Vascular Institute, Houston, TX (A.M.C.-R., K.A.Y., J.D.E., M.L., G.T.-A.); Weill Cornell Medical College, New York, NY (A.A.G., D.J.H., A.M.C.-R., K.A.Y., Z.Y., J.D.E., M.L., L.E.P., C.J.L., S.T.C.W., G.T.-A., W.A.H.); Sarah W. Stedman Nutrition and Metabolism Center and Departments of Pharmacology and Cancer Biology and Medicine, Duke University Medical Center (R.D.S., B.W., O.L., C.B.N.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY (S.T.C.W.); Catedra de Cardiologia, Instituto Tecnologico de Monterrey, Monterrey, Mexico (G.T.-A.); The University of Texas Medical School at Houston, Houston, TX (H.T.).
| |
Collapse
|
37
|
Stride N, Larsen S, Hey-Mogensen M, Sander K, Lund JT, Gustafsson F, Køber L, Dela F. Decreased mitochondrial oxidative phosphorylation capacity in the human heart with left ventricular systolic dysfunction. Eur J Heart Fail 2014; 15:150-7. [DOI: 10.1093/eurjhf/hfs172] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Nis Stride
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences; University of Copenhagen; Blegdamsvej 3b DK-2200 Copenhagen Denmark
| | - Steen Larsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences; University of Copenhagen; Blegdamsvej 3b DK-2200 Copenhagen Denmark
| | - Martin Hey-Mogensen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences; University of Copenhagen; Blegdamsvej 3b DK-2200 Copenhagen Denmark
| | - Kåre Sander
- Department of Cardiothoracic Surgery; University of Copenhagen; Copenhagen Denmark
| | - Jens T. Lund
- Department of Cardiothoracic Surgery; University of Copenhagen; Copenhagen Denmark
| | - Finn Gustafsson
- Department of Cardiology, Rigshospitalet; University of Copenhagen; Copenhagen Denmark
| | - Lars Køber
- Department of Cardiology, Rigshospitalet; University of Copenhagen; Copenhagen Denmark
| | - Flemming Dela
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences; University of Copenhagen; Blegdamsvej 3b DK-2200 Copenhagen Denmark
| |
Collapse
|
38
|
Meyer A, Charles AL, Zoll J, Guillot M, Lejay A, Singh F, Schlagowski AI, Isner-Horobeti ME, Pistea C, Charloux A, Geny B. Cryopreservation with dimethyl sulfoxide prevents accurate analysis of skinned skeletal muscle fibers mitochondrial respiration. Biochimie 2014; 100:227-33. [PMID: 24472439 DOI: 10.1016/j.biochi.2014.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 01/17/2014] [Indexed: 10/25/2022]
Abstract
Impact of cryopreservation protocols on skeletal muscle mitochondrial respiration remains controversial. We showed that oxygen consumption with main mitochondrial substrates in rat skeletal muscles was higher in fresh samples than in cryopreserved samples and that this difference was not fixed but grow significantly with respiration rates with wide fluctuations around the mean difference. Very close results were observed whatever the muscle type and the substrate used. Importantly, the deleterious effects of ischemia-reperfusion observed on fresh samples vanished when cryopreserved samples were studied. These data demonstrate that this technic should probably be performed only extemporaneously.
Collapse
Affiliation(s)
- Alain Meyer
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Service de Physiologie et d'Explorations Fonctionnelles, Pôle de Pathologie Thoracique, Hôpitaux Universitaires de Strasbourg, 67000 Cedex, France.
| | - Anne-Laure Charles
- Service de Physiologie et d'Explorations Fonctionnelles, Pôle de Pathologie Thoracique, Hôpitaux Universitaires de Strasbourg, 67000 Cedex, France
| | - Joffrey Zoll
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Service de Physiologie et d'Explorations Fonctionnelles, Pôle de Pathologie Thoracique, Hôpitaux Universitaires de Strasbourg, 67000 Cedex, France
| | - Max Guillot
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Service de Réanimation Médicale, Pôle d'Urgences, Réanimations Médicales, Centre Antipoison, Hôpitaux Universitaire de Strasbourg, 67000 Cedex, France
| | - Anne Lejay
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Service de Chirurgie Vasculaire et de Transplantation Rénale, Pôle de cardiologie, Hôpitaux Universitaires de Strasbourg, 67000 Cedex, France
| | - François Singh
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Service de Physiologie et d'Explorations Fonctionnelles, Pôle de Pathologie Thoracique, Hôpitaux Universitaires de Strasbourg, 67000 Cedex, France
| | - Anna-Isabel Schlagowski
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Service de Physiologie et d'Explorations Fonctionnelles, Pôle de Pathologie Thoracique, Hôpitaux Universitaires de Strasbourg, 67000 Cedex, France
| | - Marie-Eve Isner-Horobeti
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Institut Universitaire de Réadaptation Clémenceau, Hôpitaux Universitaire de Strasbourg, 67000 Cedex, France
| | - Cristina Pistea
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Service de Physiologie et d'Explorations Fonctionnelles, Pôle de Pathologie Thoracique, Hôpitaux Universitaires de Strasbourg, 67000 Cedex, France
| | - Anne Charloux
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Service de Physiologie et d'Explorations Fonctionnelles, Pôle de Pathologie Thoracique, Hôpitaux Universitaires de Strasbourg, 67000 Cedex, France
| | - Bernard Geny
- Equipe d'Accueil 3072 "Mitochondries, stress oxydant et protection musculaire", Fédération de Médecine Translationnelle, Université de Strasbourg, Institut de Physiologie, 67000 Cedex, France; Service de Physiologie et d'Explorations Fonctionnelles, Pôle de Pathologie Thoracique, Hôpitaux Universitaires de Strasbourg, 67000 Cedex, France
| |
Collapse
|
39
|
Thibault R, Chanséaume S, Azarnoush K, Guillet C, Giraudet C, Patrac V, Lusson JR, Cano N, Boirie Y, Walrand S. Mitochondrial protein synthesis is increased in oxidative skeletal muscles of rats with cardiac cachexia. Nutr Res 2014; 34:250-7. [PMID: 24655492 DOI: 10.1016/j.nutres.2013.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 12/19/2013] [Accepted: 12/31/2013] [Indexed: 11/30/2022]
Abstract
Since cardiac cachexia could be associated with alterations in muscular mitochondrial metabolism, we hypothesized that the expected alterations in the activities of mitochondrial oxidative enzymes could be associated with changes in mitochondrial protein synthesis in oxidative skeletal muscles. Cardiac cachexia was provoked in male rats by the ligation of the left coronary artery. Six cachectic and 6 control rats were age-paired, and their food intake was observed. The synthesis of mitochondrial proteins was measured by [1-13C]-valine infusion in soleus, tibilais, myocardium, and liver. Muscles (soleus, gastrocnemius, and tibialis anterior), heart, kidneys, liver, and visceral adipose tissue were weighed. Mitochondrial cytochrome c oxydase IV as well as citrate synthase and myosin ATPase activities were measured. As expected, decreased food intake was observed in the cachectic group. Heart, kidney, and liver weights were higher in the cachectic group, while the visceral adipose tissue weight was lower (P < .01). No changes in muscle weights were observed. Soleus mitochondrial proteins fractional synthesis rate was higher in the cachectic group (P = .054). Cytochrome c oxydase IV activity was reduced (P = .009) and increased (P = .038) in the soleus and liver of the cachectic rats, respectively. No change in citrate synthase activity was observed. Myosin ATPase activity was reduced in the gastrocnemius of the cachectic group (P < .01). Mitochondrial protein synthesis is increased in the soleus of rats with cardiac cachexia, suggesting a compensatory mechanism of the impaired oxidative mitochondrial function. Further work should assess whether the mitochondrial protein synthesis is altered in chronic heart failure patients with cardiac cachexia, and whether this is the cause or the consequence of cachexia.
Collapse
Affiliation(s)
- Ronan Thibault
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 CLERMONT-FERRAND, Cedex, France; INRA, UMR 1019, UNH, CRNH Auvergne, F-63000 CLERMONT-FERRAND, Cedex, France; CHU Clermont-Ferrand, Service de Nutrition Clinique, F-63003 CLERMONT-FERRAND, Cedex, France; Nutrition Unit, Geneva University Hospital, Geneva, Switzerland
| | - Sylvain Chanséaume
- CHU Clermont-Ferrand, Service de Cardiologie, F-63003 CLERMONT-FERRAND, Cedex, France
| | - Kasra Azarnoush
- CHU Clermont-Ferrand, Service de Chirurgie Vasculaire, F-63003 CLERMONT-FERRAND, Cedex, France
| | - Christelle Guillet
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 CLERMONT-FERRAND, Cedex, France; INRA, UMR 1019, UNH, CRNH Auvergne, F-63000 CLERMONT-FERRAND, Cedex, France
| | - Christophe Giraudet
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 CLERMONT-FERRAND, Cedex, France; INRA, UMR 1019, UNH, CRNH Auvergne, F-63000 CLERMONT-FERRAND, Cedex, France
| | - Véronique Patrac
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 CLERMONT-FERRAND, Cedex, France; INRA, UMR 1019, UNH, CRNH Auvergne, F-63000 CLERMONT-FERRAND, Cedex, France
| | | | - Noël Cano
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 CLERMONT-FERRAND, Cedex, France; INRA, UMR 1019, UNH, CRNH Auvergne, F-63000 CLERMONT-FERRAND, Cedex, France; CHU Clermont-Ferrand, Service de Nutrition Clinique, F-63003 CLERMONT-FERRAND, Cedex, France
| | - Yves Boirie
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 CLERMONT-FERRAND, Cedex, France; INRA, UMR 1019, UNH, CRNH Auvergne, F-63000 CLERMONT-FERRAND, Cedex, France; CHU Clermont-Ferrand, Service de Nutrition Clinique, F-63003 CLERMONT-FERRAND, Cedex, France
| | - Stéphane Walrand
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 CLERMONT-FERRAND, Cedex, France; INRA, UMR 1019, UNH, CRNH Auvergne, F-63000 CLERMONT-FERRAND, Cedex, France.
| |
Collapse
|
40
|
Pottecher J, Santelmo N, Noll E, Charles AL, Benahmed M, Canuet M, Frossard N, Namer IJ, Geny B, Massard G, Diemunsch P. Cold ischemia with selective anterogradein situpulmonary perfusion preserves gas exchange and mitochondrial homeostasis and curbs inflammation in an experimental model of donation after cardiac death. Transpl Int 2013; 26:1027-37. [DOI: 10.1111/tri.12157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/07/2013] [Accepted: 06/28/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Julien Pottecher
- Department of Anaesthesiology and Critical Care; Hautepierre Hospital; Strasbourg University Hospital; Strasbourg Cedex France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS); Faculty of Medicine; Physiology Institute; EA 3072; Strasbourg University; Strasbourg France
| | - Nicola Santelmo
- Department of Thoracic Surgery; Nouvel Hôpital Civil; Strasbourg University Hospital; Strasbourg France
| | - Eric Noll
- Department of Anaesthesiology and Critical Care; Hautepierre Hospital; Strasbourg University Hospital; Strasbourg Cedex France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS); Faculty of Medicine; Physiology Institute; EA 3072; Strasbourg University; Strasbourg France
| | - Anne-Laure Charles
- Fédération de Médecine Translationnelle de Strasbourg (FMTS); Faculty of Medicine; Physiology Institute; EA 3072; Strasbourg University; Strasbourg France
- Department of Physiology; Nouvel Hôpital Civil; Strasbourg University Hospital; Strasbourg France
| | - Malika Benahmed
- ICube; UMR 7357 University of Strasbourg/CNRS; Strasbourg Cedex France
| | - Matthieu Canuet
- Department of Pneumology; Nouvel Hôpital Civil; Strasbourg University Hospital; FMTS, Faculty of Medicine, Strasbourg France
| | - Nelly Frossard
- Faculty of Pharmacy; Strasbourg University/CNRS UMR 7200; Illkirch France
| | - Izzie J. Namer
- ICube; UMR 7357 University of Strasbourg/CNRS; Strasbourg Cedex France
- Department of Biophysics and Nuclear Medicine; Hautepierre Hospital; Strasbourg University Hospital; Strasbourg Cedex France
| | - Bernard Geny
- Fédération de Médecine Translationnelle de Strasbourg (FMTS); Faculty of Medicine; Physiology Institute; EA 3072; Strasbourg University; Strasbourg France
- Department of Physiology; Nouvel Hôpital Civil; Strasbourg University Hospital; Strasbourg France
| | - Gilbert Massard
- Department of Thoracic Surgery; Nouvel Hôpital Civil; Strasbourg University Hospital; Strasbourg France
| | - Pierre Diemunsch
- Department of Anaesthesiology and Critical Care; Hautepierre Hospital; Strasbourg University Hospital; Strasbourg Cedex France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS); Faculty of Medicine; Physiology Institute; EA 3072; Strasbourg University; Strasbourg France
| |
Collapse
|
41
|
Kackstein K, Teren A, Matsumoto Y, Mangner N, Möbius-Winkler S, Linke A, Schuler G, Punkt K, Adams V. Impact of angiotensin II on skeletal muscle metabolism and function in mice: contribution of IGF-1, Sirtuin-1 and PGC-1α. Acta Histochem 2013; 115:363-70. [PMID: 23092805 DOI: 10.1016/j.acthis.2012.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Revised: 09/11/2012] [Accepted: 09/12/2012] [Indexed: 01/03/2023]
Abstract
Activation of the renin-angiotensin-aldosterone system and increased levels of angiotensin II (Ang-II) occurs in numerous cardiovascular diseases such as chronic heart failure (CHF). Another hallmark in CHF is a reduced exercise tolerance with impaired skeletal muscle function. The aim of this study was to investigate in an animal model the impact of Ang-II on skeletal muscle function and concomitant molecular alterations. Mice were infused with Ang-II for 4 weeks. Subsequently, skeletal muscle function of the soleus muscle was assessed. Expression of selected proteins was quantified by qRT-PCR and Western blot. Infusion of Ang-II resulted in a 33% reduction of contractile force, despite a lack of changes in muscle weight. At the molecular level an increased expression of NAD(P)H oxidase and a reduced expression of Sirt1, PGC-1α and IGF-1 were noticed. No change was evident for the ubiquitin E3-ligases MuRF1 and MafBx and α-sarcomeric actin expression. Cytophotometrical analysis of the soleus muscle revealed a metabolic shift toward a glycolytic profile. This study provides direct evidence of Ang-II-mediated, metabolic deterioration of skeletal muscle function despite preserved muscle mass. One may speculate that the Ang-II-mediated loss of muscle force is due to an activation of NAD(P)H oxidase expression and a subsequent ROS-induced down regulation of IGF-1, PGC-1α and Sirt1.
Collapse
|
42
|
Schultz RL, Kullman EL, Waters RP, Huang H, Kirwan JP, Gerdes AM, Swallow JG. Metabolic adaptations of skeletal muscle to voluntary wheel running exercise in hypertensive heart failure rats. Physiol Res 2013; 62:361-9. [PMID: 23590601 DOI: 10.33549/physiolres.932330] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The Spontaneously Hypertensive Heart Failure (SHHF) rat mimics the human progression of hypertension from hypertrophy to heart failure. However, it is unknown whether SHHF animals can exercise at sufficient levels to observe beneficial biochemical adaptations in skeletal muscle. Thirty-seven female SHHF and Wistar-Furth (WF) rats were randomized to sedentary (SHHFsed and WFsed) and exercise groups (SHHFex and WFex). The exercise groups had access to running wheels from 6-22 months of age. Hindlimb muscles were obtained for metabolic measures that included mitochondrial enzyme function and expression, and glycogen utilization. The SHHFex rats ran a greater distance and duration as compared to the WFex rats (P<0.05), but the WFex rats ran at a faster speed (P<0.05). Skeletal muscle citrate synthase and beta-hydroxyacyl-CoA dehydrogenase enzyme activity was not altered in the SHHFex group, but was increased (P<0.05) in the WFex animals. Citrate synthase protein and gene expression were unchanged in SHHFex animals, but were increased in WFex rats (P<0.05). In the WFex animals muscle glycogen was significantly depleted after exercise (P<0.05), but not in the SHHFex group. We conclude that despite robust amounts of aerobic activity, voluntary wheel running exercise was not sufficiently intense to improve the oxidative capacity of skeletal muscle in adult SHHF animals, indicating an inability to compensate for declining heart function by improving peripheral oxidative adaptations in the skeletal muscle.
Collapse
Affiliation(s)
- R L Schultz
- Department of Integrative Biology, University of Colorado Denver, Denver, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Ahuja P, Wanagat J, Wang Z, Wang Y, Liem DA, Ping P, Antoshechkin IA, Margulies KB, Maclellan WR. Divergent mitochondrial biogenesis responses in human cardiomyopathy. Circulation 2013; 127:1957-67. [PMID: 23589024 DOI: 10.1161/circulationaha.112.001219] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Mitochondria are key players in the development and progression of heart failure (HF). Mitochondrial (mt) dysfunction leads to diminished energy production and increased cell death contributing to the progression of left ventricular failure. The fundamental mechanisms that underlie mt dysfunction in HF have not been fully elucidated. METHODS AND RESULTS To characterize mt morphology, biogenesis, and genomic integrity in human HF, we investigated left ventricular tissue from nonfailing hearts and end-stage ischemic (ICM) or dilated (DCM) cardiomyopathic hearts. Although mt dysfunction was present in both types of cardiomyopathy, mt were smaller and increased in number in DCM compared with ICM or nonfailing hearts. mt volume density and mtDNA copy number was increased by ≈2-fold (P<0.001) in DCM hearts in comparison with ICM hearts. These changes were accompanied by an increase in the expression of mtDNA-encoded genes in DCM versus no change in ICM. mtDNA repair and antioxidant genes were reduced in failing hearts, suggestive of a defective repair and protection system, which may account for the 4.1-fold increase in mtDNA deletion mutations in DCM (P<0.05 versus nonfailing hearts, P<0.05 versus ICM). CONCLUSIONS In DCM, mt dysfunction is associated with mtDNA damage and deletions, which could be a consequence of mutating stress coupled with a peroxisome proliferator-activated receptor γ coactivator 1α-dependent stimulus for mt biogenesis. However, this maladaptive compensatory response contributes to additional oxidative damage. Thus, our findings support further investigations into novel mechanisms and therapeutic strategies for mt dysfunction in DCM.
Collapse
Affiliation(s)
- Preeti Ahuja
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at UCLA, BH-569 CHS, BOX 957115, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Duicu O, Juşcă C, Falniţă L, Mirică S, Maximov D, Firă-Mladinescu O, Muntean D. Substrate-specific impairment of mitochondrial respiration in permeabilized fibers from patients with coronary heart disease versus valvular disease. Mol Cell Biochem 2013; 379:229-34. [PMID: 23568503 DOI: 10.1007/s11010-013-1644-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/28/2013] [Indexed: 12/15/2022]
Abstract
High-resolution respirometry of permeabilized myocardial fibers offers reliable insights concerning the integrated mitochondrial function while using small amounts of cardiac tissue. The aim of the present study was to assess the respiratory function in permeabilized fibers of human right atrial appendages harvested from patients with coronary heart disease (CHD) (n = 6) versus patients with valvular disease (n = 5) and preserved ejection fraction that underwent non-emergency cardiac surgery. Human bundle samples (1-3 mg wet weight) permeabilized with saponin were transferred into the 2 ml Oxygraph-2 k chambers to measure complex I(CI) and II (CII)-dependent respiration, respectively. The following values (expressed in pmol/s mg) were obtained for CI-dependent respiration: oxidative phosphorylation (OXPHOS), 35.65 ± 1.10 versus 42.43 ± 1.08, electron transport system (ETS), 37.87 ± 1.72 versus. 46.58 ± 1.85, and respiratory control ratio (RCR, calculated as the ratio between OXPHOS and LEAK states), 2.43 ± 0.09 versus 2.73 ± 0.068 (p < 0.05). In conclusion, in patients with CHD we showed a significant decline for the OXPHOS capacity, ETS and RCR for mitochondria energized with CI (but not with CII) substrates. These observations are suggestive for an early impairment of complex I supported respiration in ischemic heart disease, as previously demonstrated in the setting of experimental ischemia/reperfusion in several animal species.
Collapse
Affiliation(s)
- Oana Duicu
- Department of Pathophysiology, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | | | | | | | | | | | | |
Collapse
|
45
|
Collange O, Charles AL, Bouitbir J, Chenard MP, Zoll J, Diemunsch P, Thaveau F, Chakfé N, Piquard F, Geny B. Methylene blue protects liver oxidative capacity after gut ischaemia-reperfusion in the rat. Eur J Vasc Endovasc Surg 2012; 45:168-75. [PMID: 23246335 DOI: 10.1016/j.ejvs.2012.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 11/11/2012] [Indexed: 01/15/2023]
Abstract
OBJECTIVES Mesenteric ischaemia/reperfusion (IR) may lead to liver mitochondrial dysfunction and multiple organ failure. We determined whether gut IR induces early impairment of liver mitochondrial oxidative activity and whether methylene blue (MB) might afford protection. DESIGN Controlled animal study. MATERIALS AND METHODS Rats were randomised into three groups: controls (n = 18), gut IR group (mesenteric ischaemia (60 min)/reperfusion (60 min)) (n = 18) and gut IR + MB group (15 mg kg(-1) MB intra-peritoneally) (n = 16). Study parameters were: serum liver function markers, blood lactate, standard histology and DNA fragmentation (apoptosis) on intestinal and liver tissue, maximal oxidative capacity of liver mitochondria (state 3) and activity of complexes II, III and IV of the respiratory chain measured using a Clark oxygen electrode. RESULTS Gut IR increased lactate deshydrogenase (+982%), aspartate and alanine aminotransferases (+43% and +74%, respectively) and lactate levels (+271%). It induced segmental loss of intestinal villi and cryptic apoptosis. It reduced liver state 3 respiration by 30% from 50.1 ± 3 to 35.2 ± 3.5 μM O(2) min(-1) g(-1) (P < 0.01) and the activity of complexes II, III and IV of the mitochondrial respiratory chain. Early impairment of liver mitochondrial respiration was related to blood lactate levels (r(2) = 0.45). MB restored liver mitochondrial function. CONCLUSIONS MB protected against gut IR-induced liver mitochondria dysfunction.
Collapse
Affiliation(s)
- O Collange
- Pôle Anesthésie, Réanimation Chirurgicale, SAMU, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
An increasing body of clinical observations and experimental evidence suggests that cardiac dysfunction results from autonomic dysregulation of the contractile output of the heart. Excessive activation of the sympathetic nervous system and a decrease in parasympathetic tone are associated with increased mortality. Elevated levels of circulating catecholamines closely correlate with the severity and poor prognosis in heart failure. Sympathetic over-stimulation causes increased levels of catecholamines, which induce excessive aerobic metabolism leading to excessive cardiac oxygen consumption. Resulting impaired mitochondrial function causes acidosis, which results in reduction in blood flow by impairment of contractility. To the extent that the excessive aerobic metabolism resulting from adrenergic stimulation comes to a halt the energy deficit has to be compensated for by anaerobic metabolism. Glucose and glycogen become the essential nutrients. Beta-adrenergic blockade is used successfully to decrease hyperadrenergic drive. Neurohumoral antagonists block adrenergic over-stimulation but do not provide the heart with fuel for compensatory anaerobic metabolism. The endogenous hormone ouabain reduces catecholamine levels in healthy volunteers, promotes the secretion of insulin, induces release of acetylcholine from synaptosomes and potentiates the stimulation of glucose metabolism by insulin and acetylcholine. Ouabain stimulates glycogen synthesis and increases lactate utilisation by the myocardium. Decades of clinical experience with ouabain confirm the cardioprotective effects of this endogenous hormone. The so far neglected sympatholytic and vagotonic effects of ouabain on myocardial metabolism clearly make a clinical re-evaluation of this endogenous hormone necessary. Clinical studies with ouabain that correspond to current standards are warranted.
Collapse
|
47
|
Charles AL, Meyer A, Dal-Ros S, Auger C, Keller N, Ramamoorthy TG, Zoll J, Metzger D, Schini-Kerth V, Geny B. Polyphenols prevent ageing-related impairment in skeletal muscle mitochondrial function through decreased reactive oxygen species production. Exp Physiol 2012; 98:536-45. [DOI: 10.1113/expphysiol.2012.067496] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
48
|
Ruiz M, Courilleau D, Jullian JC, Fortin D, Ventura-Clapier R, Blondeau JP, Garnier A. A cardiac-specific robotized cellular assay identified families of human ligands as inducers of PGC-1α expression and mitochondrial biogenesis. PLoS One 2012; 7:e46753. [PMID: 23056435 PMCID: PMC3463514 DOI: 10.1371/journal.pone.0046753] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 09/05/2012] [Indexed: 12/12/2022] Open
Abstract
Background Mitochondrial function is dramatically altered in heart failure (HF). This is associated with a decrease in the expression of the transcriptional coactivator PGC-1α, which plays a key role in the coordination of energy metabolism. Identification of compounds able to activate PGC-1α transcription could be of future therapeutic significance. Methodology/Principal Findings We thus developed a robotized cellular assay to screen molecules in order to identify new activators of PGC-1α in a cardiac-like cell line. This screening assay was based on both the assessment of activity and gene expression of a secreted luciferase under the control of the human PGC-1α promoter, stably expressed in H9c2 cells. We screened part of a library of human endogenous ligands and steroid hormones, B vitamins and fatty acids were identified as activators of PGC-1α expression. The most responsive compounds of these families were then tested for PGC-1α gene expression in adult rat cardiomyocytes. These data highly confirmed the primary screening, and the increase in PGC-1α mRNA correlated with an increase in several downstream markers of mitochondrial biogenesis. Moreover, respiration rates of H9c2 cells treated with these compounds were increased evidencing their effectiveness on mitochondrial biogenesis. Conclusions/Significance Using our cellular reporter assay we could identify three original families, able to activate mitochondrial biogenesis both in cell line and adult cardiomyocytes. This first screening can be extended to chemical libraries in order to increase our knowledge on PGC-1α regulation in the heart and to identify potential therapeutic compounds able to improve mitochondrial function in HF.
Collapse
Affiliation(s)
- Matthieu Ruiz
- INSERM, U-769, Univ Paris-Sud, Châtenay-Malabry, France
| | | | - Jean-Christophe Jullian
- IFR141, CIBLOT platform, Univ Paris-Sud, Châtenay-Malabry, France
- BIOCIS, UMR 8076, Univ Paris-Sud, Châtenay-Malabry, France
| | | | | | | | - Anne Garnier
- INSERM, U-769, Univ Paris-Sud, Châtenay-Malabry, France
- * E-mail:
| |
Collapse
|
49
|
Aubert G, Vega RB, Kelly DP. Perturbations in the gene regulatory pathways controlling mitochondrial energy production in the failing heart. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:840-7. [PMID: 22964268 DOI: 10.1016/j.bbamcr.2012.08.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 08/21/2012] [Accepted: 08/24/2012] [Indexed: 12/15/2022]
Abstract
The heart is an omnivore organ that requires constant energy production to match its functional demands. In the adult heart, adenosine-5'-triphosphate (ATP) production occurs mainly through mitochondrial fatty acid and glucose oxidation. The heart must constantly adapt its energy production in response to changes in substrate supply and work demands across diverse physiologic and pathophysiologic conditions. The cardiac myocyte maintains a high level of mitochondrial ATP production through a complex transcriptional regulatory network that is orchestrated by the members of the peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family. There is increasing evidence that during the development of cardiac hypertrophy and in the failing heart, the activity of this network, including PGC-1, is altered. This review summarizes our current understanding of the perturbations in the gene regulatory pathways that occur during the development of heart failure. An appreciation of the role this regulatory circuitry serves in the regulation of cardiac energy metabolism may unveil novel therapeutic targets aimed at the metabolic disturbances that presage heart failure. This article is part of a Special Issue entitled:Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.
Collapse
Affiliation(s)
- Gregory Aubert
- Sanford-Burnham Medical Research Institute, Orlando, FL 32827, USA
| | | | | |
Collapse
|
50
|
Abstract
Studies in humans and in mice have highlighted the importance of short telomeres and impaired mitochondrial function in driving age-related functional decline in the heart. Although telomere and mitochondrial dysfunction have been viewed mainly in isolation, recent studies in telomerase-deficient mice have provided evidence for an intimate link between these two processes. Telomere dysfunction induces a profound p53-dependent repression of the master regulators of mitochondrial biogenesis and function, peroxisome proliferator-activated receptor gamma coactivator (PGC)-1α and PGC-1β in the heart, which leads to bioenergetic compromise due to impaired oxidative phosphorylation and ATP generation. This telomere-p53-PGC mitochondrial/metabolic axis integrates many factors linked to heart aging including increased DNA damage, p53 activation, mitochondrial, and metabolic dysfunction and provides a molecular basis of how dysfunctional telomeres can compromise cardiomyocytes and stem cell compartments in the heart to precipitate cardiac aging.
Collapse
Affiliation(s)
- Javid Moslehi
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | | |
Collapse
|