1
|
Iboleon-Jimenez A, Sánchez-Quintero MJ, Carmona-Segovia ADM, Sojo B, Fernández-Ramos AM, García-Rodríguez L, Molina-Ramos AI, García-Pinilla JM, Jimenez-Navarro M, Ortega-Gomez A. Circulating mitochondrial biomarkers in acute coronary syndrome. Front Med (Lausanne) 2025; 12:1568305. [PMID: 40443515 PMCID: PMC12119305 DOI: 10.3389/fmed.2025.1568305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/29/2025] [Indexed: 06/02/2025] Open
Abstract
Background Acute coronary syndrome (ACS) is the leading cause of mortality in developed countries. Mitochondrial dysfunction is a hallmark of various cardiometabolic diseases, including ACS. Emerging evidence suggests that evaluating mitochondrial biomarkers in plasma may offer valuable insights into the pathophysiology and management of these conditions. The present study aims to analyse the effect of ACS, sex and their interaction on plasma levels of mitochondrial markers, such as peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), mitochondrial open reading frame of the 12S rRNA type-c (MOTS-c) and citrate syntetase (CS). Methods A total of 18 ACS patients (8 women and 10 men) and 20 controls (8 women and 12 men) were included in this study. Venous blood samples were collected from participants after a 12-h overnight fast. Plasma levels of mitochondrial PGC-1α, MOTS-c and CS were measured. Results ACS significantly reduced plasma levels of PGC-1α and MOTS-c. Sex did not shown a significant effect on these markers. Additionally, MOTS-c positively correlated with the first troponin and hemoglobin, PGC-1α negatively correlated with glucose and positively with HDL-cholesterol, and CS showed negative correlations with NT-proBNP, C-reactive protein, and hemoglobin. Conclusion Mitochondria markers, MOTS-c and PGC-1α, are altered in ACS patients, with no observed sex differences. These findings represent an initial step toward integrating personalized medicine into the clinical management of ACS. Nonetheless, further studies are required to fully elucidate the role of these markers in this pathology.
Collapse
Affiliation(s)
- Andrea Iboleon-Jimenez
- Unidad Docente Multiprofesional de Atención Familiar y Comunitaria, Distrito de Atención Primaria Málaga-Guadalhorce; Faculty of Medicine, University of Málaga, Málaga, Spain
| | - María J. Sánchez-Quintero
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ada D. M. Carmona-Segovia
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Bélen Sojo
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Endocrinology and Nutrition UGC, Victoria Virgen University Hospital, Málaga, Spain
| | - Ana María Fernández-Ramos
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Clinical Analysis UGC, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Luis García-Rodríguez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
| | - Ana I. Molina-Ramos
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - José Manuel García-Pinilla
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Málaga, Spain
| | - Manuel Jimenez-Navarro
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Málaga, Spain
| | - Almudena Ortega-Gomez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Endocrinology and Nutrition UGC, Victoria Virgen University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Odendaal‐Gambrell CP, O'Brien C, Cairns M, Maarman GJ, Joseph DE, Smith C, Rautenbach F, Marnewick JL, Essop MF. Chronic stress elicits sex-specific mitochondrial respiratory functional changes in the rat heart. Physiol Rep 2025; 13:e70371. [PMID: 40356314 PMCID: PMC12069860 DOI: 10.14814/phy2.70371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/11/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Although chronic psychosocial stress is linked to cardiovascular diseases, the underlying mechanisms remain elusive. For this study, we focused on the mitochondrion as a putative mediator of stress-related cardiac pathologies in a sex-dependent manner. Male and female Wistar rats were subjected to chronic stress for 4 weeks (mimicking an anxious phenotype) versus matched controls. Cardiac redox status, mitochondrial respiration parameters, and expression levels of proteins involved in mitochondrial oxidative phosphorylation, dynamics, and biogenesis were evaluated. Despite limited changes in behavior and circulating stress hormones (both sexes), stressed males exhibited altered cardiac oxidative phosphorylation via β-oxidation- and glucose oxidation-linked respiratory pathways together with increased myocardial antioxidant capacity and decreased lipid peroxidation. Conversely, stressed females exhibited a protective and resilient phenotype by displaying augmented levels of major mitochondrial respiratory complexes (complex I, III, and ATP synthase) and a fusion marker (mitofusin-2 [Mfn2]), together with attenuated expression of a fission marker (dynamin-related protein-1 [Drp1]) despite decreased estradiol levels. In contrast, stressed males displayed increased cardiac ATP synthase levels together with diminished peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α) expression versus controls. These findings indicate that male mitochondria are more prone to stress-related functional changes, while females exhibited a more protective and resilient phenotype.
Collapse
Affiliation(s)
- Caitlin P. Odendaal‐Gambrell
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Cassidy O'Brien
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Megan Cairns
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Gerald J. Maarman
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Danzil E. Joseph
- Centre for Cardiometabolic Research in Africa (CARMA), Department of Physiological Sciences, Faculty of ScienceStellenbosch UniversityStellenboschSouth Africa
| | - Carine Smith
- Experimental Medicine Unit, Department of Medicine, Faculty of Medicine and Health SciencesStellenbosch UniversityStellenboschSouth Africa
| | - Fanie Rautenbach
- Applied Microbial and Health Biotechnology InstituteCape Peninsula University of TechnologyCape TownSouth Africa
| | - Jeanine L. Marnewick
- Applied Microbial and Health Biotechnology InstituteCape Peninsula University of TechnologyCape TownSouth Africa
| | - M. Faadiel Essop
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Biomedical Research Institute, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| |
Collapse
|
3
|
Fan Q, Wang Y, An Q, Ling Y. Right ventricular dysfunction following surgical repair of tetralogy of Fallot: Molecular pathways and therapeutic prospects. Biomed Pharmacother 2025; 184:117924. [PMID: 39983432 DOI: 10.1016/j.biopha.2025.117924] [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: 01/03/2025] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025] Open
Abstract
Tetralogy of Fallot (TOF) is the most common cyanotic congenital heart disease (CHD). Although surgical correction of TOF is possible, patients often face challenges related to right ventricle dysfunction (RVD) post-surgery, which can significantly impact their long-term survival. The causes of RVD in TOF patients are complex, involving both the unique structural characteristics of the TOF heart and damage resulting from surgical interventions. Residual anatomical issues following TOF repair are often unavoidable, placing the RV under stress and leading to the activation of multiple molecular pathways. This review comprehensively outlines the causes of RVD in patients after TOF surgery, particularly focusing the molecular pathways that contribute to RVD, including established signaling pathways as well as emerging pathways identified through transcriptomic analysis of RV myocardium in TOF patients. We also highlight the features of these molecular pathways concerning RVD, as well as the influence of gender disparities on these molecular pathways. By interpreting the causes and molecular mechanisms underlying RVD after TOF surgery, this review provides new insights for managing RVD in repaired TOF, potentially paving the way for targeted therapies aimed at improving long-term outcomes for those affected by RVD.
Collapse
Affiliation(s)
- Qiang Fan
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yabo Wang
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| | - Yunfei Ling
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
4
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
5
|
Paraskevaidis I, Kourek C, Farmakis D, Tsougos E. Heart Failure: A Deficiency of Energy-A Path Yet to Discover and Walk. Biomedicines 2024; 12:2589. [PMID: 39595155 PMCID: PMC11592498 DOI: 10.3390/biomedicines12112589] [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: 10/15/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Heart failure is a complex syndrome and our understanding and therapeutic approach relies mostly on its phenotypic presentation. Notably, the heart is characterized as the most energy-consuming organ, being both a producer and consumer, in order to satisfy multiple cardiac functions: ion exchange, electromechanical coordination, excitation-contraction coupling, etc. By obtaining further knowledge of the cardiac energy field, we can probably better characterize the basic pathophysiological events occurring in heart disease patients and understand the metabolic substance changes, the relationship between the alteration of energy production/consumption, and hence energetic deficiency not only in the heart as a whole but in every single cardiac territory, which will hopefully provide us with the opportunity to uncover the beginning of the heart failure process. In this respect, using (a) newer imaging techniques, (b) biomedicine, (c) nanotechnology, and (d) artificial intelligence, we can gain a deeper understanding of this complex syndrome. This, in turn, can lead to earlier and more effective therapeutic approaches, ultimately improving human health. To date, the scientific community has not given sufficient attention to the energetic starvation model. In our view, this review aims to encourage scientists and the medical community to conduct studies for a better understanding and treatment of this syndrome.
Collapse
Affiliation(s)
- Ioannis Paraskevaidis
- 6th Department of Cardiology, Hygeia Hospital, 151 23 Athens, Greece; (I.P.); (E.T.)
| | - Christos Kourek
- Department of Cardiology, 417 Army Share Fund Hospital of Athens (NIMTS), 115 21 Athens, Greece;
| | - Dimitrios Farmakis
- Heart Failure Unit, Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistiran University of Athens, 124 62 Athens, Greece
| | - Elias Tsougos
- 6th Department of Cardiology, Hygeia Hospital, 151 23 Athens, Greece; (I.P.); (E.T.)
| |
Collapse
|
6
|
Zhou R, Barnes K, Gibson S, Fillmore N. Dual-edged role of SIRT1 in energy metabolism and cardiovascular disease. Am J Physiol Heart Circ Physiol 2024; 327:H1162-H1173. [PMID: 39269450 DOI: 10.1152/ajpheart.00001.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 08/13/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Regulation of energy metabolism is pivotal in the development of cardiovascular diseases. Dysregulation in mitochondrial fatty acid oxidation has been linked to cardiac lipid accumulation and diabetic cardiomyopathy. Sirtuin 1 (SIRT1) is a deacetylase that regulates the acetylation of various proteins involved in mitochondrial energy metabolism. SIRT1 mediates energy metabolism by directly and indirectly affecting multiple aspects of mitochondrial processes, such as mitochondrial biogenesis. SIRT1 interacts with essential mitochondrial energy regulators such as peroxisome proliferator-activated receptor-α (PPARα), PPARγ coactivator-1α, estrogen-related receptor-α, and their downstream targets. Apart from that, SIRT1 regulates additional proteins, including forkhead box protein O1 and AMP-activated protein kinase in cardiac disease. Interestingly, studies have also shown that the expression of SIRT1 plays a dual-edged role in energy metabolism. Depending on the physiological state, SIRT1 expression can be detrimental or protective. This review focuses on the molecular pathways through which SIRT1 regulates energy metabolism in cardiovascular diseases. We will review SIRT1 and discuss its role in cardiac energy metabolism and its benefits and detrimental effects in heart disease.
Collapse
Affiliation(s)
- Redemptor Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Kaleb Barnes
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Savannah Gibson
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Natasha Fillmore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| |
Collapse
|
7
|
Bi F, Gao C, Guo H. Epigenetic regulation of cardiovascular diseases induced by behavioral and environmental risk factors: Mechanistic, diagnostic, and therapeutic insights. FASEB Bioadv 2024; 6:477-502. [PMID: 39512842 PMCID: PMC11539034 DOI: 10.1096/fba.2024-00080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024] Open
Abstract
Behavioral and environmental risk factors are critical in the development and progression of cardiovascular disease (CVD). Understanding the molecular mechanisms underlying these risk factors will offer valuable insights for targeted preventive and therapeutic strategies. Epigenetic modifications, including DNA methylation, histone modifications, chromatin remodeling, noncoding RNA (ncRNA) expression, and epitranscriptomic modifications, have emerged as key mediators connecting behavioral and environmental risk factors to CVD risk and progression. These epigenetic alterations can profoundly impact on cardiovascular health and susceptibility to CVD by influencing cellular processes, development, and disease risk over an individual's lifetime and potentially across generations. This review examines how behavioral and environmental risk factors affect CVD risk and health outcomes through epigenetic regulation. We review the epigenetic effects of major behavioral risk factors (such as smoking, alcohol consumption, physical inactivity, unhealthy diet, and obesity) and environmental risk factors (including air and noise pollution) in the context of CVD pathogenesis. Additionally, we explore epigenetic biomarkers, considering their role as causal or surrogate indicators, and discuss epigenetic therapeutics targeting the mechanisms through which these risk factors contribute to CVD. We also address future research directions and challenges in leveraging epigenetic insights to reduce the burden of CVD related to behavioral and environmental factors and improve public health outcomes. This review aims to provide a comprehensive understanding of behavioral and environmental epigenetics in CVD and offer valuable strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Feifei Bi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake CityUtahUSA
- Division of Cardiothoracic Surgery, Department of SurgerySchool of Medicine, University of UtahSalt Lake CityUtahUSA
| | - Chen Gao
- Department of Pharmacology and Systems PhysiologyUniversity of CincinnatiCincinnatiOhioUSA
| | - Hongchao Guo
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake CityUtahUSA
- Division of Cardiothoracic Surgery, Department of SurgerySchool of Medicine, University of UtahSalt Lake CityUtahUSA
| |
Collapse
|
8
|
Salis Torres A, Lee JE, Caporali A, Semple RK, Horrocks MH, MacRae VE. Mitochondrial Dysfunction as a Potential Mechanism Mediating Cardiac Comorbidities in Parkinson's Disease. Int J Mol Sci 2024; 25:10973. [PMID: 39456761 PMCID: PMC11507255 DOI: 10.3390/ijms252010973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Individuals diagnosed with Parkinson's disease (PD) often exhibit heightened susceptibility to cardiac dysfunction, reflecting a complex interaction between these conditions. The involvement of mitochondrial dysfunction in the development and progression of cardiac dysfunction and PD suggests a plausible commonality in some aspects of their molecular pathogenesis, potentially contributing to the prevalence of cardiac issues in PD. Mitochondria, crucial organelles responsible for energy production and cellular regulation, play important roles in tissues with high energetic demands, such as neurons and cardiac cells. Mitochondrial dysfunction can occur in different and non-mutually exclusive ways; however, some mechanisms include alterations in mitochondrial dynamics, compromised bioenergetics, biogenesis deficits, oxidative stress, impaired mitophagy, and disrupted calcium balance. It is plausible that these factors contribute to the increased prevalence of cardiac dysfunction in PD, suggesting mitochondrial health as a potential target for therapeutic intervention. This review provides an overview of the physiological mechanisms underlying mitochondrial quality control systems. It summarises the diverse roles of mitochondria in brain and heart function, highlighting shared pathways potentially exhibiting dysfunction and driving cardiac comorbidities in PD. By highlighting strategies to mitigate dysfunction associated with mitochondrial impairment in cardiac and neural tissues, our review aims to provide new perspectives on therapeutic approaches.
Collapse
Affiliation(s)
- Agustina Salis Torres
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
| | - Ji-Eun Lee
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- IRR Chemistry Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Andrea Caporali
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Robert K. Semple
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Mathew H. Horrocks
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Vicky E. MacRae
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
| |
Collapse
|
9
|
Xu Z, Li M, Lyu D, Xiao H, Li S, Li Z, Li M, Xiao J, Huang H. Cinnamaldehyde activates AMPK/PGC-1α pathway via targeting GRK2 to ameliorate heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155894. [PMID: 39089090 DOI: 10.1016/j.phymed.2024.155894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/25/2024] [Accepted: 07/15/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND According to recent research, treating heart failure (HF) by inhibiting G protein-coupled receptor kinase 2 (GRK2) to improve myocardial energy metabolism has been identified as a potential approach. Cinnamaldehyde (CIN), a phenylpropyl aldehyde compound, has been demonstrated to exhibit beneficial effects in cardiovascular diseases. However, whether CIN inhibits GRK2 to ameliorate myocardial energy metabolism in HF is still unclear. PURPOSE This study examines the effects of CIN on GRK2 and myocardial energy metabolism to elucidate its underlying mechanism to treat HF. METHODS The isoproterenol (ISO) induced HF model in vivo and in vitro were constructed using Sprague-Dawley (SD) rats and primary neonatal rat cardiomyocytes (NRCMs). Based on this, the effects of CIN on myocardial energy metabolism and GRK2 were investigated. Additionally, validation experiments were conducted after interfering and over-expressing GRK2 in ISO-induced NRCMs to verify the regulatory effect of CIN on GRK2. Furthermore, binding capacity between GRK2 and CIN was explored by Cellular Thermal Shift Assay (CETSA) and Microscale Thermophoresis (MST). RESULTS In vivo and in vitro, CIN significantly improved HF as demonstrated by reversing abnormal changes in myocardial injury markers, inhibiting myocardial hypertrophy and decreasing myocardial fibrosis. Additionally, CIN promoted myocardial fatty acid metabolism to ameliorate myocardial energy metabolism disorder by activating AMPK/PGC-1α signaling pathway. Moreover, CIN reversed the inhibition of myocardial fatty acid metabolism and AMPK/PGC-1α signaling pathway by GRK2 over-expression in ISO-induced NRCMs. Meanwhile, CIN had no better impact on the stimulation of cardiac fatty acid metabolism and the AMPK/PGC-1α signaling pathway in ISO-induced NRCMs when GRK2 was disrupted. Noticeably, CETSA and MST confirmed that CIN binds to GRK2 directly. The binding of CIN and GRK2 promoted the ubiquitination degradation of GRK2 mediated by murine double mimute 2. CONCLUSION This study demonstrates that CIN exerts a protective intervention in HF by targeting GRK2 and promoting its ubiquitination degradation to activate AMPK/PGC-1α signaling pathway, ultimately improving myocardial fatty acid metabolism.
Collapse
Affiliation(s)
- Zhanchi Xu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou 510801, China
| | - Minghui Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Dongxin Lyu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Haiming Xiao
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shanshan Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhuoming Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Min Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Junhui Xiao
- Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou 510801, China.
| | - Heqing Huang
- Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou 510801, China.
| |
Collapse
|
10
|
Li A, Qin Y, Gong G. The Changes of Mitochondria during Aging and Regeneration. Adv Biol (Weinh) 2024; 8:e2300445. [PMID: 38979843 DOI: 10.1002/adbi.202300445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/30/2024] [Indexed: 07/10/2024]
Abstract
Aging and regeneration are opposite cellular processes. Aging refers to progressive dysfunction in most cells and tissues, and regeneration refers to the replacement of damaged or dysfunctional cells or tissues with existing adult or somatic stem cells. Various studies have shown that aging is accompanied by decreased regenerative abilities, indicating a link between them. The performance of any cellular process needs to be supported by the energy that is majorly produced by mitochondria. Thus, mitochondria may be a link between aging and regeneration. It should be interesting to discuss how mitochondria behave during aging and regeneration. The changes of mitochondria in aging and regeneration discussed in this review can provide a timely and necessary study of the causal roles of mitochondrial homeostasis in longevity and health.
Collapse
Affiliation(s)
- Anqi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuan Qin
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Guohua Gong
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
11
|
Golubnitschaja O, Kapinova A, Sargheini N, Bojkova B, Kapalla M, Heinrich L, Gkika E, Kubatka P. Mini-encyclopedia of mitochondria-relevant nutraceuticals protecting health in primary and secondary care-clinically relevant 3PM innovation. EPMA J 2024; 15:163-205. [PMID: 38841620 PMCID: PMC11148002 DOI: 10.1007/s13167-024-00358-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 06/07/2024]
Abstract
Despite their subordination in humans, to a great extent, mitochondria maintain their independent status but tightly cooperate with the "host" on protecting the joint life quality and minimizing health risks. Under oxidative stress conditions, healthy mitochondria promptly increase mitophagy level to remove damaged "fellows" rejuvenating the mitochondrial population and sending fragments of mtDNA as SOS signals to all systems in the human body. As long as metabolic pathways are under systemic control and well-concerted together, adaptive mechanisms become triggered increasing systemic protection, activating antioxidant defense and repair machinery. Contextually, all attributes of mitochondrial patho-/physiology are instrumental for predictive medical approach and cost-effective treatments tailored to individualized patient profiles in primary (to protect vulnerable individuals again the health-to-disease transition) and secondary (to protect affected individuals again disease progression) care. Nutraceuticals are naturally occurring bioactive compounds demonstrating health-promoting, illness-preventing, and other health-related benefits. Keeping in mind health-promoting properties of nutraceuticals along with their great therapeutic potential and safety profile, there is a permanently growing demand on the application of mitochondria-relevant nutraceuticals. Application of nutraceuticals is beneficial only if meeting needs at individual level. Therefore, health risk assessment and creation of individualized patient profiles are of pivotal importance followed by adapted nutraceutical sets meeting individual needs. Based on the scientific evidence available for mitochondria-relevant nutraceuticals, this article presents examples of frequent medical conditions, which require protective measures targeted on mitochondria as a holistic approach following advanced concepts of predictive, preventive, and personalized medicine (PPPM/3PM) in primary and secondary care.
Collapse
Affiliation(s)
- Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Andrea Kapinova
- Biomedical Centre Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Nafiseh Sargheini
- Max Planck Institute for Plant Breeding Research, Carl-Von-Linne-Weg 10, 50829 Cologne, Germany
| | - Bianka Bojkova
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, P. J. Šafárik University in Košice, 040 01 Košice, Slovakia
| | - Marko Kapalla
- Negentropic Systems, Ružomberok, Slovakia
- PPPM Centre, s.r.o., Ruzomberok, Slovakia
| | - Luisa Heinrich
- Institute of General Medicine, University of Leipzig, Leipzig, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Peter Kubatka
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| |
Collapse
|
12
|
Park YJ, Kim HJ, Koh DJ, Kim E, Lim YW, An HJ. Effect and mechanisms of Gambi-jung against high-fat diet-induced cardiac apoptosis in mice. Heliyon 2024; 10:e29161. [PMID: 38644871 PMCID: PMC11031771 DOI: 10.1016/j.heliyon.2024.e29161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/23/2024] Open
Abstract
Obesity is associated with an increased risk of cardiovascular disease. Gambi-jung (GBJ), a modified herbal formula of Taeumjowi-tang, induces weight loss in high-fat diet (HFD)-fed obese mice. Meanwhile, concerns have been raised regarding Ephedra sinica Stapf (ES), the primary herb of GBJ, having potential adverse cardiovascular effects. However, there have been no reports on the effects of ES and ephedrine-containing products on obesity-induced cardiac apoptosis. Therefore, to investigated the effect of GBJ and ES on HFD-induced cardiac apoptosis, we utilized Western blot analysis, TUNEL-staining, and histological staining of heart tissues from HFD-fed obese mice. Western blot analysis showed that there were significant changes in the protein levels of anti-apoptotic markers (B-cell lymphoma (BCL) protein 2 (BCL-2), BCL-XL, and X-linked inhibitor of apoptosis protein) and pro-apoptotic markers (Fas, Fas-associated protein with death domain, BCL-2 agonist of cell death, BCL-2 associated X, cytochrome C, and cleaved caspase-9) in the heart of HFD-fed mice. In contrast administration of 250 mg/kg GBJ for 12 weeks significantly reversed the protein levels related to the apoptosis signaling pathway, which was greater than that of ES administration. Furthermore, GBJ-treated mice had markedly decreased number of TUNEL-stained apoptotic cells compared to the HFD group. Moreover, GBJ improved the mitochondrial function by regulating the genes expression of uncoupling protein 2, peroxisome proliferator-activated receptor-γ coactivator-1α, optic atrophy protein 1, and fission protein 1. Notably, hematoxylin and eosin histological staining showed no changes in the heart tissues of GBJ- and ES-treated mice, indicating that long-term administration of GBJ and ES did not exert any adverse effects on the cardiac tissue. The present study lays the foundation to support the efficacy of GBJ in protecting cardiac cell apoptosis induced by HFD feeding, as well as to verify the cardiac safety of GBJ administration.
Collapse
Affiliation(s)
- Yea-Jin Park
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, 26339, Republic of Korea
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyo-Jung Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Duck-Jae Koh
- Nubebe Korean Medical Clinic Jamsil Center, Seoul, 05510, Republic of Korea
| | - Eunjoo Kim
- Nubebe Obesity Research Institute, Seoul, 06634, Republic of Korea
- Nubebe Korean Medical Clinic Bundang Center, Seongnam-si, 13506, Republic of Korea
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Young-Woo Lim
- Nubebe Obesity Research Institute, Seoul, 06634, Republic of Korea
- Nubebe Korean Medical Clinic Bundang Center, Seongnam-si, 13506, Republic of Korea
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
13
|
Zhu Y, Wang F, Ma Z, Hou S, Deng W, Zhang Y, Wu Q. Anti-proliferation and apoptosis induced via the mTOR/PGC-1α signaling pathway in trophoblast cells of miscarriage. Exp Cell Res 2024; 436:113959. [PMID: 38395376 DOI: 10.1016/j.yexcr.2024.113959] [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: 11/10/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Miscarriage is a common complication during early pregnancy and affects approximately 10%-15% of all pregnant women. Several studies have reported that the abnormal expression of mitochondrial oxidative stress-related genes might be involved in the occurrence and progression of miscarriage. The present study attempted to uncover the role of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) in miscarriage chorionic villous tissue. The hypothesis that PGC-1α is crucial for glycolysis and oxidative phosphorylation during early pregnancy was tested. The results showed that the mRNA and protein levels of PGC-1α were significantly increased in the miscarriage chorionic villous tissues compared with the artificial selective abortion group, and that the expression was regulated by mTOR in knockdown and overexpression of mTOR in HTR8 cell lines. PGC-1α also promoted mitochondrion oxidative phosphorylation but inhibited glycolysis process. In addition, PGC-1α could drive ROS production, reduce mitochondrial membrane potential and block NADPH synthesis, resulting in cell cycle arrest and cell apoptosis, eventually leading to miscarriage. These results suggested that the aberrant expression of PGC-1α is involved in the etiology of early miscarriage, providing new perspectives regarding the mechanisms of miscarriage and a potential therapeutic target for miscarriage.
Collapse
Affiliation(s)
- Yuanchang Zhu
- Fertility Center, Shenzhen Hengsheng Hospital, Shenzhen, China.
| | - Feng Wang
- Fertility Center, Shenzhen Hengsheng Hospital, Shenzhen, China
| | - Zhuanghong Ma
- Fertility Center, Shenzhen Hengsheng Hospital, Shenzhen, China
| | - Sumei Hou
- Fertility Center, Shenzhen Hengsheng Hospital, Shenzhen, China
| | - Weifen Deng
- Fertility Center, Shenzhen Hengsheng Hospital, Shenzhen, China
| | - Yaou Zhang
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Qiongfang Wu
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, China.
| |
Collapse
|
14
|
Wen J, Chen C. From Energy Metabolic Change to Precision Therapy: a Holistic View of Energy Metabolism in Heart Failure. J Cardiovasc Transl Res 2024; 17:56-70. [PMID: 37450209 DOI: 10.1007/s12265-023-10412-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Heart failure (HF) is a complex and multifactorial disease that affects millions of people worldwide. It is characterized by metabolic disturbances of substrates such as glucose, fatty acids (FAs), ketone bodies, and amino acids, which lead to changes in cardiac energy metabolism pathways. These metabolic alterations can directly or indirectly promote myocardial remodeling, thereby accelerating the progression of HF, resulting in a vicious cycle of worsening symptoms, and contributing to the increased hospitalization and mortality among patients with HF. In this review, we summarized the latest researches on energy metabolic profiling in HF and provided the related translational therapeutic strategies for this devastating disease. By taking a holistic approach to understanding energy metabolism changes in HF, we hope to provide comprehensive insights into the pathophysiology of this challenging condition and identify novel precise targets for the development of more effective treatments.
Collapse
Affiliation(s)
- Jianpei Wen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| |
Collapse
|
15
|
Dörmann N, Hammer E, Struckmann K, Rüdebusch J, Bartels K, Wenzel K, Schulz J, Gross S, Schwanz S, Martin E, Fielitz B, Pablo Tortola C, Hahn A, Benkner A, Völker U, Felix SB, Fielitz J. Metabolic remodeling in cardiac hypertrophy and heart failure with reduced ejection fraction occurs independent of transcription factor EB in mice. Front Cardiovasc Med 2024; 10:1323760. [PMID: 38259303 PMCID: PMC10800928 DOI: 10.3389/fcvm.2023.1323760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Background A metabolic shift from fatty acid (FAO) to glucose oxidation (GO) occurs during cardiac hypertrophy (LVH) and heart failure with reduced ejection fraction (HFrEF), which is mediated by PGC-1α and PPARα. While the transcription factor EB (TFEB) regulates the expression of both PPARGC1A/PGC-1α and PPARA/PPARα, its contribution to metabolic remodeling is uncertain. Methods Luciferase assays were performed to verify that TFEB regulates PPARGC1A expression. Cardiomyocyte-specific Tfeb knockout (cKO) and wildtype (WT) male mice were subjected to 27G transverse aortic constriction or sham surgery for 21 and 56 days, respectively, to induce LVH and HFrEF. Echocardiographic, morphological, and histological analyses were performed. Changes in markers of cardiac stress and remodeling, metabolic shift and oxidative phosphorylation were investigated by Western blot analyses, mass spectrometry, qRT-PCR, and citrate synthase and complex II activity measurements. Results Luciferase assays revealed that TFEB increases PPARGC1A/PGC-1α expression, which was inhibited by class IIa histone deacetylases and derepressed by protein kinase D. At baseline, cKO mice exhibited a reduced cardiac function, elevated stress markers and a decrease in FAO and GO gene expression compared to WT mice. LVH resulted in increased cardiac remodeling and a decreased expression of FAO and GO genes, but a comparable decline in cardiac function in cKO compared to WT mice. In HFrEF, cKO mice showed an improved cardiac function, lower heart weights, smaller myocytes and a reduction in cardiac remodeling compared to WT mice. Proteomic analysis revealed a comparable decrease in FAO- and increase in GO-related proteins in both genotypes. A significant reduction in mitochondrial quality control genes and a decreased citrate synthase and complex II activities was observed in hearts of WT but not cKO HFrEF mice. Conclusions TFEB affects the baseline expression of metabolic and mitochondrial quality control genes in the heart, but has only minor effects on the metabolic shift in LVH and HFrEF in mice. Deletion of TFEB plays a protective role in HFrEF but does not affect the course of LVH. Further studies are needed to elucidate if TFEB affects the metabolic flux in stressed cardiomyocytes.
Collapse
Affiliation(s)
- Niklas Dörmann
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Elke Hammer
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Karlotta Struckmann
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Julia Rüdebusch
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Kirsten Bartels
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Kristin Wenzel
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Julia Schulz
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Stefan Gross
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Stefan Schwanz
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Elisa Martin
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Britta Fielitz
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Cristina Pablo Tortola
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Hahn
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Benkner
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stephan B. Felix
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| | - Jens Fielitz
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
16
|
Walker MA, Chen H, Yadav A, Ritterhoff J, Villet O, McMillen T, Wang Y, Purcell H, Djukovic D, Raftery D, Isoherranen N, Tian R. Raising NAD + Level Stimulates Short-Chain Dehydrogenase/Reductase Proteins to Alleviate Heart Failure Independent of Mitochondrial Protein Deacetylation. Circulation 2023; 148:2038-2057. [PMID: 37965787 PMCID: PMC10842390 DOI: 10.1161/circulationaha.123.066039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/04/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Strategies to increase cellular NAD+ (oxidized nicotinamide adenine dinucleotide) level have prevented cardiac dysfunction in multiple models of heart failure, but molecular mechanisms remain unclear. Little is known about the benefits of NAD+-based therapies in failing hearts after the symptoms of heart failure have appeared. Most pretreatment regimens suggested mechanisms involving activation of sirtuin, especially Sirt3 (sirtuin 3), and mitochondrial protein acetylation. METHODS We induced cardiac dysfunction by pressure overload in SIRT3-deficient (knockout) mice and compared their response with nicotinamide riboside chloride treatment with wild-type mice. To model a therapeutic approach, we initiated the treatment in mice with established cardiac dysfunction. RESULTS We found nicotinamide riboside chloride improved mitochondrial function and blunted heart failure progression. Similar benefits were observed in wild-type and knockout mice. Boosting NAD+ level improved the function of NAD(H) redox-sensitive SDR (short-chain dehydrogenase/reductase) family proteins. Upregulation of Mrpp2 (mitochondrial ribonuclease P protein 2), a multifunctional SDR protein and a subunit of mitochondrial ribonuclease P, improves mitochondrial DNA transcripts processing and electron transport chain function. Activation of SDRs in the retinol metabolism pathway stimulates RXRα (retinoid X receptor α)/PPARα (proliferator-activated receptor α) signaling and restores mitochondrial oxidative metabolism. Downregulation of Mrpp2 and impaired mitochondrial ribonuclease P were found in human failing hearts, suggesting a shared mechanism of defective mitochondrial biogenesis in mouse and human heart failure. CONCLUSIONS These findings identify SDR proteins as important regulators of mitochondrial function and molecular targets of NAD+-based therapy. Furthermore, the benefit is observed regardless of Sirt3-mediated mitochondrial protein deacetylation, a widely held mechanism for NAD+-based therapy for heart failure. The data also show that NAD+-based therapy can be useful in pre-existing heart failure.
Collapse
Affiliation(s)
- Matthew A. Walker
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Hongye Chen
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Aprajita Yadav
- Department of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, WA 98195
| | - Julia Ritterhoff
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Outi Villet
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Tim McMillen
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Yuliang Wang
- Department of Computer Science & Engineering,
University of Washington, Seattle, WA 98195
| | - Hayley Purcell
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Danijel Djukovic
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Daniel Raftery
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, WA 98195
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| |
Collapse
|
17
|
Packer M. Qiliqiangxin: A multifaceted holistic treatment for heart failure or a pharmacological probe for the identification of cardioprotective mechanisms? Eur J Heart Fail 2023; 25:2130-2143. [PMID: 37877337 DOI: 10.1002/ejhf.3068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 10/26/2023] Open
Abstract
The active ingredients in many traditional Chinese medicines are isoprene oligomers with a diterpenoid or triterpenoid structure, which exert cardiovascular effects by signalling through nutrient surplus and nutrient deprivation pathways. Qiliqiangxin (QLQX) is a commercial formulation of 11 different plant ingredients, whose active compounds include astragaloside IV, tanshione IIA, ginsenosides (Rb1, Rg1 and Re) and periplocymarin. In the QUEST trial, QLQX reduced the combined risk of cardiovascular death or heart failure hospitalization (hazard ratio 0.78, 95% confidence interval 0.68-0.90), based on 859 events in 3119 patients over a median of 18.2 months; the benefits were seen in patients taking foundational drugs except for sodium-glucose cotransporter 2 (SGLT2) inhibitors. Numerous experimental studies of QLQX in diverse cardiac injuries have yielded highly consistent findings. In marked abrupt cardiac injury, QLQX mitigated cardiac injury by upregulating nutrient surplus signalling through the PI3K/Akt/mTOR/HIF-1α/NRF2 pathway; the benefits of QLQX were abrogated by suppression of PI3K, Akt, mTOR, HIF-1α or NRF2. In contrast, in prolonged measured cardiac stress (as in chronic heart failure), QLQX ameliorated oxidative stress, maladaptive hypertrophy, cardiomyocyte apoptosis, and proinflammatory and profibrotic pathways, while enhancing mitochondrial health and promoting glucose and fatty acid oxidation and ATP production. These effects are achieved by an action of QLQX to upregulate nutrient deprivation signalling through SIRT1/AMPK/PGC-1α and enhanced autophagic flux. In particular, QLQX appears to enhance the interaction of PGC-1α with PPARα, possibly by direct binding to RXRα; silencing of SIRT1, PGC-1α and RXRα abrogated the favourable effects of QLQX in the heart. Since PGC-1α/RXRα is also a downstream effector of Akt/mTOR signalling, the actions of QLQX on PGC-1α/RXRα may explain its favourable effects in both acute and chronic stress. Intriguingly, the individual ingredients in QLQX - astragaloside IV, ginsenosides, and tanshione IIA - share QLQX's effects on PGC-1α/RXRα/PPARα signalling. QXQL also contains periplocymarin, a cardiac glycoside that inhibits Na+ -K+ -ATPase. Taken collectively, these observations support a conceptual framework for understanding the mechanism of action for QLQX in heart failure. The high likelihood of overlap in the mechanism of action of QLQX and SGLT2 inhibitors requires additional experimental studies and clinical trials.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Dallas, TX, USA
- Imperial College, London, UK
| |
Collapse
|
18
|
Zhang C, Yu H, Ye J, Tong H, Wang M, Sun G. Ginsenoside Rg3 Protects against Diabetic Cardiomyopathy and Promotes Adiponectin Signaling via Activation of PPAR-γ. Int J Mol Sci 2023; 24:16736. [PMID: 38069059 PMCID: PMC10705869 DOI: 10.3390/ijms242316736] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Ginsenoside Rg3 extracted from Panax notoginseng has therapeutic effects on diabetes and heart diseases. However, the underlying mechanism of ginsenoside Rg3 on diabetic cardiomyopathy (DCM) remains unclear. 24-week-old diabetic db/db mice were treated with ginsenoside Rg3 for 12 weeks, then body weight, serum lipids, adiponectin levels, as well as cardiac function and pathological morphology, were measured. The targets of ginsenoside Rg3 and its regulation of the adiponectin pathway were also evaluated on 3T3-L1 or H9c2 cells. Ginsenoside Rg3 directly bound to PPAR-γ, improving adiponectin secretion and promoting adiponectin signaling. Significantly attenuated overweight, hyperglycemia, and hyperlipidemia, as well as alleviated lipid accumulation and dysfunction in adipose, liver, and heart tissues, were observed in the ginsenoside Rg3-treated group. Ginsenoside Rg3 could be a promising drug targeting PPAR-γ to treat diabetic cardiomyopathy.
Collapse
Affiliation(s)
| | | | | | | | - Min Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China; (C.Z.); (H.Y.); (J.Y.); (H.T.)
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China; (C.Z.); (H.Y.); (J.Y.); (H.T.)
| |
Collapse
|
19
|
Nelson RK, Solomon R, Hosmer E, Zuhl M. Cardiac rehabilitation utilization, barriers, and outcomes among patients with heart failure. Heart Fail Rev 2023; 28:1239-1249. [PMID: 37039955 DOI: 10.1007/s10741-023-10309-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 04/12/2023]
Abstract
Exercise-based cardiac rehabilitation (CR) is effective for improving both primary (i.e., mortality and hospitalizations) and secondary (i.e., functional capacity and quality of life among) clinical outcomes among patients with heart failure (HF). The mechanisms that explain these benefits are complex and are linked to exercise adaptations such as central and peripheral hemodynamics combined with improved overall medical management. Despite the benefits of CR, utilization rates are low among CR eligible patients. Clinician-, patient-, and health system-related barriers have been identified as primary factors contributing to the lack of CR utilization among HF patients. These include patient referrals (clinician-related), psychosocial factors (patient-related), and patient access to CR services (health system-related). The aims of this review are to detail the components of each barrier as well as identify evidence-based strategies to improve CR utilization and adherence among HF. The improvements in primary and secondary outcomes along with the mechanisms that are linked to these changes will also be examined.
Collapse
Affiliation(s)
- Rachael K Nelson
- School of Health Sciences, Central Michigan University, Mt. Pleasant, MI, USA
| | - Robert Solomon
- Department of Cardiology, Henry Ford Hospital, Detroit, MI, USA
| | - Emily Hosmer
- School of Health Sciences, Central Michigan University, Mt. Pleasant, MI, USA
| | - Micah Zuhl
- School of Health Sciences, Central Michigan University, Mt. Pleasant, MI, USA.
| |
Collapse
|
20
|
Boovarahan SR, Balu K, Prem P, Sivakumar B, Kurian GA. DNA hypomethylation by fisetin preserves mitochondria functional genes and contributes to the protection of I/R rat heart. Funct Integr Genomics 2023; 23:325. [PMID: 37880513 DOI: 10.1007/s10142-023-01257-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023]
Abstract
Myocardial I/R can alter the expression of different sets of cardiac genes that negatively influence the I/R outcome via epigenetic modifications. Fisetin is known to be cardioprotective against I/R, but its underlying epigenetic mode of action is not known and is addressed in the present study. Male Wistar rats were subjected to I/R by using the Langendorff perfusion system. Fisetin (20 mg/kg; i.p.) was administered before I/R induction, followed by the measurement of cardiac injury, hemodynamics, physiological indices, the differential expression of genes that regulate DNA methylation, and the function of mitochondria were performed. Fisetin administered I/R rat heart significantly reduced the global DNA hypermethylation and infarct size with an improved physiological recovery, measured via RPP (81%) and LVDP (82%) from the I/R control. Additionally, we noted decreased expression of the DNMT1 gene by 35% and increased expression of the TET1, TET2, and TET3 genes in fisetin-treated I/R rat hearts. Molecular docking analysis data reveals that the fisetin inhibits DNMT1 at the substrate binding site with minimum binding energy (- 8.2 kcal/mol) compared to the DNMT1 inhibitor, 5-azacytidine. Moreover, fisetin-treated I/R heart reversed the expression of the I/R-linked declined expression of bioenergetics genes (MT-ND1, MT-ND2, MT-ND4, MT-Cyt B, MT-COX1, MT-COX2, MT-ATP6), mitochondrial fission gene (Fis1), replication control genes PGC-1α, POLG, and TFAM to near-normal level. Based on the above findings, we demonstrated that fisetin possesses the ability to modulate the expression of different mitochondrial genes via influencing the global DNA methylation in cardiac tissue, which contributes significantly to the improved contractile function and thereby renders cardioprotection against I/R.
Collapse
Affiliation(s)
- Sri Rahavi Boovarahan
- Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
| | - Kirankumar Balu
- Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
| | - Priyanka Prem
- Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
| | - Bhavana Sivakumar
- Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India
| | - Gino A Kurian
- Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, 613401, India.
| |
Collapse
|
21
|
Malhan D, Schoenrock B, Yalçin M, Blottner D, Relόgio A. Circadian regulation in aging: Implications for spaceflight and life on earth. Aging Cell 2023; 22:e13935. [PMID: 37493006 PMCID: PMC10497835 DOI: 10.1111/acel.13935] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/27/2023] Open
Abstract
Alterations in the circadian system are characteristic of aging on Earth. With the decline in physiological processes due to aging, several health concerns including vision loss, cardiovascular disorders, cognitive impairments, and muscle mass loss arise in elderly populations. Similar health risks are reported as "red flag" risks among astronauts during and after a long-term Space exploration journey. However, little is known about the common molecular alterations underlying terrestrial aging and space-related aging in astronauts, and controversial conclusions have been recently reported. In light of the regulatory role of the circadian clock in the maintenance of human health, we review here the overlapping role of the circadian clock both on aging on Earth and spaceflight with a focus on the four most affected systems: visual, cardiovascular, central nervous, and musculoskeletal systems. In this review, we briefly introduce the regulatory role of the circadian clock in specific cellular processes followed by alterations in those processes due to aging. We next summarize the known molecular alterations associated with spaceflight, highlighting involved clock-regulated genes in space flown Drosophila, nematodes, small mammals, and astronauts. Finally, we discuss common genes that are altered in terms of their expression due to aging on Earth and spaceflight. Altogether, the data elaborated in this review strengthen our hypothesis regarding the timely need to include circadian dysregulation as an emerging hallmark of aging on Earth and beyond.
Collapse
Affiliation(s)
- Deeksha Malhan
- Institute for Systems Medicine and Faculty of Human MedicineMSH Medical School HamburgHamburgGermany
| | - Britt Schoenrock
- Institute of Integrative NeuroanatomyCharité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Müge Yalçin
- Institute for Systems Medicine and Faculty of Human MedicineMSH Medical School HamburgHamburgGermany
- Institute for Theoretical Biology (ITB)Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Dieter Blottner
- Institute of Integrative NeuroanatomyCharité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Neuromuscular System and Neuromuscular SignalingBerlin Center of Space Medicine & Extreme EnvironmentsBerlinGermany
| | - Angela Relόgio
- Institute for Systems Medicine and Faculty of Human MedicineMSH Medical School HamburgHamburgGermany
- Institute for Theoretical Biology (ITB)Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| |
Collapse
|
22
|
Gambardella J, Jankauskas SS, Kansakar U, Varzideh F, Avvisato R, Prevete N, Sidoli S, Mone P, Wang X, Lombardi A, Santulli G. Ketone Bodies Rescue Mitochondrial Dysfunction Via Epigenetic Remodeling. JACC Basic Transl Sci 2023; 8:1123-1137. [PMID: 37791311 PMCID: PMC10543927 DOI: 10.1016/j.jacbts.2023.03.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 10/05/2023]
Abstract
Ischemic cardiac disease is a major cause of mortality worldwide. However, the exact molecular processes underlying this disorder are not fully known. This study includes a comprehensive and coordinated set of in vivo and in vitro experiments using human cardiac specimens from patients with postischemic heart failure (HF) and healthy control subjects, a murine model of HF, and cellular systems. These approaches identified for the first time a specific pattern of maladaptive chromatin remodeling, namely a double methylation of histone 3 at lysine 27 and a single methylation at lysine 36 (H3_K27me2K36me1) consistently induced by ischemic injury in all these settings: human HF; murine HF; and in vitro models. Mechanistically, this work demonstrates that this histone modification mediates the ischemia-induced transcriptional repression of PPARG coactivator 1α (PGC1α), master regulator of mitochondrial function and biogenesis. Intriguingly, both the augmented H3_K27me2K36me1 and the mitochondrial dysfunction ensued by PGC1α down-regulation were significantly attenuated by the treatment with β-hydroxybutyrate, the most abundant ketone body in humans, revealing a novel pathway coupling metabolism to gene expression. Taken together, these findings establish maladaptive chromatin remodeling as a key mechanism in postischemic heart injury, functionally modulated by ketone bodies.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
- Department of Advanced Biomedical Science, “Federico II” University, Naples, Italy
- International Translational Research and Medical Education (ITME) Consortium, Naples, Italy
| | - Stanislovas S. Jankauskas
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Urna Kansakar
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Fahimeh Varzideh
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Roberta Avvisato
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Nella Prevete
- Department of Translational Medical Sciences, “Federico II” University, Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | - Pasquale Mone
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Xujun Wang
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Angela Lombardi
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, New York, New York, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism, Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
- Department of Advanced Biomedical Science, “Federico II” University, Naples, Italy
- International Translational Research and Medical Education (ITME) Consortium, Naples, Italy
- Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
23
|
Parey E, Fernandez-Aroca D, Frost S, Uribarren A, Park TJ, Zöttl M, St John Smith E, Berthelot C, Villar D. Phylogenetic modeling of enhancer shifts in African mole-rats reveals regulatory changes associated with tissue-specific traits. Genome Res 2023; 33:1513-1526. [PMID: 37625847 PMCID: PMC10620049 DOI: 10.1101/gr.277715.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 08/24/2023] [Indexed: 08/27/2023]
Abstract
Changes in gene regulation are thought to underlie most phenotypic differences between species. For subterranean rodents such as the naked mole-rat, proposed phenotypic adaptations include hypoxia tolerance, metabolic changes, and cancer resistance. However, it is largely unknown what regulatory changes may associate with these phenotypic traits, and whether these are unique to the naked mole-rat, the mole-rat clade, or are also present in other mammals. Here, we investigate regulatory evolution in the heart and liver from two African mole-rat species and two rodent outgroups using genome-wide epigenomic profiling. First, we adapted and applied a phylogenetic modeling approach to quantitatively compare epigenomic signals at orthologous regulatory elements and identified thousands of promoter and enhancer regions with differential epigenomic activity in mole-rats. These elements associate with known mole-rat adaptations in metabolic and functional pathways and suggest candidate genetic loci that may underlie mole-rat innovations. Second, we evaluated ancestral and species-specific regulatory changes in the study phylogeny and report several candidate pathways experiencing stepwise remodeling during the evolution of mole-rats, such as the insulin and hypoxia response pathways. Third, we report nonorthologous regulatory elements overlap with lineage-specific repetitive elements and appear to modify metabolic pathways by rewiring of HNF4 and RAR/RXR transcription factor binding sites in mole-rats. These comparative analyses reveal how mole-rat regulatory evolution informs previously reported phenotypic adaptations. Moreover, the phylogenetic modeling framework we propose here improves upon the state of the art by addressing known limitations of inter-species comparisons of epigenomic profiles and has broad implications in the field of comparative functional genomics.
Collapse
Affiliation(s)
- Elise Parey
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Diego Fernandez-Aroca
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - Stephanie Frost
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom
| | - Ainhoa Uribarren
- Cambridge Institute, Cancer Research UK and University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Thomas J Park
- Department of Biological Sciences and Laboratory of Integrative Neuroscience, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Markus Zöttl
- Department of Biology and Environmental Science, Linnaeus University, 44054 Kalmar, Sweden
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, United Kingdom
| | - Camille Berthelot
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France;
- Institut Pasteur, Université Paris Cité, CNRS UMR 3525, INSERM UA12, Comparative Functional Genomics Group, F-75015 Paris, France
| | - Diego Villar
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, United Kingdom;
| |
Collapse
|
24
|
Patintingan CG, Louisa M, Juniantito V, Arozal W, Hanifah S, Wanandi SI, Thandavarayan R. Moringa oleifera Leaves Extract Ameliorates Doxorubicin-Induced Cardiotoxicity via Its Mitochondrial Biogenesis Modulatory Activity in Rats. J Exp Pharmacol 2023; 15:307-319. [PMID: 37525636 PMCID: PMC10387274 DOI: 10.2147/jep.s413256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 07/12/2023] [Indexed: 08/02/2023] Open
Abstract
Background Doxorubicin, an anthracycline class of anticancer, is an effective chemotherapeutic agent with serious adverse effects, mainly cardiotoxicity. Several possible causes of doxorubicin cardiotoxicity are increased oxidative stress, nucleic acid and protein synthesis inhibition, cardiomyocyte apoptosis, and mitochondrial biogenesis disruptions. Moringa oleifera (MO), a naturally derived medicine, is known for its antioxidative properties and activity in alleviating mitochondrial dysfunction. To determine the potency and possible cardioprotective mechanism of MO leaves aqueous extract via the mitochondrial biogenesis pathway in doxorubicin-induced rats. Methods Twenty-four Sprague-Dawley rats were divided into four groups of six. The first group was normal rats; the second group was treated with doxorubicin 4 mg/kg BW intraperitoneally once weekly for four weeks; the third and fourth groups were treated with doxorubicin 4 mg/kg BW intraperitoneally once weekly, and MO leaves extract at 200 mg/kg BW or 400 mg/kg BW orally daily, for four weeks. At the end of the fourth week, blood and cardiac tissues were obtained and analyzed for cardiac biomarkers, mitochondrial DNA copy number, mRNA expressions of peroxisome-activated receptor-gamma coactivator-1 alpha (PGC-1α), the nuclear factor erythroid 2-related factor 2 (Nrf2), superoxide dismutase 2 (SOD2), caspase 3, the activity of glutathione peroxidase (GPx), levels of 8-hydroxy-2-deoxyguanosine (8-OH-dG), and malondialdehyde. Results MO leaves extract was shown to decrease biomarkers of cardiac damage (LDH and CK-MB), malondialdehyde levels, and GPx activity. These changes align with the reduction of mRNA expressions of caspase-3, the increase of mRNA expressions of PGC-1α and Nrf2, and the elevation of mitochondrial DNA copy number. MO leaves extracts did not influence the mRNA expressions of superoxide dismutase 2 (SOD2) or the levels of 8-OH-dG. Conclusion Moringa oleifera leaves extract ameliorates doxorubicin-induced cardiotoxicity by reducing apoptosis and restoring gene expression of PGC-1α and Nrf2, a key regulator in mitochondrial biogenesis.
Collapse
Affiliation(s)
| | - Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Vetnizah Juniantito
- Department of Veterinary Clinic Reproduction and Pathology, Faculty of Veterinary Medicine, Agriculture Institute of Bogor, Bogor, Indonesia
| | - Wawaimuli Arozal
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Silmi Hanifah
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Septelia Inawati Wanandi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Rajarajan Thandavarayan
- Department of Cardiovascular Sciences Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
25
|
Szulik MW, Valdez S, Walsh M, Davis K, Bia R, Horiuchi E, O'Very S, Laxman AK, Sandaklie-Nicolova L, Eberhardt DR, Durrant JR, Sheikh H, Hickenlooper S, Creed M, Brady C, Miller M, Wang L, Garcia-Llana J, Tracy C, Drakos SG, Funai K, Chaudhuri D, Boudina S, Franklin S. SMYD1a protects the heart from ischemic injury by regulating OPA1-mediated cristae remodeling and supercomplex formation. Basic Res Cardiol 2023; 118:20. [PMID: 37212935 PMCID: PMC10203008 DOI: 10.1007/s00395-023-00991-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 05/23/2023]
Abstract
SMYD1, a striated muscle-specific lysine methyltransferase, was originally shown to play a key role in embryonic cardiac development but more recently we demonstrated that loss of Smyd1 in the murine adult heart leads to cardiac hypertrophy and failure. However, the effects of SMYD1 overexpression in the heart and its molecular function in the cardiomyocyte in response to ischemic stress are unknown. In this study, we show that inducible, cardiomyocyte-specific overexpression of SMYD1a in mice protects the heart from ischemic injury as seen by a > 50% reduction in infarct size and decreased myocyte cell death. We also demonstrate that attenuated pathological remodeling is a result of enhanced mitochondrial respiration efficiency, which is driven by increased mitochondrial cristae formation and stabilization of respiratory chain supercomplexes within the cristae. These morphological changes occur concomitant with increased OPA1 expression, a known driver of cristae morphology and supercomplex formation. Together, these analyses identify OPA1 as a novel downstream target of SMYD1a whereby cardiomyocytes upregulate energy efficiency to dynamically adapt to the energy demands of the cell. In addition, these findings highlight a new epigenetic mechanism by which SMYD1a regulates mitochondrial energetics and functions to protect the heart from ischemic injury.
Collapse
Affiliation(s)
- Marta W Szulik
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA.
| | - Steven Valdez
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Maureen Walsh
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
| | - Kathryn Davis
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Ryan Bia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Emilee Horiuchi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Sean O'Very
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Anil K Laxman
- Metabolic Phenotypic Core Facility, University of Utah, Salt Lake City, UT, USA
| | | | - David R Eberhardt
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Hanin Sheikh
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Samuel Hickenlooper
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Magnus Creed
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Cameron Brady
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Mickey Miller
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Li Wang
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - June Garcia-Llana
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Christopher Tracy
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Sarah Franklin
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA.
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
26
|
Ali AU, Abd-Elkareem M, Kamel AA, Abou Khalil NS, Hamad D, Nasr NEH, Hassan MA, El Faham TH. Impact of porous microsponges in minimizing myotoxic side effects of simvastatin. Sci Rep 2023; 13:5790. [PMID: 37031209 PMCID: PMC10082807 DOI: 10.1038/s41598-023-32545-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 03/29/2023] [Indexed: 04/10/2023] Open
Abstract
Simvastatin (SV) is a poorly soluble drug; its oral administration is associated with a significant problem: Myopathy. The present study aims to formulate SV microsponges that have the potential to minimize the myotoxicity accompanying the oral administration of the drug. SV microsponges were prepared by exploiting the emulsion solvent evaporation technique. The % entrapment efficiency (%EE) of the drug approached 82.54 ± 1.27%, the mean particle size of SV microsponges ranged from 53.80 ± 6.35 to 86.03 ± 4.79 µm in diameter, and the % cumulative drug release (%CDR) of SV from microsponges was significantly higher than that from free drug dispersion much more, the specific surface area of the optimized microsponges formulation was found to be 16.6 m2/g revealed the porosity of prepared microsponges. Histological and glycogen histochemical studies in the skeletal muscles of male albino rats revealed that microsponges were safer than free SV in minimizing myotoxicity. These findings were proven by Gene expression of Mitochondrial fusion and fission (Mfn1) & (Fis1) and (Peroxisome proliferator-activated receptor gamma co-activator 1α) PGC-1α. Finally, our study ascertained that SV microsponges significantly decreased the myotoxicity of SV.
Collapse
Affiliation(s)
- Ahmed U Ali
- Department of Pharmaceutics, Faculty of Pharmacy, Merit University, Sohag, Egypt.
| | - Mahmoud Abd-Elkareem
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Amira A Kamel
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Nasser S Abou Khalil
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - D Hamad
- Department of Physics, Faculty of Science, Assiut University, Assiut, Egypt
| | | | - Maha A Hassan
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Tahani H El Faham
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| |
Collapse
|
27
|
Kankuri E, Finckenberg P, Leinonen J, Tarkia M, Björk S, Purhonen J, Kallijärvi J, Kankainen M, Soliymani R, Lalowski M, Mervaala E. Altered acylcarnitine metabolism and inflexible mitochondrial fuel utilization characterize the loss of neonatal myocardial regeneration capacity. Exp Mol Med 2023; 55:806-817. [PMID: 37009793 PMCID: PMC10167339 DOI: 10.1038/s12276-023-00967-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 11/29/2022] [Accepted: 12/23/2022] [Indexed: 04/04/2023] Open
Abstract
Myocardial regeneration capacity declines during the first week after birth, and this decline is linked to adaptation to oxidative metabolism. Utilizing this regenerative window, we characterized the metabolic changes in myocardial injury in 1-day-old regeneration-competent and 7-day-old regeneration-compromised mice. The mice were either sham-operated or received left anterior descending coronary artery ligation to induce myocardial infarction (MI) and acute ischemic heart failure. Myocardial samples were collected 21 days after operations for metabolomic, transcriptomic and proteomic analyses. Phenotypic characterizations were carried out using echocardiography, histology and mitochondrial structural and functional assessments. In both groups, MI induced an early decline in cardiac function that persisted in the regeneration-compromised mice over time. By integrating the findings from metabolomic, transcriptomic and proteomic examinations, we linked regeneration failure to the accumulation of long-chain acylcarnitines and insufficient metabolic capacity for fatty acid beta-oxidation. Decreased expression of the redox-sensitive mitochondrial Slc25a20 carnitine-acylcarnitine translocase together with a decreased reduced:oxidized glutathione ratio in the myocardium in the regeneration-compromised mice pointed to a defect in the redox-sensitive acylcarnitine transport to the mitochondrial matrix. Rather than a forced shift from the preferred adult myocardial oxidative fuel source, our results suggest the facilitation of mitochondrial fatty acid transport and improvement of the beta-oxidation pathway as a means to overcome the metabolic barrier for repair and regeneration in adult mammals after MI and heart failure.
Collapse
Affiliation(s)
- E Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - P Finckenberg
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J Leinonen
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Tarkia
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - S Björk
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J Purhonen
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J Kallijärvi
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Kankainen
- Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - R Soliymani
- Helsinki Institute of Life Science (HiLIFE), Meilahti Clinical Proteomics Core Facility, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Lalowski
- Helsinki Institute of Life Science (HiLIFE), Meilahti Clinical Proteomics Core Facility, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - E Mervaala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
28
|
Balatskyi VV, Sowka A, Dobrzyn P, Piven OO. WNT/β-catenin pathway is a key regulator of cardiac function and energetic metabolism. Acta Physiol (Oxf) 2023; 237:e13912. [PMID: 36599355 DOI: 10.1111/apha.13912] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/24/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
The WNT/β-catenin pathway is a master regulator of cardiac development and growth, and its activity is low in healthy adult hearts. However, even this low activity is essential for maintaining normal heart function. Acute activation of the WNT/β-catenin signaling cascade is considered to be cardioprotective after infarction through the upregulation of prosurvival genes and reprogramming of metabolism. Chronically high WNT/β-catenin pathway activity causes profibrotic and hypertrophic effects in the adult heart. New data suggest more complex functions of β-catenin in metabolic maturation of the perinatal heart, establishing an adult pattern of glucose and fatty acid utilization. Additionally, low basal activity of the WNT/β-catenin cascade maintains oxidative metabolism in the adult heart, and this pathway is reactivated by physiological or pathological stimuli to meet the higher energy needs of the heart. This review summarizes the current state of knowledge of the organization of canonical WNT signaling and its function in cardiogenesis, heart maturation, adult heart function, and remodeling. We also discuss the role of the WNT/β-catenin pathway in cardiac glucose, lipid metabolism, and mitochondrial physiology.
Collapse
Affiliation(s)
- Volodymyr V Balatskyi
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Adrian Sowka
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Oksana O Piven
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Department of Human Genetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
29
|
Ayyappan P, Larsen TD, Gandy TCT, Louwagie EJ, Baack ML. Impact of Prenatal Exposure to Maternal Diabetes and High-Fat Diet on Postnatal Myocardial Ketone Body Metabolism in Rats. Int J Mol Sci 2023; 24:3684. [PMID: 36835096 PMCID: PMC9967912 DOI: 10.3390/ijms24043684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Infants exposed to diabetic pregnancy are at higher risk of cardiomyopathy at birth and early onset cardiovascular disease (CVD) as adults. Using a rat model, we showed how fetal exposure to maternal diabetes causes cardiac disease through fuel-mediated mitochondrial dysfunction, and that a maternal high-fat diet (HFD) exaggerates the risk. Diabetic pregnancy increases circulating maternal ketones which can have a cardioprotective effect, but whether diabetes-mediated complex I dysfunction impairs myocardial metabolism of ketones postnatally remains unknown. The objective of this study was to determine whether neonatal rat cardiomyocytes (NRCM) from diabetes- and HFD-exposed offspring oxidize ketones as an alternative fuel source. To test our hypothesis, we developed a novel ketone stress test (KST) using extracellular flux analyses to compare real-time ß-hydroxybutyrate (βHOB) metabolism in NRCM. We also compared myocardial expression of genes responsible for ketone and lipid metabolism. NRCM had a dose-dependent increase in respiration with increasing concentrations of βHOB, demonstrating that both control and combination exposed NRCM can metabolize ketones postnatally. Ketone treatment also enhanced the glycolytic capacity of combination exposed NRCM with a dose-dependent increase in the glucose-mediated proton efflux rate (PER) from CO2 (aerobic glycolysis) alongside a decreased reliance on PER from lactate (anaerobic glycolysis). Expression of genes responsible for ketone body metabolism was higher in combination exposed males. Findings demonstrate that myocardial ketone body metabolism is preserved and improves fuel flexibility in NRCM from diabetes- and HFD-exposed offspring, which suggests that ketones might serve a protective role in neonatal cardiomyopathy due to maternal diabetes.
Collapse
Affiliation(s)
- Prathapan Ayyappan
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Tricia D. Larsen
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Tyler C. T. Gandy
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Eli J. Louwagie
- Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| | - Michelle L. Baack
- Environmental Influences on Health and Disease Group, Sanford Research, Sioux Falls, SD 57104, USA
- Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
30
|
Morciano G, Boncompagni C, Ramaccini D, Pedriali G, Bouhamida E, Tremoli E, Giorgi C, Pinton P. Comprehensive Analysis of Mitochondrial Dynamics Alterations in Heart Diseases. Int J Mol Sci 2023; 24:ijms24043414. [PMID: 36834825 PMCID: PMC9961104 DOI: 10.3390/ijms24043414] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
The most common alterations affecting mitochondria, and associated with cardiac pathological conditions, implicate a long list of defects. They include impairments of the mitochondrial electron transport chain activity, which is a crucial element for energy formation, and that determines the depletion of ATP generation and supply to metabolic switches, enhanced ROS generation, inflammation, as well as the dysregulation of the intracellular calcium homeostasis. All these signatures significantly concur in the impairment of cardiac electrical characteristics, loss of myocyte contractility and cardiomyocyte damage found in cardiac diseases. Mitochondrial dynamics, one of the quality control mechanisms at the basis of mitochondrial fitness, also result in being dysregulated, but the use of this knowledge for translational and therapeutic purposes is still in its infancy. In this review we tried to understand why this is, by summarizing methods, current opinions and molecular details underlying mitochondrial dynamics in cardiac diseases.
Collapse
Affiliation(s)
- Giampaolo Morciano
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- GVM Care & Research, Maria Cecilia Hospital, 48033 Cotignola, Italy
- Correspondence: (G.M.); (P.P.); Tel.: +05-32-455-802 (G.M. & P.P.)
| | | | | | - Gaia Pedriali
- GVM Care & Research, Maria Cecilia Hospital, 48033 Cotignola, Italy
| | - Esmaa Bouhamida
- GVM Care & Research, Maria Cecilia Hospital, 48033 Cotignola, Italy
| | - Elena Tremoli
- GVM Care & Research, Maria Cecilia Hospital, 48033 Cotignola, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- GVM Care & Research, Maria Cecilia Hospital, 48033 Cotignola, Italy
- Correspondence: (G.M.); (P.P.); Tel.: +05-32-455-802 (G.M. & P.P.)
| |
Collapse
|
31
|
Xu Q, Zhao YM, He NQ, Gao R, Xu WX, Zhuo XJ, Ren Z, Wu CY, Liu LS. PCSK9: A emerging participant in heart failure. Biomed Pharmacother 2023; 158:114106. [PMID: 36535197 DOI: 10.1016/j.biopha.2022.114106] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Heart failure (HF) is a complex clinical syndrome caused by various cardiovascular diseases. Its main pathogenesis includes cardiomyocyte loss, myocardial energy metabolism disorder, and activation of cardiac inflammation. Due to the clinically unsatisfactory treatment of heart failure, different mechanisms need to be explored to provide new targets for the treatment of this disease. Proprotein convertase subtilisin/kexin type 9 (PCSK9), a gene mainly related to familial hypercholesterolemia, was discovered in 2003. Aside from regulating lipid metabolism, PCSK9 may be involved in other biological processes such as apoptosis, autophagy, pyroptosis, inflammation, and tumor immunity and related to diabetes and neurodegenerative diseases. Recently, clinical data have shown that the circulating PCSK9 level is significantly increased in patients with heart failure, and it is related to the prognosis for heart failure. Furthermore, in animal models and patients with myocardial infarction, PCSK9 in the infarct margin area was also found to be significantly increased, which further suggested that PCSK9 might be closely related to heart failure. However, the specific mechanism of how PCSK9 participates in heart failure remains to be further explored. The purpose of this review is to summarize the potential mechanism of PCSK9's involvement in heart failure, thereby providing a new treatment strategy for heart failure.
Collapse
Affiliation(s)
- Qian Xu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Yi-Meng Zhao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Nai-Qi He
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Rong Gao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Wen-Xin Xu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Xiu-Juan Zhuo
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China
| | - Chun-Yan Wu
- The Third Affiliated Hospital, Department of Cardiovascular Medicine, University of South China, Hengyang, Hunan Province 421001, PR China.
| | - Lu-Shan Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan Province 421001, PR China.
| |
Collapse
|
32
|
Álvarez M, Ruiz-Sala P, Pérez B, Desviat LR, Richard E. Dysregulated Cell Homeostasis and miRNAs in Human iPSC-Derived Cardiomyocytes from a Propionic Acidemia Patient with Cardiomyopathy. Int J Mol Sci 2023; 24:ijms24032182. [PMID: 36768524 PMCID: PMC9916417 DOI: 10.3390/ijms24032182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Propionic acidemia (PA) disorder shows major involvement of the heart, among other alterations. A significant number of PA patients develop cardiac complications, and available evidence suggests that this cardiac dysfunction is driven mainly by the accumulation of toxic metabolites. To contribute to the elucidation of the mechanistic basis underlying this dysfunction, we have successfully generated cardiomyocytes through the differentiation of induced pluripotent stem cells (iPSCs) from a PCCB patient and its isogenic control. In this human cellular model, we aimed to examine microRNAs (miRNAs) profiles and analyze several cellular pathways to determine miRNAs activity patterns associated with PA cardiac phenotypes. We have identified a series of upregulated cardiac-enriched miRNAs and alterations in some of their regulated signaling pathways, including an increase in the expression of cardiac damage markers and cardiac channels, an increase in oxidative stress, a decrease in mitochondrial respiration and autophagy; and lipid accumulation. Our findings indicate that miRNA activity patterns from PA iPSC-derived cardiomyocytes are biologically informative and advance the understanding of the molecular mechanisms of this rare disease, providing a basis for identifying new therapeutic targets for intervention strategies.
Collapse
Affiliation(s)
- Mar Álvarez
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Pedro Ruiz-Sala
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, 28029 Madrid, Spain
| | - Belén Pérez
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, 28029 Madrid, Spain
| | - Lourdes Ruiz Desviat
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, 28029 Madrid, Spain
| | - Eva Richard
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
33
|
Monterrubio-Ledezma F, Navarro-García F, Massieu L, Mondragón-Flores R, Soto-Ponce LA, Magaña JJ, Cisneros B. Rescue of Mitochondrial Function in Hutchinson-Gilford Progeria Syndrome by the Pharmacological Modulation of Exportin CRM1. Cells 2023; 12:275. [PMID: 36672210 PMCID: PMC9856861 DOI: 10.3390/cells12020275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/14/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disorder caused by the expression of progerin, a mutant variant of Lamin A. Recently, HGPS studies have gained relevance because unraveling its underlying mechanism would help to understand physiological aging. We previously reported that the CRM1-mediated nuclear protein export pathway is exacerbated in HGPS cells, provoking the mislocalization of numerous protein targets of CRM1. We showed that normalization of this mechanism by pharmacologically inhibiting CRM1 with LMB (specific CRM1 inhibitor), mitigates the senescent phenotype of HGPS cells. Since mitochondrial dysfunction is a hallmark of HGPS, in this study we analyze the effect of LMB on mitochondrial function. Remarkably, LMB treatment induced the recovery of mitochondrial function in HGPS cells, as shown by the improvement in mitochondrial morphology, mitochondrial membrane potential, and ATP levels, which consequently impeded the accumulation of ROS but not mitochondrial superoxide. We provide evidence that the beneficial effect of LMB is mechanistically based on a combinatory effect on mitochondrial biogenesis via upregulation of PGC-1α expression (master transcription cofactor of mitochondrial genes), and mitophagy through the recovery of lysosomal content. The use of exportin CRM1 inhibitors constitutes a promising strategy to treat HGPS and other diseases characterized by mitochondrial impairment.
Collapse
Affiliation(s)
- Feliciano Monterrubio-Ledezma
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| | - Fernando Navarro-García
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| | - Lourdes Massieu
- Department of Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
| | - Ricardo Mondragón-Flores
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| | - Luz Adriana Soto-Ponce
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| | - Jonathan J. Magaña
- Departament of Bioengineering, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey-Campus Ciudad de México, Ciudad de Mexico 14380, Mexico
- Laboratory of Genomic Medicine, Department of Genetics, National Rehabilitation Institute-Luis Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Bulmaro Cisneros
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| |
Collapse
|
34
|
Booth LK, Redgrave RE, Tual-Chalot S, Spyridopoulos I, Phillips HM, Richardson GD. Heart Disease and Ageing: The Roles of Senescence, Mitochondria, and Telomerase in Cardiovascular Disease. Subcell Biochem 2023; 103:45-78. [PMID: 37120464 DOI: 10.1007/978-3-031-26576-1_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
During ageing molecular damage leads to the accumulation of several hallmarks of ageing including mitochondrial dysfunction, cellular senescence, genetic instability and chronic inflammation, which contribute to the development and progression of ageing-associated diseases including cardiovascular disease. Consequently, understanding how these hallmarks of biological ageing interact with the cardiovascular system and each other is fundamental to the pursuit of improving cardiovascular health globally. This review provides an overview of our current understanding of how candidate hallmarks contribute to cardiovascular diseases such as atherosclerosis, coronary artery disease and subsequent myocardial infarction, and age-related heart failure. Further, we consider the evidence that, even in the absence of chronological age, acute cellular stress leading to accelerated biological ageing expedites cardiovascular dysfunction and impacts on cardiovascular health. Finally, we consider the opportunities that modulating hallmarks of ageing offer for the development of novel cardiovascular therapeutics.
Collapse
Affiliation(s)
- Laura K Booth
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Rachael E Redgrave
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Helen M Phillips
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
35
|
Kim YS, Hollmann MW, Van Lieshout JJ. Editorial: Insights in clinical and translational physiology: 2022. Front Physiol 2023; 14:1170997. [PMID: 36960157 PMCID: PMC10028462 DOI: 10.3389/fphys.2023.1170997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023] Open
Affiliation(s)
- Yu-Sok Kim
- Department of Internal Medicine, Medisch Centrum Leeuwarden, Amsterdam, Netherlands
- Laboratory for Clinical Cardiovascular Physiology, University of Amsterdam, Amsterdam, Netherlands
| | - Markus W. Hollmann
- Department of Anaesthesiology, University Medical Center (UMC), Amsterdam, Netherlands
| | - Johannes J. Van Lieshout
- Department of Internal Medicine, Medisch Centrum Leeuwarden, Amsterdam, Netherlands
- Department of Internal Medicine, University Medical Center (UMC), Amsterdam, Netherlands
- The David Greenfield Human Physiology Unit, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Pharmacology and Neuroscience, School of Life Sciences, The Medical School, University of Nottingham Medical School, Queen’s Medical Centre, Nottingham, United Kingdom
- *Correspondence: Johannes J. Van Lieshout,
| |
Collapse
|
36
|
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: 12] [Impact Index Per Article: 4.0] [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
|
37
|
Lim AY, Chen YC, Hsu CC, Fu TC, Wang JS. The Effects of Exercise Training on Mitochondrial Function in Cardiovascular Diseases: A Systematic Review and Meta-Analysis. Int J Mol Sci 2022; 23:12559. [PMID: 36293409 PMCID: PMC9603958 DOI: 10.3390/ijms232012559] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 11/25/2022] Open
Abstract
Mitochondria dysfunction is implicated in the pathogenesis of cardiovascular diseases (CVD). Exercise training is potentially an effective non-pharmacological strategy to restore mitochondrial health in CVD. However, how exercise modifies mitochondrial functionality is inconclusive. We conducted a systematic review using the PubMed; Scopus and Web of Science databases to investigate the effect of exercise training on mitochondrial function in CVD patients. Search terms included “mitochondria”, “exercise”, “aerobic capacity”, and “cardiovascular disease” in varied combination. The search yielded 821 records for abstract screening, of which 20 articles met the inclusion criteria. We summarized the effect of exercise training on mitochondrial morphology, biogenesis, dynamics, oxidative capacity, antioxidant capacity, and quality. Amongst these parameters, only oxidative capacity was suitable for a meta-analysis, which demonstrated a significant effect size of exercise in improving mitochondrial oxidative capacity in CVD patients (SMD = 4.78; CI = 2.99 to 6.57; p < 0.01), but with high heterogeneity among the studies (I2 = 75%, p = 0.003). Notably, aerobic exercise enhanced succinate-involved oxidative phosphorylation. The majority of the results suggested that exercise improves morphology and biogenesis, whereas findings on dynamic, antioxidant capacity, and quality, were inadequate or inconclusive. A further randomized controlled trial is clearly required to explain how exercise modifies the pathway of mitochondrial quantity and quality in CVD patients.
Collapse
Affiliation(s)
- Ai Yin Lim
- Healthy Aging Research Center, Graduate Institute of Rehabilitation Science, Chang Gung University, Taoyuan 333, Taiwan
| | - Yi-Ching Chen
- Department of Information Management, Chang Gung University, Taoyuan 333, Taiwan
| | - Chih-Chin Hsu
- Heart Failure Center, Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Tieh-Cheng Fu
- Heart Failure Center, Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Jong-Shyan Wang
- Healthy Aging Research Center, Graduate Institute of Rehabilitation Science, Chang Gung University, Taoyuan 333, Taiwan
- Heart Failure Center, Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| |
Collapse
|
38
|
Guajardo-Correa E, Silva-Agüero JF, Calle X, Chiong M, Henríquez M, García-Rivas G, Latorre M, Parra V. Estrogen signaling as a bridge between the nucleus and mitochondria in cardiovascular diseases. Front Cell Dev Biol 2022; 10:968373. [PMID: 36187489 PMCID: PMC9516331 DOI: 10.3389/fcell.2022.968373] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Epidemiological studies indicate that pre-menopausal women are more protected against the development of CVDs compared to men of the same age. This effect is attributed to the action/effects of sex steroid hormones on the cardiovascular system. In this context, estrogen modulates cardiovascular function in physiological and pathological conditions, being one of the main physiological cardioprotective agents. Here we describe the common pathways and mechanisms by which estrogens modulate the retrograde and anterograde communication between the nucleus and mitochondria, highlighting the role of genomic and non-genomic pathways mediated by estrogen receptors. Additionally, we discuss the presumable role of bromodomain-containing protein 4 (BRD4) in enhancing mitochondrial biogenesis and function in different CVD models and how this protein could act as a master regulator of estrogen protective activity. Altogether, this review focuses on estrogenic control in gene expression and molecular pathways, how this activity governs nucleus-mitochondria communication, and its projection for a future generation of strategies in CVDs treatment.
Collapse
Affiliation(s)
- Emanuel Guajardo-Correa
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Juan Francisco Silva-Agüero
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Ximena Calle
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
- Center of Applied Nanoscience (CANS), Facultad de Ciencias Exactas, Universidad Andres Bello, Santiago, Chile
| | - Mario Chiong
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Mauricio Henríquez
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Red para el Estudio de Enfermedades Cardiopulmonares de Alta Letalidad (REECPAL), Universidad de Chile, Santiago, Chile
| | - Gerardo García-Rivas
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, Mexico
- Tecnológico de Monterrey, The Institute for Obesity Research, Hospital Zambrano Hellion, San Pedro Garza Garcia, Nuevo León, Mexico
| | - Mauricio Latorre
- Laboratorio de Bioingeniería, Instituto de Ciencias de la Ingeniería, Universidad de O’Higgins, Rancagua, Chile
- Laboratorio de Bioinformática y Expresión Génica, INTA, Universidad de Chile, Santiago, Chile
| | - Valentina Parra
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Red para el Estudio de Enfermedades Cardiopulmonares de Alta Letalidad (REECPAL), Universidad de Chile, Santiago, Chile
| |
Collapse
|
39
|
Stamerra CA, Di Giosia P, Giorgini P, Ferri C, Sukhorukov VN, Sahebkar A. Mitochondrial Dysfunction and Cardiovascular Disease: Pathophysiology and Emerging Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9530007. [PMID: 35958017 PMCID: PMC9363184 DOI: 10.1155/2022/9530007] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022]
Abstract
Mitochondria ensure the supply of cellular energy through the production of ATP via oxidative phosphorylation. The alteration of this process, called mitochondrial dysfunction, leads to a reduction in ATP and an increase in the production of reactive oxygen species (ROS). Mitochondrial dysfunction can be caused by mitochondrial/nuclear DNA mutations, or it can be secondary to pathological conditions such as cardiovascular disease, aging, and environmental stress. The use of therapies aimed at the prevention/correction of mitochondrial dysfunction, in the context of the specific treatment of cardiovascular diseases, is a topic of growing interest. In this context, the data are conflicting since preclinical studies are numerous, but there are no large randomized studies.
Collapse
Affiliation(s)
- Cosimo Andrea Stamerra
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
- Department of Internal Medicine, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Paolo Di Giosia
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
- Department of Internal Medicine, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Paolo Giorgini
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
| | - Claudio Ferri
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
| | - Vasily N. Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, Moscow 121609, Russia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
40
|
Investigating the role of DNMT1 gene expression on myocardial ischemia reperfusion injury in rat and associated changes in mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148566. [PMID: 35489443 DOI: 10.1016/j.bbabio.2022.148566] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 12/31/2022]
Abstract
Altered DNA methylation and mitochondrial dysfunction are the two key features of myocardial ischemia reperfusion injury (I/R), but their association with I/R remains unknown. In the present study, the relationship between DNA methyl transferase1 (DNMT1), the key methylation gene, and the mitochondrial quality control genes in rat heart during I/R was explored. We used the Langendorff rat heart model with 30 min of ischemia followed by 60 min of reperfusion and subsequent inhibition of DNMT1 with 5-azacytidine to evaluate the role of DNA methylation in I/R. Reperfusion significantly increased the expression of the DNMT1 gene, enzyme activity, and global DNA methylation levels, along with decreased mitochondrial copy, electron transport chain (ETC) activities, and ATP level. This was in agreement with the significant downregulation of 11 mitochondrial genes PGC-1α, TFAM, POLG, MFN1 and MFN2, FIS1, PARKIN, OPTN, ND1, ND4L, Cyt B and COX1 in I/R induced rat hearts. The expression pattern of the mitochondrial genes PGC-1α, TFAM, ND1 and Cyt B showed a significant negative correlation with DNMT1 expression. Rate pressure product, index of cardiac performance negatively correlated with DNMT1 expression (r = -0.8231, p = 0.0456). However, DNMT1 inhibited rat hearts via 5-azacytidine significantly improved the heart from I/R injury and reversed the I/R associated changes in the gene expression of TFAM, POLG, PGC-1α, ND1, COX1 and Cyt B, and improved the overall mtDNA copies, with a subsequent improvement in the ETC enzyme activity and ATP levels. To conclude, I/R augmented the DNMT1 activity with a subsequent increase in cardiac injury via downregulating the mitochondrial functional genes.
Collapse
|
41
|
Schillemans T, Tragante V, Maitusong B, Gigante B, Cresci S, Laguzzi F, Vikström M, Richards M, Pilbrow A, Cameron V, Foco L, Doughty RN, Kuukasjärvi P, Allayee H, Hartiala JA, Tang WHW, Lyytikäinen LP, Nikus K, Laurikka JO, Srinivasan S, Mordi IR, Trompet S, Kraaijeveld A, van Setten J, Gijsberts CM, Maitland-van der Zee AH, Saely CH, Gong Y, Johnson JA, Cooper-DeHoff RM, Pepine CJ, Casu G, Leiherer A, Drexel H, Horne BD, van der Laan SW, Marziliano N, Hazen SL, Sinisalo J, Kähönen M, Lehtimäki T, Lang CC, Burkhardt R, Scholz M, Jukema JW, Eriksson N, Åkerblom A, James S, Held C, Hagström E, Spertus JA, Algra A, de Faire U, Åkesson A, Asselbergs FW, Patel RS, Leander K. Associations of Polymorphisms in the Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1 Alpha Gene With Subsequent Coronary Heart Disease: An Individual-Level Meta-Analysis. Front Physiol 2022; 13:909870. [PMID: 35812313 PMCID: PMC9260705 DOI: 10.3389/fphys.2022.909870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The knowledge of factors influencing disease progression in patients with established coronary heart disease (CHD) is still relatively limited. One potential pathway is related to peroxisome proliferator–activated receptor gamma coactivator-1 alpha (PPARGC1A), a transcription factor linked to energy metabolism which may play a role in the heart function. Thus, its associations with subsequent CHD events remain unclear. We aimed to investigate the effect of three different SNPs in the PPARGC1A gene on the risk of subsequent CHD in a population with established CHD. Methods: We employed an individual-level meta-analysis using 23 studies from the GENetIcs of sUbSequent Coronary Heart Disease (GENIUS-CHD) consortium, which included participants (n = 80,900) with either acute coronary syndrome, stable CHD, or a mixture of both at baseline. Three variants in the PPARGC1A gene (rs8192678, G482S; rs7672915, intron 2; and rs3755863, T528T) were tested for their associations with subsequent events during the follow-up using a Cox proportional hazards model adjusted for age and sex. The primary outcome was subsequent CHD death or myocardial infarction (CHD death/myocardial infarction). Stratified analyses of the participant or study characteristics as well as additional analyses for secondary outcomes of specific cardiovascular disease diagnoses and all-cause death were also performed. Results: Meta-analysis revealed no significant association between any of the three variants in the PPARGC1A gene and the primary outcome of CHD death/myocardial infarction among those with established CHD at baseline: rs8192678, hazard ratio (HR): 1.01, 95% confidence interval (CI) 0.98–1.05 and rs7672915, HR: 0.97, 95% CI 0.94–1.00; rs3755863, HR: 1.02, 95% CI 0.99–1.06. Similarly, no significant associations were observed for any of the secondary outcomes. The results from stratified analyses showed null results, except for significant inverse associations between rs7672915 (intron 2) and the primary outcome among 1) individuals aged ≥65, 2) individuals with renal impairment, and 3) antiplatelet users. Conclusion: We found no clear associations between polymorphisms in the PPARGC1A gene and subsequent CHD events in patients with established CHD at baseline.
Collapse
Affiliation(s)
- Tessa Schillemans
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vinicius Tragante
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Buamina Maitusong
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bruna Gigante
- Division of Cardiovascular Medicine, Department of Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Sciences, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Sharon Cresci
- Cardiovascular Division, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Federica Laguzzi
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Max Vikström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mark Richards
- Department of Medicine, Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Anna Pilbrow
- Department of Medicine, Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Vicky Cameron
- Department of Medicine, Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Luisa Foco
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | - Robert N. Doughty
- Heart Health Research Group, The University of Auckland, Auckland, New Zealand
| | - Pekka Kuukasjärvi
- Finnish Cardiovascular Research Center - Tampere, Department of Cardio-Thoracic Surgery, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Hooman Allayee
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jaana A. Hartiala
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - W. H. Wilson Tang
- Department of Cardiovascular and Metabolic Sciences and Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic Ohio, Cleveland, OH, United States
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic Ohio, Cleveland, OH, United States
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories Ltd., Tampere, Finland
- Finnish Cardiovascular Research Center - Tampere, Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kjell Nikus
- Finnish Cardiovascular Research Center - Tampere, Department of Cardiology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Heart Center, Department of Cardiology, Tampere University Hospital, Tampere, Finland
| | - Jari O. Laurikka
- Finnish Cardiovascular Research Center - Tampere, Department of Cardio-Thoracic Surgery, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Heart Center, Department of Thoracic Surgery, Tampere University Hospital, Tampere, Finland
| | - Sundararajan Srinivasan
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Ify R. Mordi
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Stella Trompet
- Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
- Section of Gerontology and Geriatrics, and Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Adriaan Kraaijeveld
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jessica van Setten
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Crystel M. Gijsberts
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Cardiology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Anke H. Maitland-van der Zee
- Amsterdam University Medical Centers, Department of Respiratory Medicine, University of Amsterdam, Amsterdam, Netherlands
| | - Christoph H. Saely
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
- Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Yan Gong
- Center for Pharmacogenomics and Precision Medicine, Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL, United States
| | - Julie A. Johnson
- Center for Pharmacogenomics and Precision Medicine, Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL, United States
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Rhonda M. Cooper-DeHoff
- Center for Pharmacogenomics and Precision Medicine, Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL, United States
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Gavino Casu
- Azienda Ospedaliero Universitaria, Sassari, Italy
| | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
- Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
- Department of Medicine and Intensive Care, County Hospital Bregenz, Bregenz, Austria
| | - Benjamin D. Horne
- Intermountain Medical Center Heart Institute, Salt Lake City, UT, United States
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, United States
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Nicola Marziliano
- Medicine Laboratory Unit, ASST Rhodense (Rho-Milano), Lombardy, Italy
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences and Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic Ohio, Cleveland, OH, United States
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic Ohio, Cleveland, OH, United States
| | - Juha Sinisalo
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Mika Kähönen
- Finnish Cardiovascular Research Center - Tampere, Department of Clinical Physiology, Faculty of Medicine and Health Technology, Department of Clinical Physiology, Tampere University, Tampere, Finland
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories Ltd., Tampere, Finland
- Finnish Cardiovascular Research Center - Tampere, Department of Clinical Physiology, Faculty of Medicine and Health Technology, Department of Clinical Physiology, Tampere University, Tampere, Finland
| | - Chim C. Lang
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
- LIFE Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Markus Scholz
- LIFE Research Center for Civilization Diseases, Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
- Netherlands Heart Institute, Utrecht, Netherlands
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Axel Åkerblom
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Stefan James
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Claes Held
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Emil Hagström
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - John A. Spertus
- Saint Luke´s Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Ale Algra
- Department of Neurology and Neurosurgery, Brain Centre Rudolf Magnus and Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ulf de Faire
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Åkesson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Folkert W. Asselbergs
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Faculty of Population Health Sciences, Institute of Cardiovascular Science and Institute of Health Informatics, University College London, London, United Kingdom
| | - Riyaz S. Patel
- Faculty of Population Health Sciences, Institute of Cardiovascular Science and Institute of Health Informatics, University College London, London, United Kingdom
- Bart’s Heart Centre, St Bartholomew’s Hospital, London, United Kingdom
| | - Karin Leander
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- *Correspondence: Karin Leander,
| |
Collapse
|
42
|
Abstract
As a muscular pump that contracts incessantly throughout life, the heart must constantly generate cellular energy to support contractile function and fuel ionic pumps to maintain electrical homeostasis. Thus, mitochondrial metabolism of multiple metabolic substrates such as fatty acids, glucose, ketones, and lactate is essential to ensuring an uninterrupted supply of ATP. Multiple metabolic pathways converge to maintain myocardial energy homeostasis. The regulation of these cardiac metabolic pathways has been intensely studied for many decades. Rapid adaptation of these pathways is essential for mediating the myocardial adaptation to stress, and dysregulation of these pathways contributes to myocardial pathophysiology as occurs in heart failure and in metabolic disorders such as diabetes. The regulation of these pathways reflects the complex interactions of cell-specific regulatory pathways, neurohumoral signals, and changes in substrate availability in the circulation. Significant advances have been made in the ability to study metabolic regulation in the heart, and animal models have played a central role in contributing to this knowledge. This review will summarize metabolic pathways in the heart and describe their contribution to maintaining myocardial contractile function in health and disease. The review will summarize lessons learned from animal models with altered systemic metabolism and those in which specific metabolic regulatory pathways have been genetically altered within the heart. The relationship between intrinsic and extrinsic regulators of cardiac metabolism and the pathophysiology of heart failure and how these have been informed by animal models will be discussed.
Collapse
Affiliation(s)
- Heiko Bugger
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - Nikole J Byrne
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (E.D.A.)
| |
Collapse
|
43
|
Li A, Gao M, Liu B, Qin Y, Chen L, Liu H, Wu H, Gong G. Mitochondrial autophagy: molecular mechanisms and implications for cardiovascular disease. Cell Death Dis 2022; 13:444. [PMID: 35534453 PMCID: PMC9085840 DOI: 10.1038/s41419-022-04906-6] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/13/2022]
Abstract
Mitochondria are highly dynamic organelles that participate in ATP generation and involve calcium homeostasis, oxidative stress response, and apoptosis. Dysfunctional or damaged mitochondria could cause serious consequences even lead to cell death. Therefore, maintaining the homeostasis of mitochondria is critical for cellular functions. Mitophagy is a process of selectively degrading damaged mitochondria under mitochondrial toxicity conditions, which plays an essential role in mitochondrial quality control. The abnormal mitophagy that aggravates mitochondrial dysfunction is closely related to the pathogenesis of many diseases. As the myocardium is a highly oxidative metabolic tissue, mitochondria play a central role in maintaining optimal performance of the heart. Dysfunctional mitochondria accumulation is involved in the pathophysiology of cardiovascular diseases, such as myocardial infarction, cardiomyopathy and heart failure. This review discusses the most recent progress on mitophagy and its role in cardiovascular disease.
Collapse
Affiliation(s)
- Anqi Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Meng Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bilin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuan Qin
- Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Lei Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hanyu Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Huayan Wu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guohua Gong
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
44
|
AMPK Activation Is Indispensable for the Protective Effects of Caloric Restriction on Left Ventricular Function in Postinfarct Myocardium. BIOLOGY 2022; 11:biology11030448. [PMID: 35336822 PMCID: PMC8945456 DOI: 10.3390/biology11030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022]
Abstract
Background: Caloric restriction (CR) extends lifespan in many species, including mammals. CR is cardioprotective in senescent myocardium by correcting pre-existing mitochondrial dysfunction and apoptotic activation. Furthermore, it confers cardioprotection against acute ischemia-reperfusion injury. Here, we investigated the role of AMP-activated protein kinase (AMPK) in mediating the cardioprotective CR effects in failing, postinfarct myocardium. Methods: Ligation of the left coronary artery or sham operation was performed in rats and mice. Four weeks after surgery, left ventricular (LV) function was analyzed by echocardiography, and animals were assigned to different feeding groups (control diet or 40% CR, 8 weeks) as matched pairs. The role of AMPK was investigated with an AMPK inhibitor in rats or the use of alpha 2 AMPK knock-out mice. Results: CR resulted in a significant improvement in LV function, compared to postinfarct animals receiving control diet in both species. The improvement in LV function was accompanied by a reduction in serum BNP, decrease in LV proapoptotic activation, and increase in mitochondrial biogenesis in the LV. Inhibition or loss of AMPK prevented most of these changes. Conclusions: The failing, postischemic heart is protected from progressive loss of LV systolic function by CR. AMPK activation is indispensable for these protective effects.
Collapse
|
45
|
Maurice NM, Bedi B, Yuan Z, Lin KC, Goldberg JB, Hart CM, Bailey KL, Sadikot RT. The Effect of PGC-1alpha-SIRT3 Pathway Activation on Pseudomonas aeruginosa Infection. Pathogens 2022; 11:pathogens11020116. [PMID: 35215060 PMCID: PMC8875424 DOI: 10.3390/pathogens11020116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 02/01/2023] Open
Abstract
The innate immune response to P. aeruginosa pulmonary infections relies on a network of pattern recognition receptors, including intracellular inflammasome complexes, which can recognize both pathogen- and host-derived signals and subsequently promote downstream inflammatory signaling. Current evidence suggests that the inflammasome does not contribute to bacterial clearance and, in fact, that dysregulated inflammasome activation is harmful in acute and chronic P. aeruginosa lung infection. Given the role of mitochondrial damage signals in recruiting inflammasome signaling, we investigated whether mitochondrial-targeted therapies could attenuate inflammasome signaling in response to P. aeruginosa and decrease pathogenicity of infection. In particular, we investigated the small molecule, ZLN005, which transcriptionally activates peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a master regulator of mitochondrial biogenesis, antioxidant defense, and cellular respiration. We demonstrate that P. aeruginosa infection promotes the expression of inflammasome components and attenuates several components of mitochondrial repair pathways in vitro in lung epithelial cells and in vivo in an acute pneumonia model. ZLN005 activates PGC-1α and its downstream effector, Sirtuin 3 (SIRT3), a mitochondrial-localized deacetylase important for cellular metabolic processes and for reactive oxygen species homeostasis. ZLN005 also attenuates inflammasome signaling induced by P. aeruginosa in bronchial epithelial cells and this action is dependent on ZLN005 activation of SIRT3. ZLN005 treatment reduces epithelial-barrier dysfunction caused by P. aeruginosa and decreases pathogenicity in an in vivo pneumonia model. Therapies that activate the PGC-1α—SIRT3 axis may provide a complementary approach in the treatment of P. aeruginosa infection.
Collapse
Affiliation(s)
- Nicholas M. Maurice
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (N.M.M.); (B.B.); (K.-C.L.); (C.M.H.)
- Atlanta Veterans Affairs Health Care System, Decatur, GA 30033, USA
| | - Brahmchetna Bedi
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (N.M.M.); (B.B.); (K.-C.L.); (C.M.H.)
- Atlanta Veterans Affairs Health Care System, Decatur, GA 30033, USA
| | - Zhihong Yuan
- VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA; (Z.Y.); (K.L.B.)
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kuo-Chuan Lin
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (N.M.M.); (B.B.); (K.-C.L.); (C.M.H.)
- Atlanta Veterans Affairs Health Care System, Decatur, GA 30033, USA
| | - Joanna B. Goldberg
- Department of Pediatrics, Division of Pulmonology, Allergy/Immunology, Cystic Fibrosis, and Sleep, Emory University, Atlanta, GA 30322, USA;
- Children’s Healthcare of Atlanta, Center for CF and Airways Disease Research, Atlanta, GA 30322, USA
| | - C. Michael Hart
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; (N.M.M.); (B.B.); (K.-C.L.); (C.M.H.)
- Atlanta Veterans Affairs Health Care System, Decatur, GA 30033, USA
| | - Kristina L. Bailey
- VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA; (Z.Y.); (K.L.B.)
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ruxana T. Sadikot
- VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA; (Z.Y.); (K.L.B.)
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence:
| |
Collapse
|
46
|
Chaanine AH. Metabolic Remodeling and Implicated Calcium and Signal Transduction Pathways in the Pathogenesis of Heart Failure. Int J Mol Sci 2021; 22:ijms221910579. [PMID: 34638917 PMCID: PMC8508915 DOI: 10.3390/ijms221910579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/20/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
The heart is an organ with high-energy demands in which the mitochondria are most abundant. They are considered the powerhouse of the cell and occupy a central role in cellular metabolism. The intermyofibrillar mitochondria constitute the majority of the three-mitochondrial subpopulations in the heart. They are also considered to be the most important in terms of their ability to participate in calcium and cellular signaling, which are critical for the regulation of mitochondrial function and adenosine triphosphate (ATP) production. This is because they are located in very close proximity with the endoplasmic reticulum (ER), and for the presence of tethering complexes enabling interorganelle crosstalk via calcium signaling. Calcium is an important second messenger that regulates mitochondrial function. It promotes ATP production and cellular survival under physiological changes in cardiac energetic demand. This is accomplished in concert with signaling pathways that regulate both calcium cycling and mitochondrial function. Perturbations in mitochondrial homeostasis and metabolic remodeling occupy a central role in the pathogenesis of heart failure. In this review we will discuss perturbations in ER-mitochondrial crosstalk and touch on important signaling pathways and molecular mechanisms involved in the dysregulation of calcium homeostasis and mitochondrial function in heart failure.
Collapse
Affiliation(s)
- Antoine H. Chaanine
- Department of Medicine, Heart and Vascular Institute, Tulane University, New Orleans, LA 70112, USA; ; Tel.: +1-(504)-988-1612
- Department of Physiology, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
47
|
Dubinin MV, Starinets VS, Talanov EY, Mikheeva IB, Belosludtseva NV, Serov DA, Tenkov KS, Belosludtseva EV, Belosludtsev KN. Effect of the Non-Immunosuppressive MPT Pore Inhibitor Alisporivir on the Functioning of Heart Mitochondria in Dystrophin-Deficient mdx Mice. Biomedicines 2021; 9:1232. [PMID: 34572419 PMCID: PMC8466941 DOI: 10.3390/biomedicines9091232] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 01/05/2023] Open
Abstract
Supporting mitochondrial function is one of the therapeutic strategies that improve the functioning of skeletal muscle in Duchenne muscular dystrophy (DMD). In this work, we studied the effect of a non-immunosuppressive inhibitor of mitochondrial permeability transition pore (MPTP) alisporivir (5 mg/kg/day), reducing the intensity of the necrotic process and inflammation in skeletal muscles on the cardiac phenotype of dystrophin-deficient mdx mice. We found that the heart mitochondria of mdx mice show an increase in the intensity of oxidative phosphorylation and an increase in the resistance of organelles to the MPT pore opening. Alisporivir had no significant effect on the hyperfunctionalization of the heart mitochondria of mdx mice, and the state of the heart mitochondria of wild-type animals did not affect the dynamics of organelles but significantly suppressed mitochondrial biogenesis and reduced the amount of mtDNA in the heart muscle. Moreover, alisporivir suppressed mitochondrial biogenesis in the heart of wild-type mice. Alisporivir treatment resulted in a decrease in heart weight in mdx mice, which was associated with a significant modification of the transmission of excitation in the heart. The latter was also noted in the case of WT mice treated with alisporivir. The paper discusses the prospects for using alisporivir to correct the function of heart mitochondria in DMD.
Collapse
Affiliation(s)
- Mikhail V. Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (V.S.S.); (K.S.T.); (E.V.B.); (K.N.B.)
| | - Vlada S. Starinets
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (V.S.S.); (K.S.T.); (E.V.B.); (K.N.B.)
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (I.B.M.); (N.V.B.)
| | - Eugeny Yu. Talanov
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (I.B.M.); (N.V.B.)
| | - Irina B. Mikheeva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (I.B.M.); (N.V.B.)
| | - Natalia V. Belosludtseva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (I.B.M.); (N.V.B.)
| | - Dmitriy A. Serov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia;
| | - Kirill S. Tenkov
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (V.S.S.); (K.S.T.); (E.V.B.); (K.N.B.)
| | - Evgeniya V. Belosludtseva
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (V.S.S.); (K.S.T.); (E.V.B.); (K.N.B.)
| | - Konstantin N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (V.S.S.); (K.S.T.); (E.V.B.); (K.N.B.)
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (I.B.M.); (N.V.B.)
| |
Collapse
|
48
|
Role of Irisin in Myocardial Infarction, Heart Failure, and Cardiac Hypertrophy. Cells 2021; 10:cells10082103. [PMID: 34440871 PMCID: PMC8392379 DOI: 10.3390/cells10082103] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Irisin is a myokine derived from the cleavage of fibronectin type III domain-containing 5. Irisin regulates mitochondrial energy, glucose metabolism, fatty acid oxidation, and fat browning. Skeletal muscle and cardiomyocytes produce irisin and affect various cardiovascular functions. In the early phase of acute myocardial infarction, an increasing irisin level can reduce endothelial damage by inhibiting inflammation and oxidative stress. By contrast, higher levels of irisin in the later phase of myocardial infarction are associated with more cardiovascular events. During different stages of heart failure, irisin has various influences on mitochondrial dysfunction, oxidative stress, metabolic imbalance, energy expenditure, and heart failure prognosis. Irisin affects blood pressure and controls hypertension through modulating vasodilatation. Moreover, irisin can enhance vasoconstriction via the hypothalamus. Because of these dual effects of irisin on cardiovascular physiology, irisin can be a critical therapeutic target in cardiovascular diseases. This review focuses on the complex functions of irisin in myocardial ischemia, heart failure, and cardiac hypertrophy.
Collapse
|
49
|
Cheng N, Mo Q, Donelson J, Wang L, Breton G, Rodney GG, Wang J, Hirschi KD, Wehrens XHT, Nakata PA. Crucial Role of Mammalian Glutaredoxin 3 in Cardiac Energy Metabolism in Diet-induced Obese Mice Revealed by Transcriptome Analysis. Int J Biol Sci 2021; 17:2871-2883. [PMID: 34345213 PMCID: PMC8326124 DOI: 10.7150/ijbs.60263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/25/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is often associated with metabolic dysregulation and oxidative stress with the latter serving as a possible unifying link between obesity and cardiovascular complications. Glutaredoxins (Grxs) comprise one of the major antioxidant systems in the heart. Although Grx3 has been shown to act as an endogenous negative regulator of cardiac hypertrophy and heart failure, its metabolic impact on cardiac function in diet-induced obese (DIO) mice remains largely unknown. In the present study, analysis of Grx3 expression indicated that Grx3 protein levels, but not mRNA levels, were significantly increased in the hearts of DIO mice. Cardiac-specific Grx3 deletion (Grx3 CKO) mice were viable and grew indistinguishably from their littermates after being fed a high fat diet (HFD) for one month, starting at 2 months of age. After being fed with a HFD for 8 months (starting at 2 months of age); however, Grx3 CKO DIO mice displayed left ventricular systolic dysfunction with a significant decrease in ejection fraction and fractional shortening that was associated with heart failure. ROS production was significantly increased in Grx3 CKO DIO cardiomyocytes compared to control cells. Gene expression analysis revealed a significant decline in the level of transcripts corresponding to genes associated with processes such as fatty acid uptake, mitochondrial fatty acid transport and oxidation, and citrate cycle in Grx3 CKO DIO mice compared to DIO controls. In contrast, an increase in the level of transcripts corresponding to genes associated with glucose uptake and utilization were found in Grx3 CKO DIO mice compared to DIO controls. Taken together, these findings indicate that Grx3 may play a critical role in redox balance and as a metabolic switch in cardiomyocytes contributing to the development and progression of heart failure.
Collapse
Affiliation(s)
- Ninghui Cheng
- USDA/ARS Children Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Qianxing Mo
- Department of Biostatistics & Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jimmonique Donelson
- USDA/ARS Children Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Lingfei Wang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ghislain Breton
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - George G Rodney
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Cardiovascular Research Institute, and Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kendal D Hirschi
- USDA/ARS Children Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Xander H T Wehrens
- USDA/ARS Children Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Cardiovascular Research Institute, and Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Paul A Nakata
- USDA/ARS Children Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
50
|
Federico M, De la Fuente S, Palomeque J, Sheu SS. The role of mitochondria in metabolic disease: a special emphasis on heart dysfunction. J Physiol 2021; 599:3477-3493. [PMID: 33932959 PMCID: PMC8424986 DOI: 10.1113/jp279376] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/18/2021] [Indexed: 01/10/2023] Open
Abstract
Metabolic diseases (MetDs) embrace a series of pathologies characterized by abnormal body glucose usage. The known diseases included in this group are metabolic syndrome, prediabetes and diabetes mellitus types 1 and 2. All of them are chronic pathologies that present metabolic disturbances and are classified as multi-organ diseases. Cardiomyopathy has been extensively described in diabetic patients without overt macrovascular complications. The heart is severely damaged during the progression of the disease; in fact, diabetic cardiomyopathies are the main cause of death in MetDs. Insulin resistance, hyperglycaemia and increased free fatty acid metabolism promote cardiac damage through mitochondria. These organelles supply most of the energy that the heart needs to beat and to control essential cellular functions, including Ca2+ signalling modulation, reactive oxygen species production and apoptotic cell death regulation. Several aspects of common mitochondrial functions have been described as being altered in diabetic cardiomyopathies, including impaired energy metabolism, compromised mitochondrial dynamics, deficiencies in Ca2+ handling, increases in reactive oxygen species production, and a higher probability of mitochondrial permeability transition pore opening. Therefore, the mitochondrial role in MetD-mediated heart dysfunction has been studied extensively to identify potential therapeutic targets for improving cardiac performance. Herein we review the cardiac pathology in metabolic syndrome, prediabetes and diabetes mellitus, focusing on the role of mitochondrial dysfunctions.
Collapse
Affiliation(s)
- Marilen Federico
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Medicas, UNLP, La Plata, Argentina
| | - Sergio De la Fuente
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Medicas, UNLP, La Plata, Argentina
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, CABA, Argentina
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| |
Collapse
|