1
|
Park M, Cho S, Jeong D. Restoration of Sestrin 3 Expression Mitigates Cardiac Oxidative Damage in Ischemia-Reperfusion Injury Model. Antioxidants (Basel) 2025; 14:61. [PMID: 39857395 PMCID: PMC11763094 DOI: 10.3390/antiox14010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Cardiac ischemia-reperfusion injury (IRI) occurs when blood flow is restored to the myocardium after a period of ischemia, leading to oxidative stress and subsequent myocardial cell damage, primarily due to the accumulation of reactive oxygen species (ROS). In our previous research, we identified that miR-25 is significantly overexpressed in pressure overload-induced heart failure, and its inhibition improves cardiac function by restoring the expression of SERCA2a, a key protein involved in calcium regulation. In this study, we aimed to investigate the role of miR-25 in the context of ischemia-reperfusion injury. We found that miR-25 was markedly upregulated under hypoxic conditions in both in vitro and in vivo models. Through in silico analysis, we identified Sestrin3 (SESN3), an antioxidant protein known for its protective effects against oxidative stress, as a novel target of miR-25. Based on these findings, we hypothesized that inhibiting miR-25 would restore Sestrin3 expression, thereby reducing ROS-induced myocardial cell damage and improving cardiac function. To test this hypothesis, we employed two model systems: a hypoxia/reoxygenation (H/R) stress model using H9c2 myoblasts and a surgically induced ischemia-reperfusion injury mouse model. Our results demonstrated that the use of miR-25 inhibitors significantly improved cardiac function and reduced myocardial damage in both models through the restoration of SESN3 expression. In conclusion, our findings suggest that targeting miR-25 may serve as a novel therapeutic modality to alleviate oxidative damage in the heart.
Collapse
Affiliation(s)
| | | | - Dongtak Jeong
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University—ERICA, Ansan 15588, Republic of Korea; (M.P.); (S.C.)
| |
Collapse
|
2
|
Zhang XH, Morad M. Regulation of SR and mitochondrial Ca 2+ signaling by L-type Ca 2+ channels and Na/Ca exchanger in hiPSC-CMs. Cell Calcium 2025; 125:102985. [PMID: 39693912 DOI: 10.1016/j.ceca.2024.102985] [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: 09/24/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024]
Abstract
RATIONALE & METHODS While signaling of cardiac SR by surface membrane proteins (ICa & INCX) is well studied, the regulation of mitochondrial Ca2+ by plasmalemmal proteins remains less explored. Here we have examined the signaling of mitochondria and SR by surface-membrane calcium-transporting proteins, using genetically engineered targeted fluorescent probes, mito-GCamP6 and R-CEPIA1er. RESULTS In voltage-clamped and TIRF-imaged cardiomyocytes, low Na+ induced SR Ca2+ release was suppressed by short pre-exposures to ∼100 nM FCCP, suggesting mitochondrial Ca2+ contribution to low Na+ triggered SR Ca2+release. Even though low Na+- or caffeine-triggered SR Ca2+ release activated global mitochondrial Ca2+ uptake, focal mitochondrial Ca2+ signals varied in kinetics and magnitude, showing uptake or release of calcium, depending on cellular location of mitochondria. In spontaneously pacing cells, sustained caffeine exposures depleted the SR Ca2+ content activating mitochondrial Ca2+ uptake followed by sustained mitochondrial pacing. Spontaneous hiPSCCMs pacing was strongly suppressed by L-type calcium channels blockers, but not by inhibiting SERCA2a by CPA. CONCLUSION Spontaneous hiPSCCMs pacing is triggered by influx of calcium through L-type Ca2+ channel that gates the release of SR pools supplemented by NCX-mediated mitochondrial calcium contribution.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- Cardiac Signaling Center of USC, MUSC and Clemson University, 68 President St BEB 306, Charleston, SC 29425, USA
| | - Martin Morad
- Cardiac Signaling Center of USC, MUSC and Clemson University, 68 President St BEB 306, Charleston, SC 29425, USA.
| |
Collapse
|
3
|
Sharma G, Chaurasia SS, Carlson MA, Mishra PK. Recent advances associated with cardiometabolic remodeling in diabetes-induced heart failure. Am J Physiol Heart Circ Physiol 2024; 327:H1327-H1342. [PMID: 39453429 DOI: 10.1152/ajpheart.00539.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
Diabetes mellitus (DM) is characterized by chronic hyperglycemia, and despite intensive glycemic control, the risk of heart failure in patients with diabetes remains high. Diabetes-induced heart failure (DHF) presents a unique metabolic challenge, driven by significant alterations in cardiac substrate metabolism, including increased reliance on fatty acid oxidation, reduced glucose utilization, and impaired mitochondrial function. These metabolic alterations lead to oxidative stress, lipotoxicity, and energy deficits, contributing to the progression of heart failure. Emerging research has identified novel mechanisms involved in the metabolic remodeling of diabetic hearts, such as autophagy dysregulation, epigenetic modifications, polyamine regulation, and branched-chain amino acid (BCAA) metabolism. These processes exacerbate mitochondrial dysfunction and metabolic inflexibility, further impairing cardiac function. Therapeutic interventions targeting these pathways-such as enhancing glucose oxidation, modulating fatty acid metabolism, and optimizing ketone body utilization-show promise in restoring metabolic homeostasis and improving cardiac outcomes. This review explores the key molecular mechanisms driving metabolic remodeling in diabetic hearts, highlights advanced methodologies, and presents the latest therapeutic strategies for mitigating the progression of DHF. Understanding these emerging pathways offers new opportunities to develop targeted therapies that address the root metabolic causes of heart failure in diabetes.
Collapse
Affiliation(s)
- Gaurav Sharma
- Department of Cardiovascular and Thoracic Surgery, UT Southwestern Medical Center, Dallas, Texas, United States
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, Texas, United States
- Department of Biomedical Engineering, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Shyam S Chaurasia
- Ocular Immunology and Angiogenesis Lab, Department Ophthalmology & Visual Sciences, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Mark A Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| |
Collapse
|
4
|
Kavalenia TA, Lapshina EA, Ilyich TV, Zhao HC, Zavodnik IB. Functional activity and morphology of isolated rat cardiac mitochondria under calcium overload. Effect of naringin. Mol Cell Biochem 2024; 479:3329-3340. [PMID: 38332449 DOI: 10.1007/s11010-024-04935-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: 07/19/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
The function of mitochondria as a regulator of myocyte calcium homeostasis has been extensively discussed. The aim of the present work was further clarification of the details of modulation of the functional activity of rat cardiac mitochondria by exogenous Ca2+ ions either in the absence or in the presence of the plant flavonoid naringin. Low free Ca2+ concentrations (40-250 nM) effectively inhibited the respiratory activity of heart mitochondria, remaining unaffected the efficacy of oxygen consumption. In the presence of high exogenous Ca2+ ion concentrations (Ca2+ free was 550 µM), we observed a dramatic increase in mitochondrial heterogeneity in size and electron density, which was related to calcium-induced opening of the mitochondrial permeability transition pores (MPTP) and membrane depolarization (Ca2+free ions were from 150 to 750 µM). Naringin partially prevented Ca2+-induced cardiac mitochondrial morphological transformations (200 µM) and dose-dependently inhibited the respiratory activity of mitochondria (10-75 µM) in the absence or in the presence of calcium ions. Our data suggest that naringin (75 µM) promoted membrane potential dissipation, diminishing the potential-dependent accumulation of calcium ions by mitochondria and inhibiting calcium-induced MPTP formation. The modulating effect of the flavonoid on Ca2+-induced mitochondria alterations may be attributed to the weak-acidic nature of the flavonoid and its protonophoric/ionophoric properties. Our results show that the sensitivity of rat heart mitochondria to Ca2+ ions was much lower in the case of MPTP opening and much higher in the case of respiration inhibition as compared to liver mitochondria.
Collapse
Affiliation(s)
- T A Kavalenia
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola, 5, 230009, Grodno, Belarus
| | - E A Lapshina
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola, 5, 230009, Grodno, Belarus
| | - T V Ilyich
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola, 5, 230009, Grodno, Belarus
| | - Hu-Cheng Zhao
- Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - I B Zavodnik
- Department of Biochemistry, Yanka Kupala State University of Grodno, Bulvar Leninskogo Komsomola, 5, 230009, Grodno, Belarus.
| |
Collapse
|
5
|
Martinez CS, Zheng A, Xiao Q. Mitochondrial Reactive Oxygen Species Dysregulation in Heart Failure with Preserved Ejection Fraction: A Fraction of the Whole. Antioxidants (Basel) 2024; 13:1330. [PMID: 39594472 PMCID: PMC11591317 DOI: 10.3390/antiox13111330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a multifarious syndrome, accounting for over half of heart failure (HF) patients receiving clinical treatment. The prevalence of HFpEF is rapidly increasing in the coming decades as the global population ages. It is becoming clearer that HFpEF has a lot of different causes, which makes it challenging to find effective treatments. Currently, there are no proven treatments for people with deteriorating HF or HFpEF. Although the pathophysiologic foundations of HFpEF are complex, excessive reactive oxygen species (ROS) generation and increased oxidative stress caused by mitochondrial dysfunction seem to play a critical role in the pathogenesis of HFpEF. Emerging evidence from animal models and human myocardial tissues from failed hearts shows that mitochondrial aberrations cause a marked increase in mitochondrial ROS (mtROS) production and oxidative stress. Furthermore, studies have reported that common HF medications like beta blockers, angiotensin receptor blockers, angiotensin-converting enzyme inhibitors, and mineralocorticoid receptor antagonists indirectly reduce the production of mtROS. Despite the harmful effects of ROS on cardiac remodeling, maintaining mitochondrial homeostasis and cardiac functions requires small amounts of ROS. In this review, we will provide an overview and discussion of the recent findings on mtROS production, its threshold for imbalance, and the subsequent dysfunction that leads to related cardiac and systemic phenotypes in the context of HFpEF. We will also focus on newly discovered cellular and molecular mechanisms underlying ROS dysregulation, current therapeutic options, and future perspectives for treating HFpEF by targeting mtROS and the associated signal molecules.
Collapse
Affiliation(s)
| | | | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (C.S.M.); (A.Z.)
| |
Collapse
|
6
|
Ravindran S, Rau CD. The multifaceted role of mitochondria in cardiac function: insights and approaches. Cell Commun Signal 2024; 22:525. [PMID: 39472951 PMCID: PMC11523909 DOI: 10.1186/s12964-024-01899-x] [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: 03/22/2024] [Accepted: 10/19/2024] [Indexed: 11/02/2024] Open
Abstract
Cardiovascular disease (CVD) remains a global economic burden even in the 21st century with 85% of deaths resulting from heart attacks. Despite efforts in reducing the risk factors, and enhancing pharmacotherapeutic strategies, challenges persist in early identification of disease progression and functional recovery of damaged hearts. Targeting mitochondrial dysfunction, a key player in the pathogenesis of CVD has been less successful due to its role in other coexisting diseases. Additionally, it is the only organelle with an agathokakological function that is a remedy and a poison for the cell. In this review, we describe the origins of cardiac mitochondria and the role of heteroplasmy and mitochondrial subpopulations namely the interfibrillar, subsarcolemmal, perinuclear, and intranuclear mitochondria in maintaining cardiac function and in disease-associated remodeling. The cumulative evidence of mitochondrial retrograde communication with the nucleus is addressed, highlighting the need to study the genotype-phenotype relationships of specific organelle functions with CVD by using approaches like genome-wide association study (GWAS). Finally, we discuss the practicality of computational methods combined with single-cell sequencing technologies to address the challenges of genetic screening in the identification of heteroplasmy and contributory genes towards CVD.
Collapse
Affiliation(s)
- Sriram Ravindran
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA
| | - Christoph D Rau
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA.
| |
Collapse
|
7
|
Guo Z, Tian Y, Gao J, Zhou B, Zhou X, Chang X, Zhou H. Enhancement of Mitochondrial Homeostasis: A Novel Approach to Attenuate Hypoxic Myocardial Injury. Int J Med Sci 2024; 21:2897-2911. [PMID: 39628681 PMCID: PMC11610329 DOI: 10.7150/ijms.103986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/18/2024] [Indexed: 12/06/2024] Open
Abstract
Cardiomyocytes are highly oxygen-dependent cells, relying on oxygen-driven oxidative phosphorylation to maintain their function. During hypoxia, mitochondrial ATP production decreases, leading to calcium overload, acidosis, and oxidative stress, which collectively trigger myocardial injury. Ischemic heart disease, caused by coronary atherosclerosis, results in myocardial ischemia and hypoxia, leading to ischemia-reperfusion (I/R) injury. Early myocardial injury is attributed to ischemia and hypoxia, but even after thrombolytic therapy, interventional surgery, or coronary artery bypass grafting (CABG) restores local blood flow and oxygen supply, myocardial reperfusion injury (I/R) may still occur. Mitochondria, often referred to as the "powerhouses" of the cell, play a crucial role in cellular energy production. In the early stages of ischemia and hypoxia, mitochondrial dysfunction disrupts mitochondrial homeostasis, causing the accumulation of unfolded or misfolded proteins in the mitochondria. This activates the mitochondrial unfolded protein response (mtUPR) and mitophagy, which work to clear damaged proteins and mitochondria, playing a key role during this period. This review focuses on mitochondrial mechanisms during the ischemic phase of ischemia-reperfusion injury, aiming to provide new theoretical foundations and potential therapeutic strategies to reduce myocardial damage.
Collapse
Affiliation(s)
- Zhijiang Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yingjie Tian
- Beijing University of Chinese Medicine, Beijing, 100028, China
| | - Jing Gao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Bei Zhou
- Center for Drug Evaluation, National Medical Products Administration, Beijing, China
| | - XiuTeng Zhou
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences/State Key Laboratory for Quality Assurance and Sustainable Use of Dao-di Herbs, Beijing, 100700, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
8
|
Lees JG, Greening DW, Rudd DA, Cross J, Rosdah AA, Lai X, Lin TW, Phang RJ, Kong AM, Deng Y, Crawford S, Holien JK, Hausenloy DJ, Shen HH, Lim SY. Cardiac-targeted delivery of a novel Drp1 inhibitor for acute cardioprotection. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 9:100085. [PMID: 39803589 PMCID: PMC11708310 DOI: 10.1016/j.jmccpl.2024.100085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 01/16/2025]
Abstract
Dynamin-related protein 1 (Drp1) is a mitochondrial fission protein and a viable target for cardioprotection against myocardial ischaemia-reperfusion injury. Here, we reported a novel Drp1 inhibitor (DRP1i1), delivered using a cardiac-targeted nanoparticle drug delivery system, as a more effective approach for achieving acute cardioprotection. DRP1i1 was encapsulated in cubosome nanoparticles with conjugated cardiac-homing peptides (NanoDRP1i1) and the encapsulation efficiency was 99.3 ± 0.1 %. In vivo, following acute myocardial ischaemia-reperfusion injury in mice, NanoDRP1i1 significantly reduced infarct size and serine-616 phosphorylation of Drp1, and restored cardiomyocyte mitochondrial size to that of sham group. Imaging by mass spectrometry revealed higher accumulation of DRP1i1 in the heart tissue when delivered as NanoDRP1i1. In human cardiac organoids subjected to simulated ischaemia-reperfusion injury, treatment with NanoDRP1i1 at reperfusion significantly reduced cardiac cell death, contractile dysfunction, and mitochondrial superoxide levels. Following NanoDRP1i1 treatment, cardiac organoid proteomics further confirmed reprogramming of contractile dysfunction markers and enrichment of the mitochondrial protein network, cytoskeletal and metabolic regulation networks when compared to the simulated injury group. These proteins included known cardioprotective regulators identified in human organoids and in vivo murine studies following ischaemia-reperfusion injury. DRP1i1 is a promising tool compound to study Drp1-mediated mitochondrial fission and exhibits promising therapeutic potential for acute cardioprotection, especially when delivered using the cardiac-targeted cubosome nanoparticles.
Collapse
Affiliation(s)
- Jarmon G. Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Victoria 3065, Australia
- Department of Medicine and Surgery, University of Melbourne, VIC, Australia
| | - David W. Greening
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria 3086, Australia
- Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
- Baker department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - David A. Rudd
- Monash Institute of Pharmaceutical Sciences, Monash University Parkville, Victoria 3052, Australia
| | - Jonathon Cross
- Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Ayeshah A. Rosdah
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Victoria 3065, Australia
- Department of Medicine and Surgery, University of Melbourne, VIC, Australia
- Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia
| | - Xiangfeng Lai
- Department of Materials Science and Engineering, Faulty of Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Tsung Wu Lin
- Department of Materials Science and Engineering, Faulty of Engineering, Monash University, Clayton, Victoria 3800, Australia
- Department of Chemistry, Tunghai University, No.1727, Sec.4, Taiwan Boulevard, Xitun District, Taichung 40704, Taiwan
| | - Ren Jie Phang
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Victoria 3065, Australia
| | - Anne M. Kong
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Victoria 3065, Australia
| | - Yali Deng
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Victoria 3065, Australia
- Department of Medicine and Surgery, University of Melbourne, VIC, Australia
| | - Simon Crawford
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Victoria 3800, Australia
| | - Jessica K. Holien
- Department of Medicine and Surgery, University of Melbourne, VIC, Australia
- School of Science, STEM College, Engineering and Health, RMIT University, Melbourne, Victoria, Australia
| | - Derek J. Hausenloy
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 67 Chenies Mews, WC1E 6HX London, UK
- Cardiovascular and Metabolic Disorder Programme, Duke-NUS Medical School, 8 College Road, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, 5 Hospital Drive, 169609, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, 119228, Singapore
| | - Hsin-Hui Shen
- Department of Materials Science and Engineering, Faulty of Engineering, Monash University, Clayton, Victoria 3800, Australia
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Shiang Y. Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Victoria 3065, Australia
- Department of Medicine and Surgery, University of Melbourne, VIC, Australia
- National Heart Research Institute Singapore, National Heart Centre, 5 Hospital Drive, 169609, Singapore
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Victoria, Australia
| |
Collapse
|
9
|
Hinton A, Claypool SM, Neikirk K, Senoo N, Wanjalla CN, Kirabo A, Williams CR. Mitochondrial Structure and Function in Human Heart Failure. Circ Res 2024; 135:372-396. [PMID: 38963864 PMCID: PMC11225798 DOI: 10.1161/circresaha.124.323800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Despite clinical and scientific advancements, heart failure is the major cause of morbidity and mortality worldwide. Both mitochondrial dysfunction and inflammation contribute to the development and progression of heart failure. Although inflammation is crucial to reparative healing following acute cardiomyocyte injury, chronic inflammation damages the heart, impairs function, and decreases cardiac output. Mitochondria, which comprise one third of cardiomyocyte volume, may prove a potential therapeutic target for heart failure. Known primarily for energy production, mitochondria are also involved in other processes including calcium homeostasis and the regulation of cellular apoptosis. Mitochondrial function is closely related to morphology, which alters through mitochondrial dynamics, thus ensuring that the energy needs of the cell are met. However, in heart failure, changes in substrate use lead to mitochondrial dysfunction and impaired myocyte function. This review discusses mitochondrial and cristae dynamics, including the role of the mitochondria contact site and cristae organizing system complex in mitochondrial ultrastructure changes. Additionally, this review covers the role of mitochondria-endoplasmic reticulum contact sites, mitochondrial communication via nanotunnels, and altered metabolite production during heart failure. We highlight these often-neglected factors and promising clinical mitochondrial targets for heart failure.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Steven M. Claypool
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Nanami Senoo
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation (A.K.)
- Vanderbilt Institute for Global Health (A.K.)
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH (C.R.W.)
| |
Collapse
|
10
|
Nakamura E, Aoki T, Endo Y, Kazmi J, Hagiwara J, Kuschner CE, Yin T, Kim J, Becker LB, Hayashida K. Organ-Specific Mitochondrial Alterations Following Ischemia-Reperfusion Injury in Post-Cardiac Arrest Syndrome: A Comprehensive Review. Life (Basel) 2024; 14:477. [PMID: 38672748 PMCID: PMC11050834 DOI: 10.3390/life14040477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Mitochondrial dysfunction, which is triggered by systemic ischemia-reperfusion (IR) injury and affects various organs, is a key factor in the development of post-cardiac arrest syndrome (PCAS). Current research on PCAS primarily addresses generalized mitochondrial responses, resulting in a knowledge gap regarding organ-specific mitochondrial dynamics. This review focuses on the organ-specific mitochondrial responses to IR injury, particularly examining the brain, heart, and kidneys, to highlight potential therapeutic strategies targeting mitochondrial dysfunction to enhance outcomes post-IR injury. METHODS AND RESULTS We conducted a narrative review examining recent advancements in mitochondrial research related to IR injury. Mitochondrial responses to IR injury exhibit considerable variation across different organ systems, influenced by unique mitochondrial structures, bioenergetics, and antioxidative capacities. Each organ demonstrates distinct mitochondrial behaviors that have evolved to fulfill specific metabolic and functional needs. For example, cerebral mitochondria display dynamic responses that can be both protective and detrimental to neuronal activity and function during ischemic events. Cardiac mitochondria show vulnerability to IR-induced oxidative stress, while renal mitochondria exhibit a unique pattern of fission and fusion, closely linked to their susceptibility to acute kidney injury. This organ-specific heterogeneity in mitochondrial responses requires the development of tailored interventions. Progress in mitochondrial medicine, especially in the realms of genomics and metabolomics, is paving the way for innovative strategies to combat mitochondrial dysfunction. Emerging techniques such as mitochondrial transplantation hold the potential to revolutionize the management of IR injury in resuscitation science. CONCLUSIONS The investigation into organ-specific mitochondrial responses to IR injury is pivotal in the realm of resuscitation research, particularly within the context of PCAS. This nuanced understanding holds the promise of revolutionizing PCAS management, addressing the unique mitochondrial dysfunctions observed in critical organs affected by IR injury.
Collapse
Affiliation(s)
- Eriko Nakamura
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Tomoaki Aoki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Yusuke Endo
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Jacob Kazmi
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Jun Hagiwara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Cyrus E. Kuschner
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Tai Yin
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Junhwan Kim
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
| | - Lance B. Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY 11030, USA; (E.N.); (T.A.); (Y.E.); (J.K.); (J.H.); (C.E.K.); (T.Y.); (J.K.); (L.B.B.)
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
11
|
Somers T, Siddiqi S, Maas RGC, Sluijter JPG, Buikema JW, van den Broek PHH, Meuwissen TJ, Morshuis WJ, Russel FGM, Schirris TJJ. Statins affect human iPSC-derived cardiomyocytes by interfering with mitochondrial function and intracellular acidification. Basic Res Cardiol 2024; 119:309-327. [PMID: 38305903 DOI: 10.1007/s00395-023-01025-x] [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: 02/06/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 02/03/2024]
Abstract
Statins are effective drugs in reducing cardiovascular morbidity and mortality by inhibiting cholesterol synthesis. These effects are primarily beneficial for the patient's vascular system. A significant number of statin users suffer from muscle complaints probably due to mitochondrial dysfunction, a mechanism that has recently been elucidated. This has raised our interest in exploring the effects of statins on cardiac muscle cells in an era where the elderly and patients with poorer functioning hearts and less metabolic spare capacity start dominating our patient population. Here, we investigated the effects of statins on human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-derived CMs). hiPSC-derived CMs were exposed to simvastatin, atorvastatin, rosuvastatin, and cerivastatin at increasing concentrations. Metabolic assays and fluorescent microscopy were employed to evaluate cellular viability, metabolic capacity, respiration, intracellular acidity, and mitochondrial membrane potential and morphology. Over a concentration range of 0.3-100 µM, simvastatin lactone and atorvastatin acid showed a significant reduction in cellular viability by 42-64%. Simvastatin lactone was the most potent inhibitor of basal and maximal respiration by 56% and 73%, respectively, whereas simvastatin acid and cerivastatin acid only reduced maximal respiration by 50% and 42%, respectively. Simvastatin acid and lactone and atorvastatin acid significantly decreased mitochondrial membrane potential by 20%, 6% and 3%, respectively. The more hydrophilic atorvastatin acid did not seem to affect cardiomyocyte metabolism. This calls for further research on the translatability to the clinical setting, in which a more conscientious approach to statin prescribing might be considered, especially regarding the current shift in population toward older patients with poor cardiac function.
Collapse
Affiliation(s)
- Tim Somers
- Department of Cardiothoracic Surgery, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Sailay Siddiqi
- Department of Cardiothoracic Surgery, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Renee G C Maas
- Department of Cardiology, Experimental Cardiology Laboratory, Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Jan W Buikema
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam Heart Center, Amsterdam University Medical Center, De Boelelaan 1117, 1081 HZ, Amsterdam, The Netherlands
| | - Petra H H van den Broek
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Tanne J Meuwissen
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Wim J Morshuis
- Department of Cardiothoracic Surgery, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Frans G M Russel
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.
| | - Tom J J Schirris
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Dos Santos B, Bion MC, Goujon-Svrzic M, Maher P, Dafre AL. REAP+: A single preparation for rapid isolation of nuclei, cytoplasm, and mitochondria. Anal Biochem 2024; 687:115445. [PMID: 38135241 PMCID: PMC10843687 DOI: 10.1016/j.ab.2023.115445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/03/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
REAP+ is an enhanced version of the rapid, efficient, and practical (REAP) method designed for the isolation of nuclear fractions. This improved version, REAP+, enables fast and effective extraction of mitochondria, cytoplasm, and nuclei. The mechanical cell disruption process has been optimized to cerebral tissues, snap-frozen liver, and HT22 cells with remarkable fraction enrichment. REAP+ is well-suited for samples containing minimal protein quantities, such as mouse hippocampal slices. The method was validated by Western blot and marker enzyme activities, such as LDH and G6PDH for the cytoplasmic fraction and succinate dehydrogenase and cytochrome c oxidase for the mitochondrial fraction. One of the outstanding features of this method is its rapid execution, yielding fractions within 15 min, allowing for simultaneous preparation of multiple samples. In essence, REAP+ emerges as a swift, efficient, and practical technique for the concurrent isolation of nuclei, cytoplasm, and mitochondria from various cell types and tissues. The method would be suitable to study the multicompartment translocation of proteins, such as metabolic enzymes and transcription factors migrating from cytosol to the mitochondria and nuclei. Moreover, its compatibility with small samples, such as hippocampal slices, and its potential applicability to human biopsies, highlights the potential application in medical research.
Collapse
Affiliation(s)
- Barbara Dos Santos
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil; Post-Graduation Program in Biochemistry, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil.
| | - Monique Coelho Bion
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil; Post-Graduation Program in Cell Biology and Development, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil.
| | - Marie Goujon-Svrzic
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, CA, 92037, La Jolla, United States.
| | - Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, CA, 92037, La Jolla, United States.
| | - Alcir Luiz Dafre
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil.
| |
Collapse
|
13
|
Heyne E, Zeeb S, Junker C, Petzinna A, Schrepper A, Doenst T, Koch LG, Britton SL, Schwarzer M. Exercise Training Differentially Affects Skeletal Muscle Mitochondria in Rats with Inherited High or Low Exercise Capacity. Cells 2024; 13:393. [PMID: 38474357 PMCID: PMC10931189 DOI: 10.3390/cells13050393] [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: 12/31/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Exercise capacity has been related to morbidity and mortality. It consists of an inherited and an acquired part and is dependent on mitochondrial function. We assessed skeletal muscle mitochondrial function in rats with divergent inherited exercise capacity and analyzed the effect of exercise training. Female high (HCR)- and low (LCR)-capacity runners were trained with individually adapted high-intensity intervals or kept sedentary. Interfibrillar (IFM) and subsarcolemmal (SSM) mitochondria from gastrocnemius muscle were isolated and functionally assessed (age: 15 weeks). Sedentary HCR presented with higher exercise capacity than LCR paralleled by higher citrate synthase activity and IFM respiratory capacity in skeletal muscle of HCR. Exercise training increased exercise capacity in both HCR and LCR, but this was more pronounced in LCR. In addition, exercise increased skeletal muscle mitochondrial mass more in LCR. Instead, maximal respiratory capacity was increased following exercise in HCRs' IFM only. The results suggest that differences in skeletal muscle mitochondrial subpopulations are mainly inherited. Exercise training resulted in different mitochondrial adaptations and in higher trainability of LCR. HCR primarily increased skeletal muscle mitochondrial quality while LCR increased mitochondrial quantity in response to exercise training, suggesting that inherited aerobic exercise capacity differentially affects the mitochondrial response to exercise training.
Collapse
Affiliation(s)
- Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Susanne Zeeb
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Celina Junker
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Andreas Petzinna
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University Toledo, Toledo, OH 43606, USA;
| | - Steven L. Britton
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| | - Michael Schwarzer
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany; (E.H.); (A.P.); (A.S.); (T.D.)
| |
Collapse
|
14
|
Favero G, Golic I, Arnaboldi F, Cappella A, Korac A, Monsalve M, Stacchiotti A, Rezzani R. Cardiometabolic Changes in Sirtuin1-Heterozygous Mice on High-Fat Diet and Melatonin Supplementation. Int J Mol Sci 2024; 25:860. [PMID: 38255934 PMCID: PMC10815439 DOI: 10.3390/ijms25020860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
A hypercaloric fatty diet predisposes an individual to metabolic syndrome and cardiovascular complications. Sirtuin1 (SIRT1) belongs to the class III histone deacetylase family and sustains anabolism, mitochondrial biogenesis, and fat distribution. Epididymal white adipose tissue (eWAT) is involved in inflammation, whilst interscapular brown adipose tissue (iBAT) drives metabolism in obese rodents. Melatonin, a pineal indoleamine, acting as a SIRT1 modulator, may alleviate cardiometabolic damage. In the present study, we morphologically characterized the heart, eWAT, and iBAT in male heterozygous SIRT1+/- mice (HET mice) on a high-fat diet (60%E lard) versus a standard rodent diet (8.5% E fat) and drinking melatonin (10 mg/kg) for 16 weeks. Wild-type (WT) male C57Bl6/J mice were similarly fed for comparison. Cardiomyocyte fibrosis and endoplasmic reticulum (ER) stress response worsened in HET mice on a high-fat diet vs. other groups. Lipid peroxidation, ER, and mitochondrial stress were assessed by 4 hydroxy-2-nonenal (4HNE), glucose-regulated protein78 (GRP78), CCAA/enhancer-binding protein homologous protein (CHOP), heat shock protein 60 (HSP60), and mitofusin2 immunostainings. Ultrastructural analysis indicated the prevalence of atypical inter-myofibrillar mitochondria with short, misaligned cristae in HET mice on a lard diet despite melatonin supplementation. Abnormal eWAT adipocytes, crown-like inflammatory structures, tumor necrosis factor alpha (TNFα), and iBAT whitening characterized HET mice on a hypercaloric fatty diet and were maintained after melatonin supply. All these data suggest that melatonin's mechanism of action is strictly linked to full SIRT1 expression, which is required for the exhibition of effective antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (R.R.)
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Igor Golic
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (I.G.); (A.K.)
| | - Francesca Arnaboldi
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (F.A.); (A.C.)
| | - Annalisa Cappella
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (F.A.); (A.C.)
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Aleksandra Korac
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (I.G.); (A.K.)
| | - Maria Monsalve
- Instituto de Investigaciones Biomedicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain;
| | - Alessandra Stacchiotti
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (F.A.); (A.C.)
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (R.R.)
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale—SISDO), 25123 Brescia, Italy
| |
Collapse
|
15
|
Hernandez-Resendiz S, Prakash A, Loo SJ, Semenzato M, Chinda K, Crespo-Avilan GE, Dam LC, Lu S, Scorrano L, Hausenloy DJ. Targeting mitochondrial shape: at the heart of cardioprotection. Basic Res Cardiol 2023; 118:49. [PMID: 37955687 PMCID: PMC10643419 DOI: 10.1007/s00395-023-01019-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023]
Abstract
There remains an unmet need to identify novel therapeutic strategies capable of protecting the myocardium against the detrimental effects of acute ischemia-reperfusion injury (IRI), to reduce myocardial infarct (MI) size and prevent the onset of heart failure (HF) following acute myocardial infarction (AMI). In this regard, perturbations in mitochondrial morphology with an imbalance in mitochondrial fusion and fission can disrupt mitochondrial metabolism, calcium homeostasis, and reactive oxygen species production, factors which are all known to be critical determinants of cardiomyocyte death following acute myocardial IRI. As such, therapeutic approaches directed at preserving the morphology and functionality of mitochondria may provide an important strategy for cardioprotection. In this article, we provide an overview of the alterations in mitochondrial morphology which occur in response to acute myocardial IRI, and highlight the emerging therapeutic strategies for targeting mitochondrial shape to preserve mitochondrial function which have the future therapeutic potential to improve health outcomes in patients presenting with AMI.
Collapse
Affiliation(s)
- Sauri Hernandez-Resendiz
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Programme, Singapore, Singapore
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| | - Aishwarya Prakash
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Programme, Singapore, Singapore
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| | - Sze Jie Loo
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Programme, Singapore, Singapore
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| | | | - Kroekkiat Chinda
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Gustavo E Crespo-Avilan
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Programme, Singapore, Singapore
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| | - Linh Chi Dam
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Programme, Singapore, Singapore
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| | - Shengjie Lu
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Programme, Singapore, Singapore
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| | - Luca Scorrano
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Biology, University of Padova, Padova, Italy
| | - Derek J Hausenloy
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Programme, Singapore, Singapore.
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore.
- National University Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore.
- University College London, The Hatter Cardiovascular Institute, London, UK.
| |
Collapse
|
16
|
Du J, Yu D, Li J, Si L, Zhu D, Li B, Gao Y, Sun L, Wang X, Wang X. Asiatic acid protects against pressure overload-induced heart failure in mice by inhibiting mitochondria-dependent apoptosis. Free Radic Biol Med 2023; 208:545-554. [PMID: 37717794 DOI: 10.1016/j.freeradbiomed.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Mitochondrial dysfunction and subsequent cardiomyocyte apoptosis significantly contribute to pressure overload-induced heart failure (HF). A highly oxidative environment leads to mitochondrial damage, further exacerbating this condition. Asiatic acid (AA), a proven antioxidant and anti-hypertrophic agent, might provide a solution, but its role and mechanisms in chronic pressure overload-induced HF remain largely unexplored. METHODS We induced pressure overload in mice using transverse aortic constriction (TAC) and treated them with AA (100 mg/kg/day) or vehicle daily by oral gavage for 8 weeks. The effects of AA on mitochondrial dysfunction, oxidative stress-associated signaling pathways, and overall survival were evaluated. Additionally, an in vitro model using hydrogen peroxide-exposed neonatal rat cardiomyocytes was established to further investigate the role of AA in oxidative stress-induced mitochondrial apoptosis. RESULTS AA treatment significantly improved survival and alleviated cardiac dysfunction in TAC-induced HF mice. It preserved mitochondrial structure, reduced the LVW/BW ratio by 20.24%, mitigated TAC-induced mitochondrial-dependent apoptosis by significantly lowering the Bax/Bcl-2 ratio and cleaved caspase-9/3 levels, and attenuated oxidative stress. AA treatment protected cardiomyocytes from hydrogen peroxide-induced apoptosis, with concurrent modulation of mitochondrial-dependent apoptosis pathway-related proteins and the JNK pathway. CONCLUSIONS Our findings suggest that AA effectively combats chronic TAC-induced and hydrogen peroxide-induced cardiomyocyte apoptosis through a mitochondria-dependent mechanism. AA reduces cellular levels of oxidative stress and inhibits the activation of the JNK pathway, highlighting its potential therapeutic value in the treatment of HF.
Collapse
Affiliation(s)
- Junjie Du
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Dongmin Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jinghang Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Linjie Si
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Dawei Zhu
- Department of Cardiothoracic Surgery, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211000, China
| | - Ben Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yizhou Gao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Lifu Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xufeng Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
17
|
do Val Lima PR, Ronconi KS, Morra EA, Rodrigues PL, Ávila RA, Merlo E, Graceli JB, Simões MR, Stefanon I, Ribeiro Júnior RF. Testosterone deficiency impairs cardiac interfibrillar mitochondrial function and myocardial contractility while inducing oxidative stress. Front Endocrinol (Lausanne) 2023; 14:1206387. [PMID: 37780627 PMCID: PMC10534000 DOI: 10.3389/fendo.2023.1206387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/06/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Clinical studies have shown that low levels of endogenous testosterone are associated with cardiovascular diseases. Considering the intimate connection between oxidative metabolism and myocardial contractility, we determined the effects of testosterone deficiency on the two spatially distinct subpopulations of cardiac mitochondria, subsarcolemmal (SSM) and interfibrillar (IFM). Methods We assessed cardiac function and cardiac mitochondria structure of SSM and IFM after 12 weeks of testosterone deficiency in male Wistar rats. Results and Discussion Results show that low testosterone reduced myocardial contractility. Orchidectomy increased total left ventricular mitochondrial protein in the SSM, but not in IFM. The membrane potential, size and internal complexity in the IFM after orchidectomy were higher compared to the SHAM group. However, the rate of oxidative phosphorylation with all substrates in the IFM after orchidectomy was lower compared to the SHAM group. Testosterone replacement restored these changes. In the testosterone-deficient SSM group, oxidative phosphorylation was decreased with palmitoyl-L-carnitine as substrate; however, the mitochondrial calcium retention capacity in IFM was increased. There was no difference in swelling of the mitochondria in either group. These changes in IFM were followed by a reduction in phosphorylated form of AMP-activated protein kinase (p-AMPK-α), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) translocation to mitochondria and decreased mitochondrial transcription factor A (TFAM). Testosterone deficiency increased NADPH oxidase (NOX), angiotensin converting enzyme (ACE) protein expression and reduced mitochondrial antioxidant proteins such as manganese superoxide dismutase (Mn-SOD) and catalase in the IFM. Treatment with apocynin (1.5 mM in drinking water) normalized myocardial contractility and interfibrillar mitochondrial function in the testosterone depleted animals. In conclusion, our findings demonstrate that testosterone deficiency leads to reduced myocardial contractility and impaired cardiac interfibrillar mitochondrial function. Our data suggest the involvement of reactive oxygen species, with a possibility of NOX as an enzymatic source.
Collapse
Affiliation(s)
| | - Karoline Sousa Ronconi
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Elis Aguiar Morra
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Paula Lopes Rodrigues
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Renata Andrade Ávila
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Eduardo Merlo
- Department of Morphology, Federal University of Espírito Santo, Vitoria, ES, Brazil
| | - Jones B. Graceli
- Department of Morphology, Federal University of Espírito Santo, Vitoria, ES, Brazil
| | - Maylla Ronacher Simões
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Ivanita Stefanon
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | |
Collapse
|
18
|
Zhang J, Wang H, Slotabec L, Cheng F, Tan Y, Li J. Alterations of SIRT1/SIRT3 subcellular distribution in aging undermine cardiometabolic homeostasis during ischemia and reperfusion. Aging Cell 2023; 22:e13930. [PMID: 37537789 PMCID: PMC10497814 DOI: 10.1111/acel.13930] [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: 05/26/2022] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 08/05/2023] Open
Abstract
Age-related sensors Sirtuin1 (SIRT1) and Sirtuin3 (SIRT3) play an essential role in the protective response upon myocardial ischemia and/or reperfusion (I/R). However, the subcellular localization and co-regulatory network between cardiac SIRT1 and SIRT3 remain unknown, especially their effects on age-related metabolic regulation during acute ischemia and I/R. Here, we found that defects of cardiac SIRT1 or SIRT3 with aging result in an exacerbated cardiac physiological structural and functional deterioration after acute ischemic stress and failed recovery through reperfusion operation. In aged hearts, SIRT1 translocated into mitochondria and recruited more mitochondria SIRT3 to enhance their interaction during acute ischemia, acting as adaptive protection for the aging hearts from further mitochondria dysfunction. Subsequently, SIRT3-targeted proteomics revealed that SIRT1 plays a crucial role in maintaining mitochondrial integrity through SIRT3-mediated substrate metabolism during acute ischemic and I/R stress. Although the loss of SIRT1/SIRT3 led to a compromised PGC-1α/PPARα-mediated transcriptional control of fatty acid oxidation in response to acute ischemia and I/R, their crosstalk in mitochondria plays a more important role in the aging heart during acute ischemia. However, the increased mitochondria SIRT1-SIRT3 interaction promoted adaptive protection to aging-related fatty acid metabolic disorder via deacetylation of long-chain acyl CoA dehydrogenase (LCAD) during ischemic insults. Therefore, the dynamic network of SIRT1/SIRT3 acts as a mediator that regulates adaptive metabolic response to improve the tolerance of aged hearts to ischemic insults, which will facilitate investigation into the role of SIRT1/SIRT3 in age-related ischemic heart disease.
Collapse
Affiliation(s)
- Jingwen Zhang
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Department of SurgeryUniversity of South FloridaTampaFloridaUSA
| | - Hao Wang
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Department of SurgeryUniversity of South FloridaTampaFloridaUSA
| | - Lily Slotabec
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Department of SurgeryUniversity of South FloridaTampaFloridaUSA
| | - Feng Cheng
- Department of Pharmaceutical Sciences, College of PharmacyUniversity of South FloridaTampaFloridaUSA
| | - Yi Tan
- Pediatric Research Institute, Department of PediatricsUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Ji Li
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Department of SurgeryUniversity of South FloridaTampaFloridaUSA
- G.V. (Sonny) Montgomery VA Medical CenterJacksonMississippiUSA
| |
Collapse
|
19
|
Montalvo RN, Boeno FP, Dowllah IM, Moritz CEJ, Nguyen BL, Doerr V, Bomkamp MP, Smuder AJ. Exercise and Doxorubicin Modify Markers of Iron Overload and Cardiolipin Deficiency in Cardiac Mitochondria. Int J Mol Sci 2023; 24:ijms24097689. [PMID: 37175395 PMCID: PMC10177936 DOI: 10.3390/ijms24097689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent highly effective at limiting cancer progression. Despite the efficacy of this anticancer drug, the clinical use of DOX is limited due to cardiotoxicity. The cardiac mitochondria are implicated as the primary target of DOX, resulting in inactivation of electron transport system complexes, oxidative stress, and iron overload. However, it is established that the cardiac mitochondrial subpopulations reveal differential responses to DOX exposure, with subsarcolemmal (SS) mitochondria demonstrating redox imbalance and the intermyofibrillar (IMF) mitochondria showing reduced respiration. In this regard, exercise training is an effective intervention to prevent DOX-induced cardiac dysfunction. Although it is clear that exercise confers mitochondrial protection, it is currently unknown if exercise training mitigates DOX cardiac mitochondrial toxicity by promoting beneficial adaptations to both the SS and IMF mitochondria. To test this, SS and IMF mitochondria were isolated from sedentary and exercise-preconditioned female Sprague Dawley rats exposed to acute DOX treatment. Our findings reveal a greater effect of exercise preconditioning on redox balance and iron handling in the SS mitochondria of DOX-treated rats compared to IMF, with rescue of cardiolipin synthase 1 expression in both subpopulations. These results demonstrate that exercise preconditioning improves mitochondrial homeostasis when combined with DOX treatment, and that the SS mitochondria display greater protection compared to the IMF mitochondria. These data provide important insights into the molecular mechanisms that are in part responsible for exercise-induced protection against DOX toxicity.
Collapse
Affiliation(s)
- Ryan N Montalvo
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Franccesco P Boeno
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Imtiaz M Dowllah
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Cesar E Jacintho Moritz
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Branden L Nguyen
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Vivian Doerr
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Matthew P Bomkamp
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Ashley J Smuder
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
20
|
Ren M, Xu Y, Phoon CKL, Erdjument-Bromage H, Neubert TA, Schlame M. Knockout of cardiolipin synthase disrupts postnatal cardiac development by inhibiting the maturation of mitochondrial cristae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.531996. [PMID: 36945411 PMCID: PMC10029008 DOI: 10.1101/2023.03.09.531996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Background Cardiomyocyte maturation requires a massive increase in respiratory enzymes and their assembly into long-lived complexes of oxidative phosphorylation (OXPHOS). The molecular mechanisms underlying the maturation of cardiac mitochondria have not been established. Methods To determine whether the mitochondria-specific lipid cardiolipin is involved in cardiac maturation, we created a cardiomyocyte-restricted knockout (KO) of cardiolipin synthase ( Crls1 ) in mice and studied the postnatal development of the heart. We also measured the turnover rates of proteins and lipids in cardiolipin-deficient flight muscle from Drosophila, a tissue that has mitochondria with high OXPHOS activity like the heart. Results Crls1KO mice survived the prenatal period but failed to accumulate OXPHOS proteins during postnatal maturation and succumbed to heart failure at the age of 2 weeks. Turnover measurements showed that the exceptionally long half-life of OXPHOS proteins is critically dependent on cardiolipin. Conclusions Cardiolipin is essential for the postnatal maturation of cardiomyocytes because it allows mitochondrial cristae to accumulate OXPHOS proteins to a high concentration and to shield them from degradation.
Collapse
|
21
|
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
|
22
|
Karsenty C, Guilbeau-Frugier C, Genet G, Seguelas MH, Alzieu P, Cazorla O, Montagner A, Blum Y, Dubroca C, Maupoint J, Tramunt B, Cauquil M, Sulpice T, Richard S, Arcucci S, Flores-Flores R, Pataluch N, Montoriol R, Sicard P, Deney A, Couffinhal T, Senard JM, Galés C. Ephrin-B1 regulates the adult diastolic function through a late postnatal maturation of cardiomyocyte surface crests. eLife 2023; 12:e80904. [PMID: 36649053 PMCID: PMC9844986 DOI: 10.7554/elife.80904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
The rod-shaped adult cardiomyocyte (CM) harbors a unique architecture of its lateral surface with periodic crests, relying on the presence of subsarcolemmal mitochondria (SSM) with unknown role. Here, we investigated the development and functional role of CM crests during the postnatal period. We found in rodents that CM crest maturation occurs late between postnatal day 20 (P20) and P60 through both SSM biogenesis, swelling and crest-crest lateral interactions between adjacent CM, promoting tissue compaction. At the functional level, we showed that the P20-P60 period is dedicated to the improvement of relaxation. Interestingly, crest maturation specifically contributes to an atypical CM hypertrophy of its short axis, without myofibril addition, but relying on CM lateral stretching. Mechanistically, using constitutive and conditional CM-specific knock-out mice, we identified ephrin-B1, a lateral membrane stabilizer, as a molecular determinant of P20-P60 crest maturation, governing both the CM lateral stretch and the diastolic function, thus highly suggesting a link between crest maturity and diastole. Remarkably, while young adult CM-specific Efnb1 KO mice essentially exhibit an impairment of the ventricular diastole with preserved ejection fraction and exercise intolerance, they progressively switch toward systolic heart failure with 100% KO mice dying after 13 months, indicative of a critical role of CM-ephrin-B1 in the adult heart function. This study highlights the molecular determinants and the biological implication of a new late P20-P60 postnatal developmental stage of the heart in rodents during which, in part, ephrin-B1 specifically regulates the maturation of the CM surface crests and of the diastolic function.
Collapse
Affiliation(s)
- Clement Karsenty
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Pediatric Cardiology, Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Celine Guilbeau-Frugier
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Forensic Medicine, Centre Hospitalier Universitaire de Toulouse, Université de ToulouseToulouseFrance
| | - Gaël Genet
- Department of Cell Biology, University of Virginia School of MedicineCharlottesvilleUnited States
| | - Marie-Helene Seguelas
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Philippe Alzieu
- Université de Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
| | - Olivier Cazorla
- Université de Montpellier, INSERM, CNRS, PhyMedExpMontpellierFrance
| | - Alexandra Montagner
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Yuna Blum
- IGDR UMR 6290, CNRS, Université de Rennes 1RennesFrance
| | | | | | - Blandine Tramunt
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Diabetology, Metabolic Diseases & Nutrition, Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Marie Cauquil
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | | | - Sylvain Richard
- Université de Montpellier, INSERM, CNRS, PhyMedExpMontpellierFrance
| | - Silvia Arcucci
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Remy Flores-Flores
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Nicolas Pataluch
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Romain Montoriol
- Department of Forensic Medicine, Centre Hospitalier Universitaire de Toulouse, Université de ToulouseToulouseFrance
| | - Pierre Sicard
- Université de Montpellier, INSERM, CNRS, PhyMedExpMontpellierFrance
| | - Antoine Deney
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Thierry Couffinhal
- Université de Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
- Service des Maladies Cardiaques et Vasculaires, CHU de BordeauxBordeauxFrance
| | - Jean-Michel Senard
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Clinical Pharmacology, Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Celine Galés
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| |
Collapse
|
23
|
Sivakumar B, AlAsmari AF, Ali N, Waseem M, Kurian GA. Consequential Impact of Particulate Matter Linked Inter-Fibrillar Mitochondrial Dysfunction in Rat Myocardium Subjected to Ischemia Reperfusion Injury. BIOLOGY 2022; 11:biology11121811. [PMID: 36552319 PMCID: PMC9775305 DOI: 10.3390/biology11121811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
A previous study has reported that exposure to PM2.5 from diesel exhaust (diesel particulate matter (DPM)) for 21 days can deteriorate the cardiac recovery from myocardial ischemia reperfusion injury (IR), where the latter is facilitated by the efficiency of mitochondrial subpopulations. Many investigators have demonstrated that IR impact on cardiac mitochondrial subpopulations is distinct. In the present study, we decipher the role of PM2.5 on IR associated mitochondrial dysfunction at the subpopulation level by administrating PM2.5 directly to isolated female rat hearts via KH buffer. Our results demonstrated that PM2.5 administered heart (PM_C) severely deteriorated ETC enzyme activity (NQR, SQR, QCR, and COX) and ATP level in both IFM and SSM from the normal control. Comparatively, the declined activity was prominent in IFM fraction. Moreover, in the presence of IR (PM_IR), mitochondrial oxidative stress was higher in both subpopulations from the normal, where the IFM fraction of mitochondria experienced elevated oxidative stress than SSM. Furthermore, we assessed the in vitro protein translation capacity of IFM and SSM and found a declined ability in both subpopulations where the inability of IFM was significant in both PM_C and PM_IR groups. In support of these results, the expression of mitochondrial genes involved in fission, fusion, and mitophagy events along with the DNA maintenance genes such as GUF1, LRPPRC, and HSD17-b10 were significantly altered from the control. Based on the above results, we conclude that PM2.5 administration to the heart inflicted mitochondrial damage especially to the IFM fraction, that not only deteriorated the cardiac physiology but also reduced its ability to resist IR injury.
Collapse
Affiliation(s)
- Bhavana Sivakumar
- Vascular Biology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India
| | - Abdullah F. AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Waseem
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
| | - Gino A. Kurian
- Vascular Biology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India
- Correspondence: ; Tel.: +91-9047965425; Fax: +91-4362-264120
| |
Collapse
|
24
|
Pedriali G, Ramaccini D, Bouhamida E, Wieckowski MR, Giorgi C, Tremoli E, Pinton P. Perspectives on mitochondrial relevance in cardiac ischemia/reperfusion injury. Front Cell Dev Biol 2022; 10:1082095. [PMID: 36561366 PMCID: PMC9763599 DOI: 10.3389/fcell.2022.1082095] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is the most common cause of death worldwide and in particular, ischemic heart disease holds the most considerable position. Even if it has been deeply studied, myocardial ischemia-reperfusion injury (IRI) is still a side-effect of the clinical treatment for several heart diseases: ischemia process itself leads to temporary damage to heart tissue and obviously the recovery of blood flow is promptly required even if it worsens the ischemic injury. There is no doubt that mitochondria play a key role in pathogenesis of IRI: dysfunctions of these important organelles alter cell homeostasis and survival. It has been demonstrated that during IRI the system of mitochondrial quality control undergoes alterations with the disruption of the complex balance between the processes of mitochondrial fusion, fission, biogenesis and mitophagy. The fundamental role of mitochondria is carried out thanks to the finely regulated connection to other organelles such as plasma membrane, endoplasmic reticulum and nucleus, therefore impairments of these inter-organelle communications exacerbate IRI. This review pointed to enhance the importance of the mitochondrial network in the pathogenesis of IRI with the aim to focus on potential mitochondria-targeting therapies as new approach to control heart tissue damage after ischemia and reperfusion process.
Collapse
Affiliation(s)
- Gaia Pedriali
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | | | - Esmaa Bouhamida
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy,*Correspondence: Paolo Pinton, ; Elena Tremoli,
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care and Research, Cotignola, Italy,Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, Section of Experimental Medicine, University of Ferrara, Ferrara, Italy,*Correspondence: Paolo Pinton, ; Elena Tremoli,
| |
Collapse
|
25
|
Birkedal R, Laasmaa M, Branovets J, Vendelin M. Ontogeny of cardiomyocytes: ultrastructure optimization to meet the demand for tight communication in excitation-contraction coupling and energy transfer. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210321. [PMID: 36189816 PMCID: PMC9527910 DOI: 10.1098/rstb.2021.0321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The ontogeny of the heart describes its development from the fetal to the adult stage. In newborn mammals, blood pressure and thus cardiac performance are relatively low. The cardiomyocytes are thin, and with a central core of mitochondria surrounded by a ring of myofilaments, while the sarcoplasmic reticulum (SR) is sparse. During development, as blood pressure and performance increase, the cardiomyocytes become more packed with structures involved in excitation–contraction (e-c) coupling (SR and myofilaments) and the generation of ATP (mitochondria) to fuel the contraction. In parallel, the e-c coupling relies increasingly on calcium fluxes through the SR, while metabolism relies increasingly on fatty acid oxidation. The development of transverse tubules and SR brings channels and transporters interacting via calcium closer to each other and is crucial for e-c coupling. However, for energy transfer, it may seem counterintuitive that the increased structural density restricts the overall ATP/ADP diffusion. In this review, we discuss how this is because of the organization of all these structures forming modules. Although the overall diffusion across modules is more restricted, the energy transfer within modules is fast. A few studies suggest that in failing hearts this modular design is disrupted, and this may compromise intracellular energy transfer. This article is part of the theme issue ‘The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease’.
Collapse
Affiliation(s)
- Rikke Birkedal
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Akadeemia 15, room SCI-218, 12618 Tallinn, Estonia
| | - Martin Laasmaa
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Akadeemia 15, room SCI-218, 12618 Tallinn, Estonia
| | - Jelena Branovets
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Akadeemia 15, room SCI-218, 12618 Tallinn, Estonia
| | - Marko Vendelin
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Akadeemia 15, room SCI-218, 12618 Tallinn, Estonia
| |
Collapse
|
26
|
Zhang Y, Chen Y, Zhao S. A cell model for evaluating mitochondrial damage in cardiomyocytes. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00313-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Abstract
Background
Various cellular models were used for assessment of mitochondrial damage in cardiomyocyte, but most of them are based on silent cells without contractility. The mitochondria in cells at working should be more sensitive to toxic or reperfusion damage due to their high level mitochondrial respiration. Therefore, contracting cells can represent inotropic agent-mediated high-energy demand states.
Objective
To establish a cellular model to detect mitochondrial damage in cardiomyocytes at contraction.
Method
Freshly isolated Sprague–Dawley rat cardiomyocytes were incubated with or without bupivacaine, in the presence or absence of isoprenaline, and electrically stimulated to induce rhythmic contractions.
Results
Contraction under electrical field stimulation did not induce mitochondrial swelling or ROS production in DMEM; the silent cells in the presence of bupivacaine showed mild mitochondrial swelling, but contracting cells exhibited significantly higher mitochondrial swelling and increased ROS production (P < 0.05, vs. silent cells). Isoprenaline induced a further enhancement in mitochondrial swelling and ROS production in contracting cells.
Conclusions
Contracting cells are more sensitive to bupivacaine toxicity and could be more accurately represent mitochondrial damage in vivo condition.
Collapse
|
27
|
Campbell T, Slone J, Huang T. Mitochondrial Genome Variants as a Cause of Mitochondrial Cardiomyopathy. Cells 2022; 11:cells11182835. [PMID: 36139411 PMCID: PMC9496904 DOI: 10.3390/cells11182835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondria are small double-membraned organelles responsible for the generation of energy used in the body in the form of ATP. Mitochondria are unique in that they contain their own circular mitochondrial genome termed mtDNA. mtDNA codes for 37 genes, and together with the nuclear genome (nDNA), dictate mitochondrial structure and function. Not surprisingly, pathogenic variants in the mtDNA or nDNA can result in mitochondrial disease. Mitochondrial disease primarily impacts tissues with high energy demands, including the heart. Mitochondrial cardiomyopathy is characterized by the abnormal structure or function of the myocardium secondary to genetic defects in either the nDNA or mtDNA. Mitochondrial cardiomyopathy can be isolated or part of a syndromic mitochondrial disease. Common manifestations of mitochondrial cardiomyopathy are a phenocopy of hypertrophic cardiomyopathy, dilated cardiomyopathy, and cardiac conduction defects. The underlying pathophysiology of mitochondrial cardiomyopathy is complex and likely involves multiple abnormal processes in the cell, stemming from deficient oxidative phosphorylation and ATP depletion. Possible pathophysiology includes the activation of alternative metabolic pathways, the accumulation of reactive oxygen species, dysfunctional mitochondrial dynamics, abnormal calcium homeostasis, and mitochondrial iron overload. Here, we highlight the clinical assessment of mtDNA-related mitochondrial cardiomyopathy and offer a novel hypothesis of a possible integrated, multivariable pathophysiology of disease.
Collapse
|
28
|
Cardiomyocyte Proliferation from Fetal- to Adult- and from Normal- to Hypertrophy and Failing Hearts. BIOLOGY 2022; 11:biology11060880. [PMID: 35741401 PMCID: PMC9220194 DOI: 10.3390/biology11060880] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 11/20/2022]
Abstract
Simple Summary Death from injury to the heart from a variety of causes remains a major cause of mortality worldwide. The cardiomyocyte, the major contracting cell of the heart, is responsible for pumping blood to the rest of the body. During fetal development, these immature cardiomyocytes are small and rapidly divide to complete development of the heart by birth when they develop structural and functional characteristics of mature cells which prevent further division. All further growth of the heart after birth is due to an increase in the size of cardiomyocytes, hypertrophy. Following the loss of functional cardiomyocytes due to coronary artery occlusion or other causes, the heart is unable to replace the lost cells. One of the significant research goals has been to induce adult cardiomyocytes to reactivate the cell cycle and repair cardiac injury. This review explores the developmental, structural, and functional changes of the growing cardiomyocyte, and particularly the sarcomere, responsible for force generation, from the early fetal period of reproductive cell growth through the neonatal period and on to adulthood, as well as during pathological response to different forms of myocardial diseases or injury. Multiple issues relative to cardiomyocyte cell-cycle regulation in normal or diseased conditions are discussed. Abstract The cardiomyocyte undergoes dramatic changes in structure, metabolism, and function from the early fetal stage of hyperplastic cell growth, through birth and the conversion to hypertrophic cell growth, continuing to the adult stage and responding to various forms of stress on the myocardium, often leading to myocardial failure. The fetal cell with incompletely formed sarcomeres and other cellular and extracellular components is actively undergoing mitosis, organelle dispersion, and formation of daughter cells. In the first few days of neonatal life, the heart is able to repair fully from injury, but not after conversion to hypertrophic growth. Structural and metabolic changes occur following conversion to hypertrophic growth which forms a barrier to further cardiomyocyte division, though interstitial components continue dividing to keep pace with cardiac growth. Both intra- and extracellular structural changes occur in the stressed myocardium which together with hemodynamic alterations lead to metabolic and functional alterations of myocardial failure. This review probes some of the questions regarding conditions that regulate normal and pathologic growth of the heart.
Collapse
|
29
|
Chapa-Dubocq XR, Garcia-Baez JF, Bazil JN, Javadov S. Crosstalk between adenine nucleotide transporter and mitochondrial swelling: experimental and computational approaches. Cell Biol Toxicol 2022:10.1007/s10565-022-09724-2. [PMID: 35606662 DOI: 10.1007/s10565-022-09724-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022]
Abstract
Mitochondrial metabolism and function are modulated by changes in matrix Ca2+. Small increases in the matrix Ca2+ stimulate mitochondrial bioenergetics, whereas excessive Ca2+ leads to cell death by causing massive matrix swelling and impairing the structural and functional integrity of mitochondria. Sustained opening of the non-selective mitochondrial permeability transition pores (PTP) is the main mechanism responsible for mitochondrial Ca2+ overload that leads to mitochondrial dysfunction and cell death. Recent studies suggest the existence of two or more types of PTP, and adenine nucleotide translocator (ANT) and FOF1-ATP synthase were proposed to form the PTP independent of each other. Here, we elucidated the role of ANT in PTP opening by applying both experimental and computational approaches. We first developed and corroborated a detailed model of the ANT transport mechanism including the matrix (ANTM), cytosolic (ANTC), and pore (ANTP) states of the transporter. Then, the ANT model was incorporated into a simple, yet effective, empirical model of mitochondrial bioenergetics to ascertain the point when Ca2+ overload initiates PTP opening via an ANT switch-like mechanism activated by matrix Ca2+ and is inhibited by extra-mitochondrial ADP. We found that encoding a heterogeneous Ca2+ response of at least three types of PTPs, weakly, moderately, and strongly sensitive to Ca2+, enabled the model to simulate Ca2+ release dynamics observed after large boluses were administered to a population of energized cardiac mitochondria. Thus, this study demonstrates the potential role of ANT in PTP gating and proposes a novel mechanism governing the cryptic nature of the PTP phenomenon.
Collapse
Affiliation(s)
- Xavier R Chapa-Dubocq
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, 00936-5067, USA
| | - Jorge F Garcia-Baez
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, 00936-5067, USA
| | - Jason N Bazil
- Department of Physiology, Michigan State University, East Lansing, MI, 48824-1046, USA
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR, 00936-5067, USA.
| |
Collapse
|
30
|
Rajagopal V, Arumugam S, Hunter PJ, Khadangi A, Chung J, Pan M. The Cell Physiome: What Do We Need in a Computational Physiology Framework for Predicting Single-Cell Biology? Annu Rev Biomed Data Sci 2022; 5:341-366. [PMID: 35576556 DOI: 10.1146/annurev-biodatasci-072018-021246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Modern biology and biomedicine are undergoing a big data explosion, needing advanced computational algorithms to extract mechanistic insights on the physiological state of living cells. We present the motivation for the Cell Physiome project: a framework and approach for creating, sharing, and using biophysics-based computational models of single-cell physiology. Using examples in calcium signaling, bioenergetics, and endosomal trafficking, we highlight the need for spatially detailed, biophysics-based computational models to uncover new mechanisms underlying cell biology. We review progress and challenges to date toward creating cell physiome models. We then introduce bond graphs as an efficient way to create cell physiome models that integrate chemical, mechanical, electromagnetic, and thermal processes while maintaining mass and energy balance. Bond graphs enhance modularization and reusability of computational models of cells at scale. We conclude with a look forward at steps that will help fully realize this exciting new field of mechanistic biomedical data science. Expected final online publication date for the Annual Review of Biomedical Data Science, Volume 5 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Vijay Rajagopal
- Department of Biomedical Engineering, University of Melbourne, Melbourne, Victoria, Australia;
| | - Senthil Arumugam
- Cellular Physiology Lab, Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences; European Molecular Biological Laboratory (EMBL) Australia; and Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton/Melbourne, Victoria, Australia
| | - Peter J Hunter
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Afshin Khadangi
- Department of Biomedical Engineering, University of Melbourne, Melbourne, Victoria, Australia;
| | - Joshua Chung
- Department of Biomedical Engineering, University of Melbourne, Melbourne, Victoria, Australia;
| | - Michael Pan
- School of Mathematics and Statistics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
31
|
Therapeutic Targets for Regulating Oxidative Damage Induced by Ischemia-Reperfusion Injury: A Study from a Pharmacological Perspective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8624318. [PMID: 35450409 PMCID: PMC9017553 DOI: 10.1155/2022/8624318] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/28/2022] [Accepted: 03/15/2022] [Indexed: 12/22/2022]
Abstract
Ischemia-reperfusion (I-R) injury is damage caused by restoring blood flow into ischemic tissues or organs. This complex and characteristic lesion accelerates cell death induced by signaling pathways such as apoptosis, necrosis, and even ferroptosis. In addition to the direct association between I-R and the release of reactive oxygen species and reactive nitrogen species, it is involved in developing mitochondrial oxidative damage. Thus, its mechanism plays a critical role via reactive species scavenging, calcium overload modulation, electron transport chain blocking, mitochondrial permeability transition pore activation, or noncoding RNA transcription. Other receptors and molecules reduce tissue and organ damage caused by this pathology and other related diseases. These molecular targets have been gradually discovered and have essential roles in I-R resolution. Therefore, the current study is aimed at highlighting the importance of these discoveries. In this review, we inquire about the oxidative damage receptors that are relevant to reducing the damage induced by oxidative stress associated with I-R. Several complications on surgical techniques and pathology interventions do not mitigate the damage caused by I-R. Nevertheless, these therapies developed using alternative targets could work as coadjuvants in tissue transplants or I-R-related pathologies
Collapse
|
32
|
Garbincius JF, Elrod JW. Mitochondrial calcium exchange in physiology and disease. Physiol Rev 2022; 102:893-992. [PMID: 34698550 PMCID: PMC8816638 DOI: 10.1152/physrev.00041.2020] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 08/16/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
The uptake of calcium into and extrusion of calcium from the mitochondrial matrix is a fundamental biological process that has critical effects on cellular metabolism, signaling, and survival. Disruption of mitochondrial calcium (mCa2+) cycling is implicated in numerous acquired diseases such as heart failure, stroke, neurodegeneration, diabetes, and cancer and is genetically linked to several inherited neuromuscular disorders. Understanding the mechanisms responsible for mCa2+ exchange therefore holds great promise for the treatment of these diseases. The past decade has seen the genetic identification of many of the key proteins that mediate mitochondrial calcium uptake and efflux. Here, we present an overview of the phenomenon of mCa2+ transport and a comprehensive examination of the molecular machinery that mediates calcium flux across the inner mitochondrial membrane: the mitochondrial uniporter complex (consisting of MCU, EMRE, MICU1, MICU2, MICU3, MCUB, and MCUR1), NCLX, LETM1, the mitochondrial ryanodine receptor, and the mitochondrial permeability transition pore. We then consider the physiological implications of mCa2+ flux and evaluate how alterations in mCa2+ homeostasis contribute to human disease. This review concludes by highlighting opportunities and challenges for therapeutic intervention in pathologies characterized by aberrant mCa2+ handling and by summarizing critical unanswered questions regarding the biology of mCa2+ flux.
Collapse
Affiliation(s)
- Joanne F Garbincius
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
33
|
Mitochondrial Ca 2+ Homeostasis: Emerging Roles and Clinical Significance in Cardiac Remodeling. Int J Mol Sci 2022; 23:ijms23063025. [PMID: 35328444 PMCID: PMC8954803 DOI: 10.3390/ijms23063025] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 01/27/2023] Open
Abstract
Mitochondria are the sites of oxidative metabolism in eukaryotes where the metabolites of sugars, fats, and amino acids are oxidized to harvest energy. Notably, mitochondria store Ca2+ and work in synergy with organelles such as the endoplasmic reticulum and extracellular matrix to control the dynamic balance of Ca2+ concentration in cells. Mitochondria are the vital organelles in heart tissue. Mitochondrial Ca2+ homeostasis is particularly important for maintaining the physiological and pathological mechanisms of the heart. Mitochondrial Ca2+ homeostasis plays a key role in the regulation of cardiac energy metabolism, mechanisms of death, oxygen free radical production, and autophagy. The imbalance of mitochondrial Ca2+ balance is closely associated with cardiac remodeling. The mitochondrial Ca2+ uniporter (mtCU) protein complex is responsible for the uptake and release of mitochondrial Ca2+ and regulation of Ca2+ homeostasis in mitochondria and consequently, in cells. This review summarizes the mechanisms of mitochondrial Ca2+ homeostasis in physiological and pathological cardiac remodeling and the regulatory effects of the mitochondrial calcium regulatory complex on cardiac energy metabolism, cell death, and autophagy, and also provides the theoretical basis for mitochondrial Ca2+ as a novel target for the treatment of cardiovascular diseases.
Collapse
|
34
|
Chandra Shekar K, Yannopoulos D, Kosmopoulos M, Riess ML. Differential Effects of Reperfusion on Cardiac Mitochondrial Subpopulations in a Preclinical Porcine Model of Acute Myocardial Infarction. Front Cell Dev Biol 2022; 10:843733. [PMID: 35356287 PMCID: PMC8959812 DOI: 10.3389/fcell.2022.843733] [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: 12/26/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022] Open
Abstract
Acute myocardial infarction (AMI) leads to localized cardiac ischemia and can be fatal if untreated. Despite being treatable, the threat of ischemia-reperfusion (IR) injury remains high. Mitochondria are central to both propagation and mitigation of IR injury, and cardiac mitochondria are categorized into two major subtypes-subsarcolemmal and interfibrillar mitochondria (SSM and IFM, respectively). We hypothesized that, in our pre-clinical porcine model of AMI, SSM and IFM are differentially affected by reperfusion. AMI was induced in female pigs by balloon occlusion of the left anterior descending artery for 45 min, followed by 4 h of reperfusion. At the end of reperfusion, animals were euthanized. Cardiac SSM and IFM from the affected ischemic area and a nearby non-ischemic area were isolated to compare mitochondrial function using substrates targeting mitochondrial electron transport chain complexes I and II. Despite detecting overall significant differences in mitochondrial function including yield, mitochondrial S3 and S4 respirations, and calcium retention, consistent individual functional differences in the two mitochondrial subpopulations were not observed, both between the two mitochondrial subtypes, as well as between the ischemic and non-ischemic tissue. Nonetheless, this study describes the mitochondrial subtype response within the initial few hours of reperfusion in a clinically relevant model of AMI, which provides valuable information needed to develop novel mitochondrially targeted therapies for AMI.
Collapse
Affiliation(s)
- Kadambari Chandra Shekar
- Integrative Biology and Physiology, University of Minnesota at Twin Cities, St. Paul, MN, United States
| | - Demetris Yannopoulos
- Department of Cardiology, Division of Medicine, University of Minnesota at Twin Cities, St. Paul, MN, United States
| | - Marinos Kosmopoulos
- Department of Cardiology, Division of Medicine, University of Minnesota at Twin Cities, St. Paul, MN, United States
| | - Matthias L. Riess
- Anesthesiology, TVHS VA Medical Center, Nashville, TN, United States
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
35
|
Durr AJ, Hathaway QA, Kunovac A, Taylor AD, Pinti MV, Rizwan S, Shepherd DL, Cook CC, Fink GK, Hollander JM. Manipulation of the miR-378a/mt-ATP6 regulatory axis rescues ATP synthase in the diabetic heart and offers a novel role for lncRNA Kcnq1ot1. Am J Physiol Cell Physiol 2022; 322:C482-C495. [PMID: 35108116 PMCID: PMC8917913 DOI: 10.1152/ajpcell.00446.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus has been linked to an increase in mitochondrial microRNA-378a (miR-378a) content. Enhanced miR-378a content has been associated with a reduction in mitochondrial genome-encoded mt-ATP6 abundance, supporting the hypothesis that miR-378a inhibition may be a therapeutic option for maintaining ATP synthase functionality during diabetes mellitus. Evidence also suggests that long noncoding RNAs (lncRNAs), including lncRNA potassium voltage-gated channel subfamily Q member 1 overlapping transcript 1 (Kcnq1ot1), participate in regulatory axes with microRNAs (miRs). Prediction analyses indicate that Kcnq1ot1 has the potential to bind miR-378a. This study aimed to determine if loss of miR-378a in a genetic mouse model could ameliorate cardiac dysfunction in type 2 diabetes mellitus (T2DM) and to ascertain whether Kcnq1ot1 interacts with miR-378a to impact ATP synthase functionality by preserving mt-ATP6 levels. MiR-378a was significantly higher in patients with T2DM and 25-wk-old Db/Db mouse mitochondria, whereas mt-ATP6 and Kcnq1ot1 levels were significantly reduced when compared with controls. Twenty-five-week-old miR-378a knockout Db/Db mice displayed preserved mt-ATP6 and ATP synthase protein content, ATP synthase activity, and preserved cardiac function, implicating miR-378a as a potential therapeutic target in T2DM. Assessments following overexpression of the 500-bp Kcnq1ot1 fragment in established mouse cardiomyocyte cell line (HL-1) cardiomyocytes overexpressing miR-378a revealed that Kcnq1ot1 may bind and significantly reduce miR-378a levels, and rescue mt-ATP6 and ATP synthase protein content. Together, these data suggest that Kcnq1ot1 and miR-378a may act as constituents in an axis that regulates mt-ATP6 content, and that manipulation of this axis may provide benefit to ATP synthase functionality in type 2 diabetic heart.
Collapse
Affiliation(s)
- Andrya J Durr
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Quincy A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Amina Kunovac
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Andrew D Taylor
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Mark V Pinti
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
- West Virginia University School of Pharmacy, Morgantown, West Virginia
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Saira Rizwan
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Danielle L Shepherd
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Chris C Cook
- Cardiovascular and Thoracic Surgery, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Garrett K Fink
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - John M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
36
|
The Role of Oxidative Stress in the Aging Heart. Antioxidants (Basel) 2022; 11:antiox11020336. [PMID: 35204217 PMCID: PMC8868312 DOI: 10.3390/antiox11020336] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/17/2022] Open
Abstract
Medical advances and the availability of diagnostic tools have considerably increased life expectancy and, consequently, the elderly segment of the world population. As age is a major risk factor in cardiovascular disease (CVD), it is critical to understand the changes in cardiac structure and function during the aging process. The phenotypes and molecular mechanisms of cardiac aging include several factors. An increase in oxidative stress is a major player in cardiac aging. Reactive oxygen species (ROS) production is an important mechanism for maintaining physiological processes; its generation is regulated by a system of antioxidant enzymes. Oxidative stress occurs from an imbalance between ROS production and antioxidant defenses resulting in the accumulation of free radicals. In the heart, ROS activate signaling pathways involved in myocyte hypertrophy, interstitial fibrosis, contractile dysfunction, and inflammation thereby affecting cell structure and function, and contributing to cardiac damage and remodeling. In this manuscript, we review recent published research on cardiac aging. We summarize the aging heart biology, highlighting key molecular pathways and cellular processes that underlie the redox signaling changes during aging. Main ROS sources, antioxidant defenses, and the role of dysfunctional mitochondria in the aging heart are addressed. As metabolism changes contribute to cardiac aging, we also comment on the most prevalent metabolic alterations. This review will help us to understand the mechanisms involved in the heart aging process and will provide a background for attractive molecular targets to prevent age-driven pathology of the heart. A greater understanding of the processes involved in cardiac aging may facilitate our ability to mitigate the escalating burden of CVD in older individuals and promote healthy cardiac aging.
Collapse
|
37
|
Renal Revascularization Attenuates Myocardial Mitochondrial Damage and Improves Diastolic Function in Pigs with Metabolic Syndrome and Renovascular Hypertension. J Cardiovasc Transl Res 2022; 15:15-26. [PMID: 34269985 PMCID: PMC8761225 DOI: 10.1007/s12265-021-10155-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/03/2021] [Indexed: 02/03/2023]
Abstract
Percutaneous transluminal renal angioplasty (PTRA) may improve cardiac function in renovascular hypertension (RVH), but its effect on the biological mechanisms implicated in cardiac damage remains unknown. We hypothesized that restoration of kidney function by PTRA ameliorates myocardial mitochondrial damage and preserves cardiac function in pigs with metabolic syndrome (MetS) and RVH. Pigs were studied after 16 weeks of MetS+RVH, MetS+RVH treated 4 weeks earlier with PTRA, and Lean and MetS Sham controls (n=6 each). Cardiac function was assessed by multi-detector CT, whereas cardiac mitochondrial morphology and function, microvascular remodeling, and injury pathways were assessed ex vivo. PTRA attenuated myocardial mitochondrial damage, improved capillary and microvascular maturity, and ameliorated oxidative stress and fibrosis, in association with attenuation of left ventricular remodeling and diastolic dysfunction. Myocardial mitochondrial damage correlated with myocardial injury and renal dysfunction. Preservation of myocardial mitochondria with PTRA can enhance cardiac recovery, underscoring its therapeutic potential in experimental MetS+RVH.
Collapse
|
38
|
Sivakumar B, Kurian GA. Diesel particulate matter exposure deteriorates cardiovascular health and increases the sensitivity of rat heart towards ischemia reperfusion injury via suppressing mitochondrial bioenergetics function. Chem Biol Interact 2022; 351:109769. [PMID: 34875278 DOI: 10.1016/j.cbi.2021.109769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 11/03/2022]
Abstract
Documents from previous studies do not sufficiently explain the pathophysiological alterations involved in rat hearts exposed to PM2.5 from diesel exhaust, termed as Diesel Particulate matter (DPM). In the present study, we explored the cardiovascular effect of DPM exposure on the recovery of heart from Ischemia reperfusion injury (IR) and explored the probable cause-effect relationship. Two groups of female Wistar rats were exposed to 0.5 mg/ml DPM for 1 h and 3 h durations daily for 21 days via a whole-body exposure system. At the end of 21st day, the animals were sacrificed and the heart was subjected to IR via Langendorff isolated rat heart perfusion system. 21 days of exposure altered cardiac electrophysiology and the ultra-structure of myocardium. Also, the same group of animals exhibited calcification in the vasculature. These changes were prominent in animals exposed to DPM for 3 h daily. Administration of DPM to H9C2 cells resulted in 15% and 36% cell death after 1hr and 3hrs of incubation, respectively. When the hearts were challenged to IR, both 1 h and 3 h exposed hearts exhibited a significant decline in IR recovery. At the sub-cellular level, DPM exposure reduced ATP levels, mitochondrial copy number, and increased oxidative stress after IR in both exposure groups. These changes were markedly seen in the interfibrillar mitochondrial fraction of the mitochondria. Hence, we conclude that exposure to PM2.5 from diesel exhaust alters electrophysiology and ultrastructure of heart and reduces the level of cellular mediators, thereby compromising the ability of heart to withstand IR injury.
Collapse
Affiliation(s)
- Bhavana Sivakumar
- Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Gino A Kurian
- Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India; School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur, 613401, Tamil Nadu, India.
| |
Collapse
|
39
|
Abdullah CS, Aishwarya R, Alam S, Remex NS, Morshed M, Nitu S, Miriyala S, Panchatcharam M, Hartman B, King J, Alfrad Nobel Bhuiyan M, Traylor J, Kevil CG, Orr AW, Bhuiyan MS. The molecular role of Sigmar1 in regulating mitochondrial function through mitochondrial localization in cardiomyocytes. Mitochondrion 2022; 62:159-175. [PMID: 34902622 PMCID: PMC8790786 DOI: 10.1016/j.mito.2021.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 01/03/2023]
Abstract
Sigmar1 is a widely expressed molecular chaperone protein in mammalian cell systems. Accumulating research demonstrated the cardioprotective roles of pharmacologic Sigmar1 activation by ligands in preclinical rodent models of cardiac injury. Extensive biochemical and immuno-electron microscopic research demonstrated Sigmar1's sub-cellular localization largely depends on cell and organ types. Despite comprehensive studies, Sigmar1's direct molecular role in cardiomyocytes remains elusive. In the present study, we determined Sigmar1's subcellular localization, transmembrane topology, and function using complementary microscopy, biochemical, and functional assays in cardiomyocytes. Quantum dots in transmission electron microscopy showed Sigmar1 labeled quantum dots on the mitochondrial membranes, lysosomes, and sarcoplasmic reticulum-mitochondrial interface. Subcellular fractionation of heart cell lysates confirmed Sigmar1's localization in purified mitochondria fraction and lysosome fraction. Immunocytochemistry confirmed Sigmar1 colocalization with mitochondrial proteins in isolated adult mouse cardiomyocytes. Sigmar1's mitochondrial localization was further confirmed by Sigmar1 colocalization with Mito-Tracker in isolated mouse heart mitochondria. A series of biochemical experiments, including alkaline extraction and proteinase K treatment of purified heart mitochondria, demonstrated Sigmar1 as an integral mitochondrial membrane protein. Sigmar1's structural requirement for mitochondrial localization was determined by expressing FLAG-tagged Sigmar1 fragments in cells. Full-length Sigmar1 and Sigmar1's C terminal-deletion fragments were able to localize to the mitochondrial membrane, whereas N-terminal deletion fragment was unable to incorporate into the mitochondria. Finally, functional assays using extracellular flux analyzer and high-resolution respirometry showed Sigmar1 siRNA knockdown significantly altered mitochondrial respiration in cardiomyocytes. Overall, we found that Sigmar1 localizes to mitochondrial membranes and is indispensable for maintaining mitochondrial respiratory homeostasis in cardiomyocytes.
Collapse
Affiliation(s)
- Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Richa Aishwarya
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Shafiul Alam
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Sadia Nitu
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Sumitra Miriyala
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Manikandan Panchatcharam
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Brandon Hartman
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Judy King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | | | - James Traylor
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71103, USA.
| |
Collapse
|
40
|
Kong L, Chen S, Zeng X, Zhao L, Chen Z. Calpain inhibitors inhibit mitochondrial calpain activity to ameliorate apoptosis of cocultured myoblast. CHINESE J PHYSIOL 2022; 65:226-232. [DOI: 10.4103/0304-4920.359797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
41
|
Papatheodorou I, Makrecka-Kuka M, Kuka J, Liepinsh E, Dambrova M, Lazou A. Pharmacological activation of PPARβ/δ preserves mitochondrial respiratory function in ischemia/reperfusion via stimulation of fatty acid oxidation-linked respiration and PGC-1α/NRF-1 signaling. Front Endocrinol (Lausanne) 2022; 13:941822. [PMID: 36046786 PMCID: PMC9420994 DOI: 10.3389/fendo.2022.941822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury leads to significant impairment of cardiac function and remains the leading cause of morbidity and mortality worldwide. Activation of peroxisome proliferator-activated receptor β/δ (PPARβ/δ) confers cardioprotection via pleiotropic effects including antioxidant and anti-inflammatory actions; however, the underlying mechanisms are not yet fully elucidated. The aim of this study was to investigate the effect of PPARβ/δ activation on myocardial mitochondrial respiratory function and link this effect with cardioprotection after ischemia/reperfusion (I/R). For this purpose, rats were treated with the PPARβ/δ agonist GW0742 and/or antagonist GSK0660 in vivo. Mitochondrial respiration and ROS production rates were determined using high-resolution fluororespirometry. Activation of PPARβ/δ did not alter mitochondrial respiratory function in the healthy heart, however, inhibition of PPARβ/δ reduced fatty acid oxidation (FAO) and complex II-linked mitochondrial respiration and shifted the substrate dependence away from succinate-related energy production and towards NADH. Activation of PPARβ/δ reduced mitochondrial stress during in vitro anoxia/reoxygenation. Furthermore, it preserved FAO-dependent mitochondrial respiration and lowered ROS production at oxidative phosphorylation (OXPHOS)-dependent state during ex vivo I/R. PPARβ/δ activation was also followed by increased mRNA expression of components of FAO -linked respiration and of transcription factors governing mitochondrial homeostasis (carnitine palmitoyl transferase 1b and 2-CPT-1b and CPT-2, electron transfer flavoprotein dehydrogenase -ETFDH, peroxisome proliferator-activated receptor gamma co-activator 1 alpha- PGC-1α and nuclear respiratory factor 1-NRF-1). In conclusion, activation of PPARβ/δ stimulated both FAO-linked respiration and PGC-1α/NRF -1 signaling and preserved mitochondrial respiratory function during I/R. These effects are associated with reduced infarct size.
Collapse
Affiliation(s)
- Ioanna Papatheodorou
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Marina Makrecka-Kuka
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Janis Kuka
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Edgars Liepinsh
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Maija Dambrova
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
- Faculty of Pharmacy, Riga Stradins University, Riga, Latvia
| | - Antigone Lazou
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
- *Correspondence: Antigone Lazou,
| |
Collapse
|
42
|
FIsetin Preserves Interfibrillar Mitochondria to Protect Against Myocardial Ischemia-Reperfusion Injury. Cell Biochem Biophys 2021; 80:123-137. [PMID: 34392494 DOI: 10.1007/s12013-021-01026-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
According to our previous study, fisetin (3,3',4',7-tetrahydroxyflavone), a bioactive phytochemical (flavonol), reportedly showed cardioprotection against ischemia-reperfusion injury (IRI) by reducing oxidative stress and inhibiting glycogen synthase kinase 3β (GSK3β) [1]. GSK3β is said to exert a non-mitochondrial mediated cardioprotection; therefore, distinct mechanisms of GSK3β on the regulatory effect of mitochondria need to be addressed. The two distinct mitochondrial subpopulations in the heart, namely interfibrillar mitochondria (IFM) and subsarcolemmal mitochondria (SSM), respond differently to disease states. The current study aimed to understand the effect of fisetin on the subpopulation-specific preservation of IFM and SSM while rendering cardioprotection against ischemia reperfusion (I/R). Rats were pre-treated with fisetin (20 mg/kg) intraperitoneally, and IRI was induced using Langendorff isolated heart perfusion technique. Hemodynamic parameters were recorded, and the cardiac injury was assessed using infarct size (IS), lactate dehydrogenase (LDH), and creatine kinase (CK) levels. Subpopulation-specific mitochondrial preservation was evaluated by electron transport chain (ETC), catalase, superoxide dismutase (SOD), and glutathione (GSH) activities. The bioavailability of fisetin in IFM and SSM was measured using the fluorescence method. The ability of fisetin to bind directly to the mitochondrial complex-1 and activating it through donating electrons to FMN was studied using molecular docking studies and further validated by in vitro rotenone sensitivity assay. Cardioprotective effects exhibited by fisetin were mainly mediated through IFM preservation. Mitochondrial bioavailability of fisetin is more in IFM than SSM in both ex vivo and in vitro conditions. Fisetin increased mitochondrial ATP production in I/R insult hearts by activating ETC complex 1. Inhibition of complex 1 prevents the ATP-producing capacity of fisetin. Our results provide evidence that fisetin plays a protective role in myocardial IRI, possibly by preserving the functional activities of IFM.
Collapse
|
43
|
Feng Y, Huang W, Paul C, Liu X, Sadayappan S, Wang Y, Pauklin S. Mitochondrial nucleoid in cardiac homeostasis: bidirectional signaling of mitochondria and nucleus in cardiac diseases. Basic Res Cardiol 2021; 116:49. [PMID: 34392401 PMCID: PMC8364536 DOI: 10.1007/s00395-021-00889-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/20/2021] [Indexed: 01/11/2023]
Abstract
Metabolic function and energy production in eukaryotic cells are regulated by mitochondria, which have been recognized as the intracellular 'powerhouses' of eukaryotic cells for their regulation of cellular homeostasis. Mitochondrial function is important not only in normal developmental and physiological processes, but also in a variety of human pathologies, including cardiac diseases. An emerging topic in the field of cardiovascular medicine is the implication of mitochondrial nucleoid for metabolic reprogramming. This review describes the linear/3D architecture of the mitochondrial nucleoid (e.g., highly organized protein-DNA structure of nucleoid) and how it is regulated by a variety of factors, such as noncoding RNA and its associated R-loop, for metabolic reprogramming in cardiac diseases. In addition, we highlight many of the presently unsolved questions regarding cardiac metabolism in terms of bidirectional signaling of mitochondrial nucleoid and 3D chromatin structure in the nucleus. In particular, we explore novel techniques to dissect the 3D structure of mitochondrial nucleoid and propose new insights into the mitochondrial retrograde signaling, and how it regulates the nuclear (3D) chromatin structures in mitochondrial diseases.
Collapse
Affiliation(s)
- Yuliang Feng
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Old Road, University of Oxford, Oxford, OX3 7LD, UK
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, Regenerative Medicine Research, University of Cincinnati College of Medicine, 231 Albert Sabin Way, CincinnatiCincinnati, OH, 45267-0529, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, Regenerative Medicine Research, University of Cincinnati College of Medicine, 231 Albert Sabin Way, CincinnatiCincinnati, OH, 45267-0529, USA
| | - Xingguo Liu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Guangzhou Medical University, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Sakthivel Sadayappan
- Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, Regenerative Medicine Research, University of Cincinnati College of Medicine, 231 Albert Sabin Way, CincinnatiCincinnati, OH, 45267-0529, USA.
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Old Road, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
44
|
Single-Particle Tracking Method in Fluorescence Microscopy to Monitor Bioenergetic Responses of Individual Mitochondria. Methods Mol Biol 2021. [PMID: 34060039 DOI: 10.1007/978-1-0716-1266-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The spectroscopic methods commonly used to study mitochondria bioenergetics do not show the diversity of responses within a population of mitochondria (isolated or in a cell), and/or cannot measure individual dynamics. New methodological developments are necessary in order to improve quantitative and kinetic resolutions and eventually gain further insights on individual mitochondrial responses, such as studying activities of the mitochondrial permeability transition pore (mPTP ). The work reported herein is devoted to study responses of single mitochondria within a large population after isolation from cardiomyocytes. Mitochondria were preloaded with a commonly used membrane potential sensitive dye (TMRM), they are then deposited on a plasma-treated glass coverslip and subsequently energized or inhibited by additions of usual bioenergetics effectors. Responses were analyzed by fluorescence microscopy over few thousands of mitochondria simultaneously with a single organelle resolution. We report an automatic method to analyze each image of time-lapse stacks based on the TrackMate-ImageJ plug-in and specially made Python scripts. Images are processed to eliminate defects of illumination inhomogeneity, improving by at least two orders of magnitude the signal/noise ratio. This method enables us to follow the track of each mitochondrion within the observed field and monitor its fluorescence changes, with a time resolution of 400 ms, uninterrupted over the course of the experiment. Such methodological improvement is a prerequisite to further study the role of mPTP in single mitochondria during calcium transient loading.
Collapse
|
45
|
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
|
46
|
Wu P, Chen L, Cheng J, Pan Y, Zhu X, Bao L, Chu W, Zhang J. The miRNA expression profile directly reflects the energy metabolic differences between slow and fast muscle with nutritional regulation of the Chinese perch (Siniperca chuatsi). Comp Biochem Physiol A Mol Integr Physiol 2021; 259:111003. [PMID: 34118407 DOI: 10.1016/j.cbpa.2021.111003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/01/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
Fish skeletal muscles are composed of two distinct types, slow and fast muscles, and they play important roles in maintaining the body's movement and energy metabolism. The two types of muscle are easy to separate, so they are often used as the model system for studies on their physiological and functional characteristics. In this study, we revealed that the carbohydrate and lipid metabolic KEGG pathways are different between slow and fast muscles of Chinese perch with transcriptome analysis. In fast muscle, glucose metabolism was catabolic with higher glycolysis capacity, while in slow muscle, glucose metabolism was anabolic with more glycogen synthesis. In addition, oxidative metabolism in slow muscle was stronger than that in fast muscle. By analyzing the expression levels of 40 miRNAs involved in metabolism in the muscles of Chinese perch, 18 miRNAs were significantly upregulated and 7 were significantly downregulated in slow muscle compared with fast muscle. Based on functional enrichment analysis of their target genes, the differential expression levels of 17 miRNAs in slow and fast muscles were reflected in their carbohydrate and lipid metabolism. Among these, 15 miRNAs were associated with carbohydrate metabolism, and 6 miRNAs were associated with lipid metabolism. After 3 days of starvation, the expression levels of 15 miRNAs involved in glucose metabolism in fast and slow muscles increased. However, after 7 days of starvation, the mRNA levels of miR-22a, miR-23a, miR-133a-3p, miR-139, miR-143, miR-144, miR-181a and miR-206 decreased to basal levels. Our data suggest that the possible reason for the difference in glucose and lipid metabolism is that more miRNAs inhibit the expression of target genes in slow muscle.
Collapse
Affiliation(s)
- Ping Wu
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, Hunan 410003, China; Key Laboratory of Protein Chemistry and Fish Developmental Biology of Education Ministry of China, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Lin Chen
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, Hunan 410003, China; Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Jia Cheng
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, Hunan 410003, China; Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Yaxiong Pan
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, Hunan 410003, China; Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Xin Zhu
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, Hunan 410003, China; Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Lingsheng Bao
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, Hunan 410003, China; Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha 410022, China
| | - Wuying Chu
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, Hunan 410003, China; Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha 410022, China.
| | - Jianshe Zhang
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, Hunan 410003, China; Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha 410022, China.
| |
Collapse
|
47
|
Modulations of Cardiac Functions and Pathogenesis by Reactive Oxygen Species and Natural Antioxidants. Antioxidants (Basel) 2021; 10:antiox10050760. [PMID: 34064823 PMCID: PMC8150787 DOI: 10.3390/antiox10050760] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 01/11/2023] Open
Abstract
Homeostasis in the level of reactive oxygen species (ROS) in cardiac myocytes plays a critical role in regulating their physiological functions. Disturbance of balance between generation and removal of ROS is a major cause of cardiac myocyte remodeling, dysfunction, and failure. Cardiac myocytes possess several ROS-producing pathways, such as mitochondrial electron transport chain, NADPH oxidases, and nitric oxide synthases, and have endogenous antioxidation mechanisms. Cardiac Ca2+-signaling toolkit proteins, as well as mitochondrial functions, are largely modulated by ROS under physiological and pathological conditions, thereby producing alterations in contraction, membrane conductivity, cell metabolism and cell growth and death. Mechanical stresses under hypertension, post-myocardial infarction, heart failure, and valve diseases are the main causes for stress-induced cardiac remodeling and functional failure, which are associated with ROS-induced pathogenesis. Experimental evidence demonstrates that many cardioprotective natural antioxidants, enriched in foods or herbs, exert beneficial effects on cardiac functions (Ca2+ signal, contractility and rhythm), myocytes remodeling, inflammation and death in pathological hearts. The review may provide knowledge and insight into the modulation of cardiac pathogenesis by ROS and natural antioxidants.
Collapse
|
48
|
Yousefi R, Fornasiero EF, Cyganek L, Montoya J, Jakobs S, Rizzoli SO, Rehling P, Pacheu‐Grau D. Monitoring mitochondrial translation in living cells. EMBO Rep 2021; 22:e51635. [PMID: 33586863 PMCID: PMC8024989 DOI: 10.15252/embr.202051635] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/05/2021] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Mitochondria possess a small genome that codes for core subunits of the oxidative phosphorylation system and whose expression is essential for energy production. Information on the regulation and spatial organization of mitochondrial gene expression in the cellular context has been difficult to obtain. Here we devise an imaging approach to analyze mitochondrial translation within the context of single cells, by following the incorporation of clickable non-canonical amino acids. We apply this method to multiple cell types, including specialized cells such as cardiomyocytes and neurons, and monitor with spatial resolution mitochondrial translation in axons and dendrites. We also show that translation imaging allows to monitor mitochondrial protein expression in patient fibroblasts. Approaching mitochondrial translation with click chemistry opens new avenues to understand how mitochondrial biogenesis is integrated into the cellular context and can be used to assess mitochondrial gene expression in mitochondrial diseases.
Collapse
Affiliation(s)
- Roya Yousefi
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
| | - Eugenio F Fornasiero
- Department of Neuro‐ and Sensory PhysiologyUniversity Medical Center GöttingenGöttingenGermany
| | - Lukas Cyganek
- Clinic for Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
- German Center for Cardiovascular Research (DZHK)GöttingenGermany
| | - Julio Montoya
- Departamento de BioquímicaBiología Molecular y CelularUniversidad de ZaragozaZaragozaSpain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER)MadridSpain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón)Universidad de ZaragozaZaragozaSpain
| | - Stefan Jakobs
- Department of NanoBiophotonicsMax Planck Institute for Biophysical ChemistryGöttingenGermany
- Clinic of NeurologyUniversity Medical Center GöttingenGöttingenGermany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC)University of GoettingenGöttingenGermany
| | - Silvio O Rizzoli
- Department of Neuro‐ and Sensory PhysiologyUniversity Medical Center GöttingenGöttingenGermany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC)University of GoettingenGöttingenGermany
| | - Peter Rehling
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC)University of GoettingenGöttingenGermany
- Max‐Planck Institute for Biophysical ChemistryGöttingenGermany
| | - David Pacheu‐Grau
- Department of Cellular BiochemistryUniversity Medical Center GöttingenGöttingenGermany
| |
Collapse
|
49
|
"Empowering" Cardiac Cells via Stem Cell Derived Mitochondrial Transplantation- Does Age Matter? Int J Mol Sci 2021; 22:ijms22041824. [PMID: 33673127 PMCID: PMC7918132 DOI: 10.3390/ijms22041824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022] Open
Abstract
With cardiovascular diseases affecting millions of patients, new treatment strategies are urgently needed. The use of stem cell based approaches has been investigated during the last decades and promising effects have been achieved. However, the beneficial effect of stem cells has been found to being partly due to paracrine functions by alterations of their microenvironment and so an interesting field of research, the “stem- less” approaches has emerged over the last years using or altering the microenvironment, for example, via deletion of senescent cells, application of micro RNAs or by modifying the cellular energy metabolism via targeting mitochondria. Using autologous muscle-derived mitochondria for transplantations into the affected tissues has resulted in promising reports of improvements of cardiac functions in vitro and in vivo. However, since the targeted treatment group represents mainly elderly or otherwise sick patients, it is unclear whether and to what extent autologous mitochondria would exert their beneficial effects in these cases. Stem cells might represent better sources for mitochondria and could enhance the effect of mitochondrial transplantations. Therefore in this review we aim to provide an overview on aging effects of stem cells and mitochondria which might be important for mitochondrial transplantation and to give an overview on the current state in this field together with considerations worthwhile for further investigations.
Collapse
|
50
|
Bisaccia G, Ricci F, Gallina S, Di Baldassarre A, Ghinassi B. Mitochondrial Dysfunction and Heart Disease: Critical Appraisal of an Overlooked Association. Int J Mol Sci 2021; 22:ijms22020614. [PMID: 33435429 PMCID: PMC7827742 DOI: 10.3390/ijms22020614] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/03/2021] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
The myocardium is among the most energy-consuming tissues in the body, burning from 6 to 30 kg of ATP per day within the mitochondria, the so-called powerhouse of the cardiomyocyte. Although mitochondrial genetic disorders account for a small portion of cardiomyopathies, mitochondrial dysfunction is commonly involved in a broad spectrum of heart diseases, and it has been implicated in the development of heart failure via maladaptive circuits producing and perpetuating mitochondrial stress and energy starvation. In this bench-to-bedside review, we aimed to (i) describe the key functions of the mitochondria within the myocardium, including their role in ischemia/reperfusion injury and intracellular calcium homeostasis; (ii) examine the contribution of mitochondrial dysfunction to multiple cardiac disease phenotypes and their transition to heart failure; and (iii) discuss the rationale and current evidence for targeting mitochondrial function for the treatment of heart failure, including via sodium-glucose cotransporter 2 inhibitors.
Collapse
Affiliation(s)
- Giandomenico Bisaccia
- MIUR Department of Excellence, Department of Neuroscience, Imaging and Clinical Sciences, University “G.d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi, 11-66100 Chieti, Italy; (G.B.); (S.G.)
| | - Fabrizio Ricci
- MIUR Department of Excellence, Department of Neuroscience, Imaging and Clinical Sciences, University “G.d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi, 11-66100 Chieti, Italy; (G.B.); (S.G.)
- Department of Clinical Sciences, Lund University, E-205 02 Malmö, Sweden
- Casa di Cura Villa Serena, Città Sant’Angelo, 65013 Pescara, Italy
- Correspondence: ; Tel./Fax: +39-871-355-6922
| | - Sabina Gallina
- MIUR Department of Excellence, Department of Neuroscience, Imaging and Clinical Sciences, University “G.d’Annunzio” of Chieti-Pescara, Via Luigi Polacchi, 11-66100 Chieti, Italy; (G.B.); (S.G.)
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.B.); (B.G.)
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.B.); (B.G.)
| |
Collapse
|