1
|
Murphy E, Eisner DA. How does mitochondrial Ca2+ change during ischemia and reperfusion? Implications for activation of the permeability transition pore. J Gen Physiol 2025; 157:e202313520. [PMID: 39699565 DOI: 10.1085/jgp.202313520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/14/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
Cardiac ischemia followed by reperfusion results in cardiac cell death, which has been attributed to an increase of mitochondrial Ca2+ concentration, resulting in activation of the mitochondrial permeability transition pore (PTP). Evaluating this hypothesis requires understanding of the mechanisms responsible for control of mitochondrial Ca2+ in physiological conditions and how they are altered during both ischemia and reperfusion. Ca2+ influx is thought to occur through the mitochondrial Ca2+ uniporter (MCU). However, with deletion of the MCU, an increase in mitochondrial Ca2+ still occurs, suggesting an alternative Ca2+ influx mechanism during ischemia. There is less certainty about the mechanisms responsible for Ca2+ efflux, with contributions from both Ca2+/H+ exchange and a Na+-dependent Ca2+ efflux pathway. The molecular details of both mechanisms are not fully resolved. We discuss this and the contributions of both pathways to the accumulation of mitochondrial Ca2+ during ischemia and reperfusion. We further discuss the role of mitochondrial Ca2+ in activation of the PTP.
Collapse
Affiliation(s)
- Elizabeth Murphy
- Cardiac Physiology Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - David A Eisner
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Takeuchi A, Matsuoka S. A simulation study on the role of mitochondria-sarcoplasmic reticulum Ca 2+ interaction in cardiomyocyte energetics during exercise. J Physiol 2024. [PMID: 39387569 DOI: 10.1113/jp286054] [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: 06/16/2024] [Accepted: 08/15/2024] [Indexed: 10/15/2024] Open
Abstract
Previous studies demonstrated that the mitochondrial Ca2+ uniporter MCU and the Na+-Ca2+ exchanger NCLX exist in proximity to the sarcoplasmic reticulum (SR) ryanodine receptor RyR and the Ca2+ pump SERCA, respectively, creating a mitochondria-SR Ca2+ interaction. However, the physiological relevance of the mitochondria-SR Ca2+ interaction has remained unsolved. Furthermore, although mitochondrial Ca2+ has been proposed to be an important factor regulating mitochondrial energy metabolism, by activating NADH-producing dehydrogenases, the contribution of the Ca2+-dependent regulatory mechanisms to cellular functions under physiological conditions has been controversial. In this study, we constructed a new integrated model of human ventricular myocyte with excitation-contraction-energetics coupling and investigated systematically the contribution of mitochondria-SR Ca2+ interaction, especially focusing on cardiac energetics during dynamic workload transitions in exercise. Simulation analyses revealed that the spatial coupling of mitochondria and SR, particularly via mitochondrial Ca2+ uniport activity-RyR, was the primary determinant of mitochondrial Ca2+ concentration, and that the Ca2+-dependent regulatory mechanism facilitated mitochondrial NADH recovery during exercise and contributed to the stability of NADH in the workload transition by about 40%, while oxygen consumption rate and cytoplasmic ATP level were not influenced. We concluded that the mitochondria-SR Ca2+ interaction, created via the uneven distribution of Ca2+ handling proteins, optimizes the contribution of the mitochondrial Ca2+-dependent regulatory mechanism to stabilizing NADH during exercise. KEY POINTS: The mitochondrial Ca2+ uniporter protein MCU and the Na+-Ca2+ exchanger protein NCLX are reported to exist in proximity to the sarcoplasmic reticulum (SR) ryanodine receptor RyR and the Ca2+ pump SERCA, respectively, creating a mitochondria-SR Ca2+ interaction in cardiomyocytes. Mitochondrial Ca2+ (Ca2+ mit) has been proposed to be an important factor regulating mitochondrial energy metabolism, by activating NADH-producing dehydrogenases. Here we constructed an integrated model of a human ventricular myocyte with excitation-contraction-energetics coupling and investigated the role of the mitochondria-SR Ca2+ interaction in cardiac energetics during exercise. Simulation analyses revealed that the spatial coupling particularly via mitochondrial Ca2+ uniport activity-RyR is the primary determinant of Ca2+ mit concentration, and that the activation of NADH-producing dehydrogenases by Ca2+ mit contributes to NADH stability during exercise. The mitochondria-SR Ca2+ interaction optimizes the contribution of Ca2+ mit to the activation of NADH-producing dehydrogenases.
Collapse
Affiliation(s)
- Ayako Takeuchi
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences and Life Science Innovation Center, University of Fukui, Fukui, Japan
| | - Satoshi Matsuoka
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences and Life Science Innovation Center, University of Fukui, Fukui, Japan
| |
Collapse
|
3
|
Balderas E, Lee SHJ, Rai NK, Mollinedo DM, Duron HE, Chaudhuri D. Mitochondrial Calcium Regulation of Cardiac Metabolism in Health and Disease. Physiology (Bethesda) 2024; 39:0. [PMID: 38713090 PMCID: PMC11460536 DOI: 10.1152/physiol.00014.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024] Open
Abstract
Oxidative phosphorylation is regulated by mitochondrial calcium (Ca2+) in health and disease. In physiological states, Ca2+ enters via the mitochondrial Ca2+ uniporter and rapidly enhances NADH and ATP production. However, maintaining Ca2+ homeostasis is critical: insufficient Ca2+ impairs stress adaptation, and Ca2+ overload can trigger cell death. In this review, we delve into recent insights further defining the relationship between mitochondrial Ca2+ dynamics and oxidative phosphorylation. Our focus is on how such regulation affects cardiac function in health and disease, including heart failure, ischemia-reperfusion, arrhythmias, catecholaminergic polymorphic ventricular tachycardia, mitochondrial cardiomyopathies, Barth syndrome, and Friedreich's ataxia. Several themes emerge from recent data. First, mitochondrial Ca2+ regulation is critical for fuel substrate selection, metabolite import, and matching of ATP supply to demand. Second, mitochondrial Ca2+ regulates both the production and response to reactive oxygen species (ROS), and the balance between its pro- and antioxidant effects is key to how it contributes to physiological and pathological states. Third, Ca2+ exerts localized effects on the electron transport chain (ETC), not through traditional allosteric mechanisms but rather indirectly. These effects hinge on specific transporters, such as the uniporter or the Na+/Ca2+ exchanger, and may not be noticeable acutely, contributing differently to phenotypes depending on whether Ca2+ transporters are acutely or chronically modified. Perturbations in these novel relationships during disease states may either serve as compensatory mechanisms or exacerbate impairments in oxidative phosphorylation. Consequently, targeting mitochondrial Ca2+ holds promise as a therapeutic strategy for a variety of cardiac diseases characterized by contractile failure or arrhythmias.
Collapse
Affiliation(s)
- Enrique Balderas
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Sandra H J Lee
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Neeraj K Rai
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - David M Mollinedo
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Hannah E Duron
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
- Division of Cardiovascular Medicine, Department of Internal Medicine, Biochemistry, Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
4
|
Hasan P, Berezhnaya E, Rodríguez-Prados M, Weaver D, Bekeova C, Cartes-Saavedra B, Birch E, Beyer AM, Santos JH, Seifert EL, Elrod JW, Hajnóczky G. MICU1 and MICU2 control mitochondrial calcium signaling in the mammalian heart. Proc Natl Acad Sci U S A 2024; 121:e2402491121. [PMID: 39163336 PMCID: PMC11363308 DOI: 10.1073/pnas.2402491121] [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: 02/05/2024] [Accepted: 07/08/2024] [Indexed: 08/22/2024] Open
Abstract
Activating Ca2+-sensitive enzymes of oxidative metabolism while preventing calcium overload that leads to mitochondrial and cellular injury requires dynamic control of mitochondrial Ca2+ uptake. This is ensured by the mitochondrial calcium uptake (MICU)1/2 proteins that gate the pore of the mitochondrial calcium uniporter (mtCU). MICU1 is relatively sparse in the heart, and recent studies claimed the mammalian heart lacks MICU1 gating of mtCU. However, genetic models have not been tested. We find that MICU1 is present in a complex with MCU in nonfailing human hearts. Furthermore, using murine genetic models and pharmacology, we show that MICU1 and MICU2 control cardiac mitochondrial Ca2+ influx, and that MICU1 deletion alters cardiomyocyte mitochondrial calcium signaling and energy metabolism. MICU1 loss causes substantial compensatory changes in the mtCU composition and abundance, increased turnover of essential MCU regulator (EMRE) early on and, later, of MCU, that limit mitochondrial Ca2+ uptake and allow cell survival. Thus, both the primary consequences of MICU1 loss and the ensuing robust compensation highlight MICU1's relevance in the beating heart.
Collapse
Affiliation(s)
- Prottoy Hasan
- Department of Pathology and Genomic Medicine, MitoCare Center, Thomas Jefferson University, Philadelphia, PA19107
| | - Elena Berezhnaya
- Department of Pathology and Genomic Medicine, MitoCare Center, Thomas Jefferson University, Philadelphia, PA19107
| | - Macarena Rodríguez-Prados
- Department of Pathology and Genomic Medicine, MitoCare Center, Thomas Jefferson University, Philadelphia, PA19107
| | - David Weaver
- Department of Pathology and Genomic Medicine, MitoCare Center, Thomas Jefferson University, Philadelphia, PA19107
| | - Carmen Bekeova
- Department of Pathology and Genomic Medicine, MitoCare Center, Thomas Jefferson University, Philadelphia, PA19107
| | - Benjamin Cartes-Saavedra
- Department of Pathology and Genomic Medicine, MitoCare Center, Thomas Jefferson University, Philadelphia, PA19107
| | - Erin Birch
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI53226
| | - Andreas M. Beyer
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI53226
| | - Janine H. Santos
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, NC27709
| | - Erin L. Seifert
- Department of Pathology and Genomic Medicine, MitoCare Center, Thomas Jefferson University, Philadelphia, PA19107
| | - John W. Elrod
- Department of Cardiovascular Sciences, Aging+Cardiovascular Discovery Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140
| | - György Hajnóczky
- Department of Pathology and Genomic Medicine, MitoCare Center, Thomas Jefferson University, Philadelphia, PA19107
| |
Collapse
|
5
|
Fakuade FE, Hubricht D, Möller V, Sobitov I, Liutkute A, Döring Y, Seibertz F, Gerloff M, Pronto JRD, Haghighi F, Brandenburg S, Alhussini K, Ignatyeva N, Bonhoff Y, Kestel S, El-Essawi A, Jebran AF, Großmann M, Danner BC, Baraki H, Schmidt C, Sossalla S, Kutschka I, Bening C, Maack C, Linke WA, Heijman J, Lehnart SE, Kensah G, Ebert A, Mason FE, Voigt N. Impaired Intracellular Calcium Buffering Contributes to the Arrhythmogenic Substrate in Atrial Myocytes From Patients With Atrial Fibrillation. Circulation 2024; 150:544-559. [PMID: 38910563 PMCID: PMC11319087 DOI: 10.1161/circulationaha.123.066577] [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: 08/01/2023] [Accepted: 05/31/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Alterations in the buffering of intracellular Ca2+, for which myofilament proteins play a key role, have been shown to promote cardiac arrhythmia. It is interesting that although studies report atrial myofibrillar degradation in patients with persistent atrial fibrillation (persAF), the intracellular Ca2+ buffering profile in persAF remains obscure. Therefore, we aimed to investigate the intracellular buffering of Ca2+ and its potential arrhythmogenic role in persAF. METHODS Transmembrane Ca2+ fluxes (patch-clamp) and intracellular Ca2+ signaling (fluo-3-acetoxymethyl ester) were recorded simultaneously in myocytes from right atrial biopsies of sinus rhythm (Ctrl) and patients with persAF, alongside human atrial subtype induced pluripotent stem cell-derived cardiac myocytes (iPSC-CMs). Protein levels were quantified by immunoblotting of human atrial tissue and induced pluripotent stem cell-derived cardiac myocytes. Mouse whole heart and atrial electrophysiology were measured on a Langendorff system. RESULTS Cytosolic Ca2+ buffering was decreased in atrial myocytes of patients with persAF because of a depleted amount of Ca2+ buffers. In agreement, protein levels of selected Ca2+ binding myofilament proteins, including cTnC (cardiac troponin C), a major cytosolic Ca2+ buffer, were significantly lower in patients with persAF. Small interfering RNA (siRNA)-mediated knockdown of cTnC (si-cTNC) in atrial iPSC-CM phenocopied the reduced cytosolic Ca2+ buffering observed in persAF. Si-cTnC treated atrial iPSC-CM exhibited a higher predisposition to spontaneous Ca2+ release events and developed action potential alternans at low stimulation frequencies. Last, indirect reduction of cytosolic Ca2+ buffering using blebbistatin in an ex vivo mouse whole heart model increased vulnerability to tachypacing-induced atrial arrhythmia, validating the direct mechanistic link between impaired cytosolic Ca2+ buffering and atrial arrhythmogenesis. CONCLUSIONS Our findings suggest that loss of myofilament proteins, particularly reduced cTnC protein levels, causes diminished cytosolic Ca2+ buffering in persAF, thereby potentiating the occurrence of spontaneous Ca2+ release events and atrial fibrillation susceptibility. Strategies targeting intracellular buffering may represent a promising therapeutic lead in persAF management.
Collapse
Affiliation(s)
- Funsho E. Fakuade
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Dominik Hubricht
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Vanessa Möller
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Izzatullo Sobitov
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Aiste Liutkute
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Yannic Döring
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Fitzwilliam Seibertz
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Marcus Gerloff
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Julius Ryan D. Pronto
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Fereshteh Haghighi
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Sören Brandenburg
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Khaled Alhussini
- Department of Thoracic and Cardiovascular Surgery (K.A., C.B.), University Clinic Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg (K.A., C.B., C.M.), University Clinic Würzburg, Germany
| | - Nadezda Ignatyeva
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Yara Bonhoff
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Stefanie Kestel
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Aschraf El-Essawi
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
- Department of Thoracic and Cardiovascular Surgery, Klinikum Braunschweig, Germany (A.E.-E.)
| | - Ahmad Fawad Jebran
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Marius Großmann
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Bernhard C. Danner
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Hassina Baraki
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Constanze Schmidt
- Department of Cardiology, University Hospital Heidelberg, Germany (C.S.)
- German Center for Cardiovascular Research Partner Site Heidelberg/Mannheim, Heidelberg University (C.S.)
| | - Samuel Sossalla
- Department of Cardiology, University Hospital Giessen & Kerckhoff Clinic, Germany (S.S.)
- Department of Cardiology, Bad Nauheim & German Center for Cardiovascular Research Partner Site Rhine-Main, Germany (S.S.)
| | - Ingo Kutschka
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Constanze Bening
- Department of Thoracic and Cardiovascular Surgery (K.A., C.B.), University Clinic Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg (K.A., C.B., C.M.), University Clinic Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center Würzburg (K.A., C.B., C.M.), University Clinic Würzburg, Germany
| | - Wolfgang A. Linke
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
- Institute of Physiology II, University of Münster, Germany (W.A.L.)
| | - Jordi Heijman
- Gottfried Schatz Research Center, Division of Medical Physics and Biophysics, Medical University of Graz, Austria (J.H.)
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands (J.H.)
| | - Stephan E. Lehnart
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - George Kensah
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Antje Ebert
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Fleur E. Mason
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Niels Voigt
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| |
Collapse
|
6
|
Eisner D, Neher E, Taschenberger H, Smith G. Physiology of intracellular calcium buffering. Physiol Rev 2023; 103:2767-2845. [PMID: 37326298 PMCID: PMC11550887 DOI: 10.1152/physrev.00042.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/08/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023] Open
Abstract
Calcium signaling underlies much of physiology. Almost all the Ca2+ in the cytoplasm is bound to buffers, with typically only ∼1% being freely ionized at resting levels in most cells. Physiological Ca2+ buffers include small molecules and proteins, and experimentally Ca2+ indicators will also buffer calcium. The chemistry of interactions between Ca2+ and buffers determines the extent and speed of Ca2+ binding. The physiological effects of Ca2+ buffers are determined by the kinetics with which they bind Ca2+ and their mobility within the cell. The degree of buffering depends on factors such as the affinity for Ca2+, the Ca2+ concentration, and whether Ca2+ ions bind cooperatively. Buffering affects both the amplitude and time course of cytoplasmic Ca2+ signals as well as changes of Ca2+ concentration in organelles. It can also facilitate Ca2+ diffusion inside the cell. Ca2+ buffering affects synaptic transmission, muscle contraction, Ca2+ transport across epithelia, and the killing of bacteria. Saturation of buffers leads to synaptic facilitation and tetanic contraction in skeletal muscle and may play a role in inotropy in the heart. This review focuses on the link between buffer chemistry and function and how Ca2+ buffering affects normal physiology and the consequences of changes in disease. As well as summarizing what is known, we point out the many areas where further work is required.
Collapse
Affiliation(s)
- David Eisner
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Erwin Neher
- Membrane Biophysics Laboratory, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Godfrey Smith
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
7
|
Lee SH, Duron HE, Chaudhuri D. Beyond the TCA cycle: new insights into mitochondrial calcium regulation of oxidative phosphorylation. Biochem Soc Trans 2023; 51:1661-1673. [PMID: 37641565 PMCID: PMC10508640 DOI: 10.1042/bst20230012] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
While mitochondria oxidative phosphorylation is broadly regulated, the impact of mitochondrial Ca2+ on substrate flux under both physiological and pathological conditions is increasingly being recognized. Under physiologic conditions, mitochondrial Ca2+ enters through the mitochondrial Ca2+ uniporter and boosts ATP production. However, maintaining Ca2+ homeostasis is crucial as too little Ca2+ inhibits adaptation to stress and Ca2+ overload can trigger cell death. In this review, we discuss new insights obtained over the past several years expanding the relationship between mitochondrial Ca2+ and oxidative phosphorylation, with most data obtained from heart, liver, or skeletal muscle. Two new themes are emerging. First, beyond boosting ATP synthesis, Ca2+ appears to be a critical determinant of fuel substrate choice between glucose and fatty acids. Second, Ca2+ exerts local effects on the electron transport chain indirectly, not via traditional allosteric mechanisms. These depend critically on the transporters involved, such as the uniporter or the Na+-Ca2+ exchanger. Alteration of these new relationships during disease can be either compensatory or harmful and suggest that targeting mitochondrial Ca2+ may be of therapeutic benefit during diseases featuring impairments in oxidative phosphorylation.
Collapse
Affiliation(s)
- Sandra H. Lee
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - Hannah E. Duron
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, Biochemistry, Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
8
|
Xu H, Yu W, Sun M, Bi Y, Wu NN, Zhou Y, Yang Q, Zhang M, Ge J, Zhang Y, Ren J. Syntaxin17 contributes to obesity cardiomyopathy through promoting mitochondrial Ca 2+ overload in a Parkin-MCUb-dependent manner. Metabolism 2023; 143:155551. [PMID: 36948287 DOI: 10.1016/j.metabol.2023.155551] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
OBJECTIVE Uncorrected obesity is accompanied by unfavorable structural and functional changes in the heart, known as obesity cardiomyopathy. Recent evidence has revealed a crucial role for mitochondria-associated endoplasmic reticulum membranes (MAMs) in obesity-induced cardiac complication. Syntaxin 17 (STX17) serves as a scaffolding molecule localized on MAMs although its role in obesity heart complication remains elusive. METHODS AND MATERIALS This study examined the role of STX17 in MAMs and mitochondrial Ca2+ homeostasis in HFD-induced obesity cardiomyopathy using tamoxifen-induced cardiac-specific STX17 knockout (STX17cko) and STX17 overexpression mice using intravenously delivered recombinant adeno-associated virus serotype-9 (AAV9-cTNT-STX17). RESULTS STX17 levels were significantly elevated in plasma from obese patients and heart tissues of HFD-fed mice. Our data revealed that cardiac STX17 knockout alleviated cardiac remodeling and dysfunction in obese hearts without eliciting any notable effect itself, while STX17 overexpression aggravated cardiac dysfunction in obese mice. STX17 deletion and STX17 overexpression annihilated and aggravated, respectively, HFD-induced oxidative stress (O2- production) and mitochondrial injury in the heart. Furthermore, STX17 transfection facilitated obesity-induced MAMs formation in cardiomyocytes and evoked excess mitochondrial Ca2+ influx, dependent upon interaction with mitochondrial Ca2+ uniporter dominant negative β (MCUb) through Habc domain. Our data also suggested that STX17 promoted ubiquitination and degradation of MCUb through the E3 ligase parkin in the face of palmitate challenging. CONCLUSION Taken together, our results identified a novel role for STX17 in facilitating obesity-induced MAMs formation, and subsequently mitochondrial Ca2+ overload, mitochondrial O2- accumulation, lipid peroxidation, resulting in cardiac impairment. Our findings denoted therapeutic promises of targeting STX17 in obesity.
Collapse
Affiliation(s)
- Haixia Xu
- Department of Cardiology, Zhongshan Hospital, Fudan University, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Department of Cardiology, Affiliated Hospital of Nantong University, Jiangsu, 226001, China
| | - Wenjun Yu
- Department of Cardiology, Zhongshan Hospital, Fudan University, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Mingming Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yaguang Bi
- Department of Cardiology, Zhongshan Hospital, Fudan University, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Ne N Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Mengjiao Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
9
|
Increased Mitochondrial Calcium Fluxes in Hypertrophic Right Ventricular Cardiomyocytes from a Rat Model of Pulmonary Artery Hypertension. Life (Basel) 2023; 13:life13020540. [PMID: 36836897 PMCID: PMC9967871 DOI: 10.3390/life13020540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Pulmonary artery hypertension causes right ventricular hypertrophy which rapidly progresses to heart failure with underlying cardiac mitochondrial dysfunction. Prior to failure, there are alterations in cytosolic Ca2+ handling that might impact mitochondrial function in the compensatory phase of RV hypertrophy. Our aims, therefore, were (i) to measure beat-to-beat mitochondrial Ca2+ fluxes, and (ii) to determine mitochondrial abundance and function in non-failing, hypertrophic cardiomyocytes. Male Wistar rats were injected with either saline (CON) or monocrotaline (MCT) to induce pulmonary artery hypertension and RV hypertrophy after four weeks. Cytosolic Ca2+ ([Ca2+]cyto) transients were obtained in isolated right ventricular (RV) cardiomyocytes, and mitochondrial Ca2+ ([Ca2+]mito) was recorded in separate RV cardiomyocytes. The distribution and abundance of key proteins was determined using confocal and stimulated emission depletion (STED) microscopy. The RV mitochondrial function was also assessed in RV homogenates using oxygraphy. The MCT cardiomyocytes had increased area, larger [Ca2+]cyto transients, increased Ca2+ store content, and faster trans-sarcolemmal Ca2+ extrusion relative to CON. The MCT cardiomyocytes also had larger [Ca2+]mito transients. STED images detected increased mitochondrial protein abundance (TOM20 clusters per μm2) in MCT, yet no difference was found when comparing mitochondrial respiration and membrane potential between the groups. We suggest that the larger [Ca2+]mito transients compensate to match ATP supply to the increased energy demands of hypertrophic cardiomyocytes.
Collapse
|
10
|
Keefe JA, Moore OM, Ho KS, Wehrens XHT. Role of Ca 2+ in healthy and pathologic cardiac function: from normal excitation-contraction coupling to mutations that cause inherited arrhythmia. Arch Toxicol 2023; 97:73-92. [PMID: 36214829 PMCID: PMC10122835 DOI: 10.1007/s00204-022-03385-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/15/2022] [Indexed: 01/19/2023]
Abstract
Calcium (Ca2+) ions are a key second messenger involved in the rhythmic excitation and contraction of cardiomyocytes throughout the heart. Proper function of Ca2+-handling proteins is required for healthy cardiac function, whereas disruption in any of these can cause cardiac arrhythmias. This comprehensive review provides a broad overview of the roles of Ca2+-handling proteins and their regulators in healthy cardiac function and the mechanisms by which mutations in these proteins contribute to inherited arrhythmias. Major Ca2+ channels and Ca2+-sensitive regulatory proteins involved in cardiac excitation-contraction coupling are discussed, with special emphasis on the function of the RyR2 macromolecular complex. Inherited arrhythmia disorders including catecholaminergic polymorphic ventricular tachycardia, long QT syndrome, Brugada syndrome, short QT syndrome, and arrhythmogenic right-ventricular cardiomyopathy are discussed with particular emphasis on subtypes caused by mutations in Ca2+-handling proteins.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Oliver M Moore
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kevin S Ho
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA. .,Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Center for Space Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
11
|
Voglhuber J, Holzer M, Radulović S, Thai PN, Djalinac N, Matzer I, Wallner M, Bugger H, Zirlik A, Leitinger G, Dedkova EN, Bers DM, Ljubojevic-Holzer S. Functional remodelling of perinuclear mitochondria alters nucleoplasmic Ca 2+ signalling in heart failure. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210320. [PMID: 36189813 PMCID: PMC9527904 DOI: 10.1098/rstb.2021.0320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/23/2022] [Indexed: 11/12/2022] Open
Abstract
Mitochondrial dysfunction in cardiomyocytes is a hallmark of heart failure development. Although initial studies recognized the importance of different mitochondrial subpopulations, there is a striking lack of direct comparison of intrafibrillar (IF) versus perinuclear (PN) mitochondria during the development of HF. Here, we use multiple approaches to examine the morphology and functional properties of IF versus PN mitochondria in pressure overload-induced cardiac remodelling in mice, and in non-failing and failing human cardiomyocytes. We demonstrate that PN mitochondria from failing cardiomyocytes are more susceptible to depolarization of mitochondrial membrane potential, reactive oxygen species generation and impairment in Ca2+ uptake compared with IF mitochondria at baseline and under physiological stress protocol. We also demonstrate, for the first time to our knowledge, that under normal conditions PN mitochondrial Ca2+ uptake shapes nucleoplasmic Ca2+ transients (CaTs) and limits nucleoplasmic Ca2+ loading. The loss of PN mitochondrial Ca2+ buffering capacity translates into increased nucleoplasmic CaTs and may explain disproportionate rise in nucleoplasmic [Ca2+] in failing cardiomyocytes at increased stimulation frequencies. Therefore, a previously unidentified benefit of restoring the mitochondrial Ca2+ uptake may be normalization of nuclear Ca2+ signalling and alleviation of altered excitation-transcription, which could be an important therapeutic approach to prevent adverse cardiac remodelling. 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)
- Julia Voglhuber
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Michael Holzer
- BioTechMed-Graz, Graz, Austria
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Snježana Radulović
- Research Unit Electron Microscopic Techniques, Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Phung N. Thai
- Department of Internal Medicine, Cardiovascular Medicine, University of California Davis, Davis, CA, USA
| | - Natasa Djalinac
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Ingrid Matzer
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Markus Wallner
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, PA, USA
| | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Andreas Zirlik
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Gerd Leitinger
- Research Unit Electron Microscopic Techniques, Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Elena N. Dedkova
- Department of Pharmacology, University of California Davis, Davis, CA, USA
- Department of Molecular Biosciences, University of California Davis, Davis, CA, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
12
|
Shi X, Jiang X, Chen C, Zhang Y, Sun X. The interconnections between the microtubules and mitochondrial networks in cardiocerebrovascular diseases: Implications for therapy. Pharmacol Res 2022; 184:106452. [PMID: 36116706 DOI: 10.1016/j.phrs.2022.106452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
Abstract
Microtubules, a highly dynamic cytoskeleton, participate in many cellular activities including mechanical support, organelles interactions, and intracellular trafficking. Microtubule organization can be regulated by modification of tubulin subunits, microtubule-associated proteins (MAPs) or agents modulating microtubule assembly. Increasing studies demonstrate that microtubule disorganization correlates with various cardiocerebrovascular diseases including heart failure and ischemic stroke. Microtubules also mediate intracellular transport as well as intercellular transfer of mitochondria, a power house in cells which produce ATP for various physiological activities such as cardiac mechanical function. It is known to all that both microtubules and mitochondria participate in the progression of cancer and Parkinson's disease. However, the interconnections between the microtubules and mitochondrial networks in cardiocerebrovascular diseases remain unclear. In this paper, we will focus on the roles of microtubules in cardiocerebrovascular diseases, and discuss the interplay of mitochondria and microtubules in disease development and treatment. Elucidation of these issues might provide significant diagnostic value as well as potential targets for cardiocerebrovascular diseases.
Collapse
Affiliation(s)
- Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| | - Xuan Jiang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Congwei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yu Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| |
Collapse
|
13
|
Ren L, Gopireddy RR, Perkins G, Zhang H, Timofeyev V, Lyu Y, Diloretto DA, Trinh P, Sirish P, Overton JL, Xu W, Grainger N, Xiang YK, Dedkova EN, Zhang XD, Yamoah EN, Navedo MF, Thai PN, Chiamvimonvat N. Disruption of mitochondria-sarcoplasmic reticulum microdomain connectomics contributes to sinus node dysfunction in heart failure. Proc Natl Acad Sci U S A 2022; 119:e2206708119. [PMID: 36044551 PMCID: PMC9456763 DOI: 10.1073/pnas.2206708119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
The sinoatrial node (SAN), the leading pacemaker region, generates electrical impulses that propagate throughout the heart. SAN dysfunction with bradyarrhythmia is well documented in heart failure (HF). However, the underlying mechanisms are not completely understood. Mitochondria are critical to cellular processes that determine the life or death of the cell. The release of Ca2+ from the ryanodine receptors 2 (RyR2) on the sarcoplasmic reticulum (SR) at mitochondria-SR microdomains serves as the critical communication to match energy production to meet metabolic demands. Therefore, we tested the hypothesis that alterations in the mitochondria-SR connectomics contribute to SAN dysfunction in HF. We took advantage of a mouse model of chronic pressure overload-induced HF by transverse aortic constriction (TAC) and a SAN-specific CRISPR-Cas9-mediated knockdown of mitofusin-2 (Mfn2), the mitochondria-SR tethering GTPase protein. TAC mice exhibited impaired cardiac function with HF, cardiac fibrosis, and profound SAN dysfunction. Ultrastructural imaging using electron microscope (EM) tomography revealed abnormal mitochondrial structure with increased mitochondria-SR distance. The expression of Mfn2 was significantly down-regulated and showed reduced colocalization with RyR2 in HF SAN cells. Indeed, SAN-specific Mfn2 knockdown led to alterations in the mitochondria-SR microdomains and SAN dysfunction. Finally, disruptions in the mitochondria-SR microdomains resulted in abnormal mitochondrial Ca2+ handling, alterations in localized protein kinase A (PKA) activity, and impaired mitochondrial function in HF SAN cells. The current study provides insights into the role of mitochondria-SR microdomains in SAN automaticity and possible therapeutic targets for SAN dysfunction in HF patients.
Collapse
Affiliation(s)
- Lu Ren
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305
| | | | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093
| | - Hao Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305
| | - Valeriy Timofeyev
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Yankun Lyu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Daphne A. Diloretto
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Pauline Trinh
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - James L. Overton
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Wilson Xu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Nathan Grainger
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616
| | - Yang K. Xiang
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Elena N. Dedkova
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616
| | - Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, NV 89557
| | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Phung N. Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
- Department of Physiology and Cell Biology, University of Nevada, Reno, NV 89557
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
- Department of Pharmacology, University of California, Davis, CA 95616
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655
| |
Collapse
|
14
|
Reggiani C, Marcucci L. A controversial issue: Can mitochondria modulate cytosolic calcium and contraction of skeletal muscle fibers? J Gen Physiol 2022; 154:e202213167. [PMID: 35849108 PMCID: PMC9297197 DOI: 10.1085/jgp.202213167] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Mitochondria are characterized by a high capacity to accumulate calcium thanks to the electrochemical gradient created by the extrusion of protons in the respiratory chain. Thereby calcium can enter crossing the inner mitochondrial membrane via MCU complex, a high-capacity, low-affinity transport mechanism. Calcium uptake serves numerous purposes, among them the regulation of three dehydrogenases of the citric cycle, apoptosis via permeability transition, and, in some cell types, modulation of cytosolic calcium transients. This Review is focused on mitochondrial calcium uptake in skeletal muscle fibers and aims to reanalyze its functional impact. In particular, we ask whether mitochondrial calcium uptake is relevant for the control of cytosolic calcium transients and therefore of contractile performance. Recent data suggest that this may be the case, at least in particular conditions, as modified expression of MCU complex subunits or of proteins involved in mitochondrial dynamics and ablation of the main cytosolic calcium buffer, parvalbumin.
Collapse
Affiliation(s)
- Carlo Reggiani
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Science and Research Center Koper, Institute for Kinesiology Research, Koper, Slovenia
| | - Lorenzo Marcucci
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Center for Biosystems Dynamics Research, RIKEN, Suita, Japan
| |
Collapse
|
15
|
VDAC2 as a novel target for heart failure: Ca2+ at the sarcomere, mitochondria and SR. Cell Calcium 2022; 104:102586. [DOI: 10.1016/j.ceca.2022.102586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 11/22/2022]
|
16
|
Lyu Y, Thai PN, Ren L, Timofeyev V, Jian Z, Park S, Ginsburg KS, Overton J, Bossuyt J, Bers DM, Yamoah EN, Chen-Izu Y, Chiamvimonvat N, Zhang XD. Beat-to-beat dynamic regulation of intracellular pH in cardiomyocytes. iScience 2022; 25:103624. [PMID: 35005560 PMCID: PMC8718820 DOI: 10.1016/j.isci.2021.103624] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/10/2021] [Accepted: 12/10/2021] [Indexed: 11/20/2022] Open
Abstract
The mammalian heart beats incessantly with rhythmic mechanical activities generating acids that need to be buffered to maintain a stable intracellular pH (pHi) for normal cardiac function. Even though spatial pHi non-uniformity in cardiomyocytes has been documented, it remains unknown how pHi is regulated to match the dynamic cardiac contractions. Here, we demonstrated beat-to-beat intracellular acidification, termed pHi transients, in synchrony with cardiomyocyte contractions. The pHi transients are regulated by pacing rate, Cl-/HCO3 - transporters, pHi buffering capacity, and β-adrenergic signaling. Mitochondrial electron-transport chain inhibition attenuates the pHi transients, implicating mitochondrial activity in sculpting the pHi regulation. The pHi transients provide dynamic alterations of H+ transport required for ATP synthesis, and a decrease in pHi may serve as a negative feedback to cardiac contractions. Current findings dovetail with the prevailing three known dynamic systems, namely electrical, Ca2+, and mechanical systems, and may reveal broader features of pHi handling in excitable cells.
Collapse
Affiliation(s)
- Yankun Lyu
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Phung N. Thai
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Lu Ren
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Valeriy Timofeyev
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Zhong Jian
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Seojin Park
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV 89557, USA
| | - Kenneth S. Ginsburg
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - James Overton
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV 89557, USA
| | - Ye Chen-Izu
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| | - Xiao-Dong Zhang
- Department of Internal Medicine, University of California, Davis, Davis, CA 95616, USA
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| |
Collapse
|
17
|
Takeuchi A, Matsuoka S. Physiological and Pathophysiological Roles of Mitochondrial Na +-Ca 2+ Exchanger, NCLX, in Hearts. Biomolecules 2021; 11:biom11121876. [PMID: 34944520 PMCID: PMC8699148 DOI: 10.3390/biom11121876] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022] Open
Abstract
It has been over 10 years since SLC24A6/SLC8B1, coding the Na+/Ca2+/Li+ exchanger (NCLX), was identified as the gene responsible for mitochondrial Na+-Ca2+ exchange, a major Ca2+ efflux system in cardiac mitochondria. This molecular identification enabled us to determine structure–function relationships, as well as physiological/pathophysiological contributions, and our understandings have dramatically increased. In this review, we provide an overview of the recent achievements in relation to NCLX, focusing especially on its heart-specific characteristics, biophysical properties, and spatial distribution in cardiomyocytes, as well as in cardiac mitochondria. In addition, we discuss the roles of NCLX in cardiac functions under physiological and pathophysiological conditions—the generation of rhythmicity, the energy metabolism, the production of reactive oxygen species, and the opening of mitochondrial permeability transition pores.
Collapse
Affiliation(s)
- Ayako Takeuchi
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan;
- Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan
- Correspondence: ; Tel.: +81-776-61-8311
| | - Satoshi Matsuoka
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan;
- Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan
| |
Collapse
|
18
|
Manousek J, Kala P, Lokaj P, Ondrus T, Helanova K, Miklikova M, Brazdil V, Tomandlova M, Parenica J, Pavkova Goldbergova M, Hlasensky J. Oxidative Stress in Takotsubo Syndrome-Is It Essential for an Acute Attack? Indirect Evidences Support Multisite Impact Including the Calcium Overload-Energy Failure Hypothesis. Front Cardiovasc Med 2021; 8:732708. [PMID: 34738019 PMCID: PMC8562109 DOI: 10.3389/fcvm.2021.732708] [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] [Received: 06/29/2021] [Accepted: 09/16/2021] [Indexed: 12/28/2022] Open
Abstract
Indirect evidences in reviews and case reports on Takotsubo syndrome (TTS) support the fact that the existence of oxidative stress (OS) might be its common feature in the pre-acute stage. The sources of OS are exogenous (environmental factors including pharmacological and toxic influences) and endogenous, the combination of both may be present, and they are being discussed in detail. OS is associated with several pathological conditions representing TTS comorbidities and triggers. The dominant source of OS electrones are mitochondria. Our analysis of drug therapy related to acute TTS shows many interactions, e.g., cytostatics and glucocorticoids with mitochondrial cytochrome P450 and other enzymes important for OS. One of the most frequently discussed mechanisms in TTS is the effect of catecholamines on myocardium. Yet, their metabolic influence is neglected. OS is associated with the oxidation of catecholamines leading to the synthesis of their oxidized forms - aminochromes. Under pathological conditions, this pathway may dominate. There are evidences of interference between OS, catecholamine/aminochrome effects, their metabolism and antioxidant protection. The OS offensive may cause fast depletion of antioxidant protection including the homocystein-methionine system, whose activity decreases with age. The alteration of effector subcellular structures (mitochondria, sarco/endoplasmic reticulum) and subsequent changes in cellular energetics and calcium turnover may also occur and lead to the disruption of cellular function, including neurons and cardiomyocytes. On the organ level (nervous system and heart), neurocardiogenic stunning may occur. The effects of OS correspond to the effect of high doses of catecholamines in the experiment. Intensive OS might represent "conditio sine qua non" for this acute clinical condition. TTS might be significantly more complex pathology than currently perceived so far.
Collapse
Affiliation(s)
- Jan Manousek
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
| | - Petr Kala
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Petr Lokaj
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Tomas Ondrus
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Katerina Helanova
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marie Miklikova
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
| | - Vojtech Brazdil
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marie Tomandlova
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Jiri Parenica
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | | | - Jiri Hlasensky
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czechia
- Department of Internal Medicine and Cardiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
19
|
Hamilton S, Terentyeva R, Perger F, Hernández Orengo B, Martin B, Gorr MW, Belevych AE, Clements RT, Györke S, Terentyev D. MCU overexpression evokes disparate dose-dependent effects on mito-ROS and spontaneous Ca 2+ release in hypertrophic rat cardiomyocytes. Am J Physiol Heart Circ Physiol 2021; 321:H615-H632. [PMID: 34415186 PMCID: PMC8794228 DOI: 10.1152/ajpheart.00126.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 12/11/2022]
Abstract
Cardiac dysfunction in heart failure (HF) and diabetic cardiomyopathy (DCM) is associated with aberrant intracellular Ca2+ handling and impaired mitochondrial function accompanied with reduced mitochondrial calcium concentration (mito-[Ca2+]). Pharmacological or genetic facilitation of mito-Ca2+ uptake was shown to restore Ca2+ transient amplitude in DCM and HF, improving contractility. However, recent reports suggest that pharmacological enhancement of mito-Ca2+ uptake can exacerbate ryanodine receptor-mediated spontaneous sarcoplasmic reticulum (SR) Ca2+ release in ventricular myocytes (VMs) from diseased animals, increasing propensity to stress-induced ventricular tachyarrhythmia. To test whether chronic recovery of mito-[Ca2+] restores systolic Ca2+ release without adverse effects in diastole, we overexpressed mitochondrial Ca2+ uniporter (MCU) in VMs from male rat hearts with hypertrophy induced by thoracic aortic banding (TAB). Measurement of mito-[Ca2+] using genetic probe mtRCamp1h revealed that mito-[Ca2+] in TAB VMs paced at 2 Hz under β-adrenergic stimulation is lower compared with shams. Adenoviral 2.5-fold MCU overexpression in TAB VMs fully restored mito-[Ca2+]. However, it failed to improve cytosolic Ca2+ handling and reduce proarrhythmic spontaneous Ca2+ waves. Furthermore, mitochondrial-targeted genetic probes MLS-HyPer7 and OMM-HyPer revealed a significant increase in emission of reactive oxygen species (ROS) in TAB VMs with 2.5-fold MCU overexpression. Conversely, 1.5-fold MCU overexpression in TABs, that led to partial restoration of mito-[Ca2+], reduced mitochondria-derived reactive oxygen species (mito-ROS) and spontaneous Ca2+ waves. Our findings emphasize the key role of elevated mito-ROS in disease-related proarrhythmic Ca2+ mishandling. These data establish nonlinear mito-[Ca2+]/mito-ROS relationship, whereby partial restoration of mito-[Ca2+] in diseased VMs is protective, whereas further enhancement of MCU-mediated Ca2+ uptake exacerbates damaging mito-ROS emission.NEW & NOTEWORTHY Defective intracellular Ca2+ homeostasis and aberrant mitochondrial function are common features in cardiac disease. Here, we directly compared potential benefits of mito-ROS scavenging and restoration of mito-Ca2+ uptake by overexpressing MCU in ventricular myocytes from hypertrophic rat hearts. Experiments using novel mito-ROS and Ca2+ biosensors demonstrated that mito-ROS scavenging rescued both cytosolic and mito-Ca2+ homeostasis, whereas moderate and high MCU overexpression demonstrated disparate effects on mito-ROS emission, with only a moderate increase in MCU being beneficial.
Collapse
MESH Headings
- Adrenergic beta-Agonists/pharmacology
- Animals
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/pathology
- Arrhythmias, Cardiac/physiopathology
- Biosensing Techniques
- Calcium/metabolism
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Signaling/drug effects
- Cells, Cultured
- Disease Models, Animal
- Heart Rate
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Microscopy, Confocal
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/genetics
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocardial Contraction
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Up-Regulation
- Ventricular Function, Left
- Ventricular Remodeling
- Rats
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Fruzsina Perger
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Benjamín Hernández Orengo
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Benjamin Martin
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Matthew W Gorr
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
- College of Nursing, The Ohio State University, Columbus, Ohio
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Richard T Clements
- Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island
| | - Sandor Györke
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| |
Collapse
|
20
|
Chaanine AH. Metabolic Remodeling and Implicated Calcium and Signal Transduction Pathways in the Pathogenesis of Heart Failure. Int J Mol Sci 2021; 22:ijms221910579. [PMID: 34638917 PMCID: PMC8508915 DOI: 10.3390/ijms221910579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/20/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
The heart is an organ with high-energy demands in which the mitochondria are most abundant. They are considered the powerhouse of the cell and occupy a central role in cellular metabolism. The intermyofibrillar mitochondria constitute the majority of the three-mitochondrial subpopulations in the heart. They are also considered to be the most important in terms of their ability to participate in calcium and cellular signaling, which are critical for the regulation of mitochondrial function and adenosine triphosphate (ATP) production. This is because they are located in very close proximity with the endoplasmic reticulum (ER), and for the presence of tethering complexes enabling interorganelle crosstalk via calcium signaling. Calcium is an important second messenger that regulates mitochondrial function. It promotes ATP production and cellular survival under physiological changes in cardiac energetic demand. This is accomplished in concert with signaling pathways that regulate both calcium cycling and mitochondrial function. Perturbations in mitochondrial homeostasis and metabolic remodeling occupy a central role in the pathogenesis of heart failure. In this review we will discuss perturbations in ER-mitochondrial crosstalk and touch on important signaling pathways and molecular mechanisms involved in the dysregulation of calcium homeostasis and mitochondrial function in heart failure.
Collapse
Affiliation(s)
- Antoine H. Chaanine
- Department of Medicine, Heart and Vascular Institute, Tulane University, New Orleans, LA 70112, USA; ; Tel.: +1-(504)-988-1612
- Department of Physiology, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
21
|
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: 4.0] [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
|
22
|
Hamilton S, Terentyeva R, Clements RT, Belevych AE, Terentyev D. Sarcoplasmic reticulum-mitochondria communication; implications for cardiac arrhythmia. J Mol Cell Cardiol 2021; 156:105-113. [PMID: 33857485 DOI: 10.1016/j.yjmcc.2021.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/15/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022]
Abstract
Sudden cardiac death due to ventricular tachyarrhythmias remains the major cause of mortality in the world. Heart failure, diabetic cardiomyopathy, old age-related cardiac dysfunction and inherited disorders are associated with enhanced propensity to malignant cardiac arrhythmias. Both defective mitochondrial function and abnormal intracellular Ca2+ homeostasis have been established as the key contributing factors in the pathophysiology and arrhythmogenesis in these conditions. This article reviews current advances in understanding of bidirectional control of ryanodine receptor-mediated sarcoplasmic reticulum Ca2+ release and mitochondrial function, and how defects in crosstalk between these two organelles increase arrhythmic risk in cardiac disease.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, United States of America
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, United States of America
| | - Richard T Clements
- Biomedical & Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, United States of America
| | - Andriy E Belevych
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, United States of America
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, United States of America.
| |
Collapse
|
23
|
Su Y, Ahn B, Macpherson PCD, Ranjit R, Claflin DR, Van Remmen H, Brooks SV. Transgenic expression of SOD1 specifically in neurons of Sod1 deficient mice prevents defects in muscle mitochondrial function and calcium handling. Free Radic Biol Med 2021; 165:299-311. [PMID: 33561489 PMCID: PMC8026109 DOI: 10.1016/j.freeradbiomed.2021.01.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/09/2021] [Accepted: 01/25/2021] [Indexed: 01/21/2023]
Abstract
Aging is accompanied by loss of muscle mass and force, known as sarcopenia. Muscle atrophy, weakness, and neuromuscular junction (NMJ) degeneration reminiscent of normal muscle aging are observed early in adulthood for mice deficient in Cu, Zn-superoxide dismutase (SOD, Sod1-/-). Muscles of Sod1-/- mice also display impaired mitochondrial ATP production and increased mitochondrial reactive oxygen species (ROS) generation implicating oxidative stress in sarcopenia. Restoration of CuZnSOD specifically in neurons of Sod1-/- mice (SynTgSod1-/-) prevents muscle atrophy and loss of force, but whether muscle mitochondrial function is preserved is not known. To establish links among CuZnSOD expression, mitochondrial function, and sarcopenia, we examined contractile properties, mitochondrial function and ROS production, intracellular calcium transients (ICT), and NMJ morphology in lumbrical muscles of 7-9 month wild type (WT), Sod1-/-, and SynTgSod1-/- mice. Compared with WT values, mitochondrial ROS production was increased 2.9-fold under basal conditions and 2.2-fold with addition of glutamate and malate in Sod1-/- muscle fibers while oxygen consumption was not significantly altered. In addition, NADH recovery was blunted following contraction and the peak of the ICT was decreased by 25%. Mitochondrial function, ROS generation and calcium handling were restored to WT values in SynTgSod1-/- mice, despite continued lack of CuZnSOD in muscle. NMJ denervation and fragmentation were also fully rescued in SynTgSod1-/- mice suggesting that muscle mitochondrial and calcium handling defects in Sod1-/- mice are secondary to neuronal oxidative stress and its effects on the NMJ rather than the lack of muscle CuZnSOD. We conclude that intact neuronal function and innervation are key to maintaining excitation-contraction coupling and muscle mitochondrial function.
Collapse
Affiliation(s)
- Yu Su
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, PR China
| | - Bumsoo Ahn
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Peter C D Macpherson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Rojina Ranjit
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Dennis R Claflin
- Department of Surgery, Section of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Physiology, Oklahoma University Health Science Center, Oklahoma City, OK, USA; VA Medical Center, Oklahoma City, OK, USA
| | - Susan V Brooks
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
24
|
Observing and Manipulating Cell-Specific Cardiac Function with Light. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33398827 DOI: 10.1007/978-981-15-8763-4_24] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
The heart is a complex multicellular organ comprising both cardiomyocytes (CM), which make up the majority of the cardiac volume, and non-myocytes (NM), which represent the majority of cardiac cells. CM drive the pumping action of the heart, triggered via rhythmic electrical activity. NM, on the other hand, have many essential functions including generating extracellular matrix, regulating CM activity, and aiding in repair following injury. NM include neurons and interstitial, immune, and endothelial cells. Understanding the role of specific cell types and their interactions with one another may be key to developing new therapies with minimal side effects to treat cardiac disease. However, assessing cell-type-specific behavior in situ using standard techniques is challenging. Optogenetics enables population-specific observation and control, facilitating studies into the role of specific cell types and subtypes. Optogenetic models targeting the most important cardiac cell types have been generated and used to investigate non-canonical roles of those cell populations, e.g., to better understand how cardiac pacing occurs and to assess potential translational possibilities of optogenetics. So far, cardiac optogenetic studies have primarily focused on validating models and tools in the healthy heart. The field is now in a position where animal models and tools should be utilized to improve our understanding of the complex heterocellular nature of the heart, how this changes in disease, and from there to enable the development of cell-specific therapies and improved treatments.
Collapse
|
25
|
Boyman L, Greiser M, Lederer WJ. Calcium influx through the mitochondrial calcium uniporter holocomplex, MCU cx. J Mol Cell Cardiol 2021; 151:145-154. [PMID: 33147447 PMCID: PMC7880866 DOI: 10.1016/j.yjmcc.2020.10.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
Ca2+ flux into the mitochondrial matrix through the MCU holocomplex (MCUcx) has recently been measured quantitatively and with milliseconds resolution for the first time under physiological conditions in both heart and skeletal muscle. Additionally, the dynamic levels of Ca2+ in the mitochondrial matrix ([Ca2+]m) of cardiomyocytes were measured as it was controlled by the balance between influx of Ca2+ into the mitochondrial matrix through MCUcx and efflux through the mitochondrial Na+ / Ca2+ exchanger (NCLX). Under these conditions [Ca2+]m was shown to regulate ATP production by the mitochondria at only a few critical sites. Additional functions attributed to [Ca2+]m continue to be reported in the literature. Here we review the new findings attributed to MCUcx function and provide a framework for understanding and investigating mitochondrial Ca2+ influx features, many of which remain controversial. The properties and functions of the MCUcx subunits that constitute the holocomplex are challenging to tease apart. Such distinct subunits include EMRE, MCUR1, MICUx (i.e. MICU1, MICU2, MICU3), and the pore-forming subunits (MCUpore). Currently, the specific set of functions of each subunit remains non-quantitative and controversial. The more contentious issues are discussed in the context of the newly measured native MCUcx Ca2+ flux from heart and skeletal muscle. These MCUcx Ca2+ flux measurements have been shown to be a highly-regulated, tissue-specific with femto-Siemens Ca2+ conductances and with distinct extramitochondrial Ca2+ ([Ca2+]i) dependencies. These data from cardiac and skeletal muscle mitochondria have been examined quantitatively for their threshold [Ca2+]i levels and for hypothesized gatekeeping function and are discussed in the context of model cell (e.g. HeLa, MEF, HEK293, COS7 cells) measurements. Our new findings on MCUcx dependent matrix [Ca2+]m signaling provide a quantitative basis for on-going and new investigations of the roles of MCUcx in cardiac function ranging from metabolic fuel selection, capillary blood-flow control and the pathological activation of the mitochondrial permeability transition pore (mPTP). Additionally, this review presents the use of advanced new methods that can be readily adapted by any investigator to enable them to carry out quantitative Ca2+ measurements in mitochondria while controlling the inner mitochondrial membrane potential, ΔΨm.
Collapse
Affiliation(s)
- Liron Boyman
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; The Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Maura Greiser
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; The Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; The Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
26
|
Salazar-Ramírez F, Ramos-Mondragón R, García-Rivas G. Mitochondrial and Sarcoplasmic Reticulum Interconnection in Cardiac Arrhythmia. Front Cell Dev Biol 2021; 8:623381. [PMID: 33585462 PMCID: PMC7876262 DOI: 10.3389/fcell.2020.623381] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/30/2020] [Indexed: 12/31/2022] Open
Abstract
Ca2+ plays a pivotal role in mitochondrial energy production, contraction, and apoptosis. Mitochondrial Ca2+-targeted fluorescent probes have demonstrated that mitochondria Ca2+ transients are synchronized with Ca2+ fluxes occurring in the sarcoplasmic reticulum (SR). The presence of specialized proteins tethering SR to mitochondria ensures the local Ca2+ flux between these organelles. Furthermore, communication between SR and mitochondria impacts their functionality in a bidirectional manner. Mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniplex is essential for ATP production and controlled reactive oxygen species levels for proper cellular signaling. Conversely, mitochondrial ATP ensures the proper functioning of SR Ca2+-handling proteins, which ensures that mitochondria receive an adequate supply of Ca2+. Recent evidence suggests that altered SR Ca2+ proteins, such as ryanodine receptors and the sarco/endoplasmic reticulum Ca2+ ATPase pump, play an important role in maintaining proper cardiac membrane excitability, which may be initiated and potentiated when mitochondria are dysfunctional. This recognized mitochondrial role offers the opportunity to develop new therapeutic approaches aimed at preventing cardiac arrhythmias in cardiac disease.
Collapse
Affiliation(s)
- Felipe Salazar-Ramírez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Cardiovascular, Monterrey, Mexico
| | - Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Cardiovascular, Monterrey, Mexico.,TecSalud, Centro de Investigación Biomédica, Hospital Zambrano-Hellion, San Pedro Garza García, Mexico.,TecSalud, Centro de Medicina Funcional, Hospital Zambrano-Hellion, San Pedro Garza García, Mexico
| |
Collapse
|
27
|
Hamilton S, Veress R, Belevych A, Terentyev D. The role of calcium homeostasis remodeling in inherited cardiac arrhythmia syndromes. Pflugers Arch 2021; 473:377-387. [PMID: 33404893 PMCID: PMC7940310 DOI: 10.1007/s00424-020-02505-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
Sudden cardiac death due to malignant ventricular arrhythmias remains the major cause of mortality in the postindustrial world. Defective intracellular Ca2+ homeostasis has been well established as a key contributing factor to the enhanced propensity for arrhythmia in acquired cardiac disease, such as heart failure or diabetic cardiomyopathy. More recent advances provide a strong basis to the emerging view that hereditary cardiac arrhythmia syndromes are accompanied by maladaptive remodeling of Ca2+ homeostasis which substantially increases arrhythmic risk. This brief review will focus on functional changes in elements of Ca2+ handling machinery in cardiomyocytes that occur secondary to genetic mutations associated with catecholaminergic polymorphic ventricular tachycardia, and long QT syndrome.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Roland Veress
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Andriy Belevych
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
28
|
Singh S, Mabalirajan U. Mitochondrial calcium in command of juggling myriads of cellular functions. Mitochondrion 2021; 57:108-118. [PMID: 33412334 DOI: 10.1016/j.mito.2020.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 12/14/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023]
Abstract
The puzzling traits related to the evolutionary aspect of mitochondria, still positions the mitochondrion at the center of the research. The theory of endosymbiosis popularized by Lynn Margulis in 1967 gained prominence wherein the mitochondrion is believed to have emerged as a prokaryote and later integrated into the eukaryotic system. This semi-autonomous organelle has bagged two responsible but perilous cellular functions: a) energy metabolism, and b) calcium buffering, though both are interdependent. While most of the mitochondrial functions are saliently regulated by calcium ions, the calcium buffering role of mitochondria decides the cellular fate. Though calcium overload in few mitochondria makes them dysfunctional at the early stage of cellular stress, this doesn't lead to sudden cell death due to critical checkpoints like mitophagy, mitochondrial fusion, etc. Thus, mitochondrion juggles with multiple crucial cellular functions with its calcium buffering skill.
Collapse
Affiliation(s)
- Sabita Singh
- Molecular Pathobiology Of Respiratory Diseases, Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ulaganathan Mabalirajan
- Molecular Pathobiology Of Respiratory Diseases, Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
29
|
Mason FE, Pronto JRD, Alhussini K, Maack C, Voigt N. Cellular and mitochondrial mechanisms of atrial fibrillation. Basic Res Cardiol 2020; 115:72. [PMID: 33258071 PMCID: PMC7704501 DOI: 10.1007/s00395-020-00827-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/26/2020] [Indexed: 11/06/2022]
Abstract
The molecular mechanisms underlying atrial fibrillation (AF), the most common form of arrhythmia, are poorly understood and therefore target-specific treatment options remain an unmet clinical need. Excitation–contraction coupling in cardiac myocytes requires high amounts of adenosine triphosphate (ATP), which is replenished by oxidative phosphorylation in mitochondria. Calcium (Ca2+) is a key regulator of mitochondrial function by stimulating the Krebs cycle, which produces nicotinamide adenine dinucleotide for ATP production at the electron transport chain and nicotinamide adenine dinucleotide phosphate for the elimination of reactive oxygen species (ROS). While it is now well established that mitochondrial dysfunction plays an important role in the pathophysiology of heart failure, this has been less investigated in atrial myocytes in AF. Considering the high prevalence of AF, investigating the role of mitochondria in this disease may guide the path towards new therapeutic targets. In this review, we discuss the importance of mitochondrial Ca2+ handling in regulating ATP production and mitochondrial ROS emission and how alterations, particularly in these aspects of mitochondrial activity, may play a role in AF. In addition to describing research advances, we highlight areas in which further studies are required to elucidate the role of mitochondria in AF.
Collapse
Affiliation(s)
- Fleur E Mason
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Khaled Alhussini
- Department of Thoracic and Cardiovascular Surgery, University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center Würzburg, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany. .,Department of Internal Medicine I, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany. .,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
30
|
Gao P, Yan Z, Zhu Z. Mitochondria-Associated Endoplasmic Reticulum Membranes in Cardiovascular Diseases. Front Cell Dev Biol 2020; 8:604240. [PMID: 33240899 PMCID: PMC7680862 DOI: 10.3389/fcell.2020.604240] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/21/2020] [Indexed: 12/20/2022] Open
Abstract
The endoplasmic reticulum (ER) and mitochondria are physically connected to form dedicated structural domains known as mitochondria-associated ER membranes (MAMs), which participate in fundamental biological processes, including lipid and calcium (Ca2+) homeostasis, mitochondrial dynamics and other related cellular behaviors such as autophagy, ER stress, inflammation and apoptosis. Many studies have proved the importance of MAMs in maintaining the normal function of both organelles, and the abnormal amount, structure or function of MAMs is related to the occurrence of cardiovascular diseases. Here, we review the knowledge regarding the components of MAMs according to their different functions and the specific roles of MAMs in cardiovascular physiology and pathophysiology, focusing on some highly prevalent cardiovascular diseases, including ischemia-reperfusion, diabetic cardiomyopathy, heart failure, pulmonary arterial hypertension and systemic vascular diseases. Finally, we summarize the possible mechanisms of MAM in cardiovascular diseases and put forward some obstacles in the understanding of MAM function we may encounter.
Collapse
Affiliation(s)
- Peng Gao
- Department of Hypertension and Endocrinology, Chongqing Institute of Hypertension, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology, Chongqing Institute of Hypertension, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Chongqing Institute of Hypertension, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
31
|
Oropeza-Almazán Y, Blatter LA. Mitochondrial calcium uniporter complex activation protects against calcium alternans in atrial myocytes. Am J Physiol Heart Circ Physiol 2020; 319:H873-H881. [PMID: 32857593 DOI: 10.1152/ajpheart.00375.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cardiac alternans, defined as beat-to-beat alternations in action potential duration, cytosolic Ca transient (CaT) amplitude, and cardiac contraction is associated with atrial fibrillation (AF) and sudden cardiac death. At the cellular level, cardiac alternans is linked to abnormal intracellular calcium handling during excitation-contraction coupling. We investigated how pharmacological activation or inhibition of cytosolic Ca sequestration via mitochondrial Ca uptake and mitochondrial Ca retention affects the occurrence of pacing-induced CaT alternans in isolated rabbit atrial myocytes. Cytosolic CaTs were recorded using Fluo-4 fluorescence microscopy. Alternans was quantified as the alternans ratio (AR = 1 - CaTsmall/CaTlarge, where CaTsmall and CaTlarge are the amplitudes of the small and large CaTs of a pair of alternating CaTs). Inhibition of mitochondrial Ca sequestration via mitochondrial Ca uniporter complex (MCUC) with Ru360 enhanced the severity of CaT alternans (AR increase) and lowered the pacing frequency threshold for alternans. In contrast, stimulation of MCUC mediated mitochondrial Ca uptake with spermine-rescued alternans (AR decrease) and increased the alternans pacing threshold. Direct measurement of mitochondrial [Ca] in membrane permeabilized myocytes with Fluo-4 loaded mitochondria revealed that spermine enhanced and accelerated mitochondrial Ca uptake. Stimulation of mitochondrial Ca retention by preventing mitochondrial Ca efflux through the mitochondrial permeability transition pore with cyclosporin A also protected from alternans and increased the alternans pacing threshold. Pharmacological manipulation of MCUC activity did not affect sarcoplasmic reticulum Ca load. Our results suggest that activation of Ca sequestration by mitochondria protects from CaT alternans and could be a potential therapeutic target for cardiac alternans and AF prevention.NEW & NOTEWORTHY This study provides conclusive evidence that mitochondrial Ca uptake and retention protects from Ca alternans, whereas uptake inhibition enhances Ca alternans. The data suggest pharmacological mitochondrial Ca cycling modulation as a potential therapeutic strategy for alternans-related cardiac arrhythmia prevention.
Collapse
Affiliation(s)
| | - Lothar A Blatter
- Department of Physiology and Biophysics, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
32
|
Gordan R, Fefelova N, Gwathmey JK, Xie LH. Iron Overload, Oxidative Stress and Calcium Mishandling in Cardiomyocytes: Role of the Mitochondrial Permeability Transition Pore. Antioxidants (Basel) 2020; 9:E758. [PMID: 32824344 PMCID: PMC7465659 DOI: 10.3390/antiox9080758] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
Iron (Fe) plays an essential role in many physiological processes. Hereditary hemochromatosis or frequent blood transfusions often cause iron overload (IO), which can lead to cardiomyopathy and arrhythmias; however, the underlying mechanism is not well defined. In the present study, we assess the hypothesis that IO promotes arrhythmias via reactive oxygen species (ROS) production, mitochondrial membrane potential (∆Ψm) depolarization, and disruption of cytosolic Ca dynamics. In ventricular myocytes isolated from wild type (WT) mice, both cytosolic and mitochondrial Fe levels were elevated following perfusion with the Fe3+/8-hydroxyquinoline (8-HQ) complex. IO promoted mitochondrial superoxide generation (measured using MitoSOX Red) and induced the depolarization of the ΔΨm (measured using tetramethylrhodamine methyl ester, TMRM) in a dose-dependent manner. IO significantly increased the rate of Ca wave (CaW) formation measured in isolated ventricular myocytes using Fluo-4. Furthermore, in ex-vivo Langendorff-perfused hearts, IO increased arrhythmia scores as evaluated by ECG recordings under programmed S1-S2 stimulation protocols. We also carried out similar experiments in cyclophilin D knockout (CypD KO) mice in which the mitochondrial permeability transition pore (mPTP) opening is impaired. While comparable cytosolic and mitochondrial Fe load, mitochondrial ROS production, and depolarization of the ∆Ψm were observed in ventricular myocytes isolated from both WT and CypD KO mice, the rate of CaW formation in isolated cells and the arrhythmia scores in ex-vivo hearts were significantly lower in CypD KO mice compared to those observed in WT mice under conditions of IO. The mPTP inhibitor cyclosporine A (CsA, 1 µM) also exhibited a protective effect. In conclusion, our results suggest that IO induces mitochondrial ROS generation and ∆Ψm depolarization, thus opening the mPTP, thereby promoting CaWs and cardiac arrhythmias. Conversely, the inhibition of mPTP ameliorates the proarrhythmic effects of IO.
Collapse
Affiliation(s)
| | | | | | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (R.G.); (N.F.); (J.K.G.)
| |
Collapse
|
33
|
Werley CA, Boccardo S, Rigamonti A, Hansson EM, Cohen AE. Multiplexed Optical Sensors in Arrayed Islands of Cells for multimodal recordings of cellular physiology. Nat Commun 2020; 11:3881. [PMID: 32753572 PMCID: PMC7403318 DOI: 10.1038/s41467-020-17607-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 07/07/2020] [Indexed: 11/29/2022] Open
Abstract
Cells typically respond to chemical or physical perturbations via complex signaling cascades which can simultaneously affect multiple physiological parameters, such as membrane voltage, calcium, pH, and redox potential. Protein-based fluorescent sensors can report many of these parameters, but spectral overlap prevents more than ~4 modalities from being recorded in parallel. Here we introduce the technique, MOSAIC, Multiplexed Optical Sensors in Arrayed Islands of Cells, where patterning of fluorescent sensor-encoding lentiviral vectors with a microarray printer enables parallel recording of multiple modalities. We demonstrate simultaneous recordings from 20 sensors in parallel in human embryonic kidney (HEK293) cells and in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), and we describe responses to metabolic and pharmacological perturbations. Together, these results show that MOSAIC can provide rich multi-modal data on complex physiological responses in multiple cell types.
Collapse
Affiliation(s)
- Christopher A Werley
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
- Q-State Biosciences, Cambridge, MA, 02139, USA
| | - Stefano Boccardo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
- Nobel Biocare AG, Kloten, Switzerland
| | - Alessandra Rigamonti
- Integrated Cardio Metabolic Centre, Department of Medicine Huddinge, Karolinska Institute, Huddinge, Sweden
| | - Emil M Hansson
- Integrated Cardio Metabolic Centre, Department of Medicine Huddinge, Karolinska Institute, Huddinge, Sweden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
34
|
Rossini M, Filadi R. Sarcoplasmic Reticulum-Mitochondria Kissing in Cardiomyocytes: Ca 2+, ATP, and Undisclosed Secrets. Front Cell Dev Biol 2020; 8:532. [PMID: 32671075 PMCID: PMC7332691 DOI: 10.3389/fcell.2020.00532] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
In cardiomyocytes, to carry out cell contraction, the distribution, morphology, and dynamic interaction of different cellular organelles are tightly regulated. For instance, the repetitive close apposition between junctional sarcoplasmic reticulum (jSR) and specialized sarcolemma invaginations, called transverse-tubules (TTs), is essential for an efficient excitation-contraction coupling (ECC). Upon an action potential, Ca2+ microdomains, generated in synchrony at the interface between TTs and jSR, underlie the prompt increase in cytosolic Ca2+ concentration, ultimately responsible for cell contraction during systole. This process requires a considerable amount of energy and the active participation of mitochondria, which encompass ∼30% of the cell volume and represent the major source of ATP in the heart. Importantly, in adult cardiomyocytes, mitochondria are distributed in a highly orderly fashion and strategically juxtaposed with SR. By taking advantage of the vicinity to Ca2+ releasing sites, they take up Ca2+ and modulate ATP synthesis according to the specific cardiac workload. Interestingly, with respect to SR, a biased, polarized positioning of mitochondrial Ca2+ uptake/efflux machineries has been reported, hinting the importance of a strictly regulated mitochondrial Ca2+ handling for heart activity. This notion, however, has been questioned by the observation that, in some mouse models, the deficiency of specific molecules, modulating mitochondrial Ca2+ dynamics, triggers non-obvious cardiac phenotypes. This review will briefly summarize the physiological significance of SR-mitochondria apposition in cardiomyocytes, as well as the pathological consequences of an altered organelle communication, focusing on Ca2+ signaling. We will discuss ongoing debates and propose future research directions.
Collapse
Affiliation(s)
- Michela Rossini
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Neuroscience Institute - Italian National Research Council (CNR), Padua, Italy
| |
Collapse
|
35
|
Kirschner Peretz N, Segal S, Yaniv Y. May the Force Not Be With You During Culture: Eliminating Mechano-Associated Feedback During Culture Preserves Cultured Atrial and Pacemaker Cell Functions. Front Physiol 2020; 11:163. [PMID: 32265724 PMCID: PMC7100534 DOI: 10.3389/fphys.2020.00163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/12/2020] [Indexed: 01/24/2023] Open
Abstract
Cultured cardiomyocytes have been shown to possess significant potential as a model for characterization of mechano-Ca2+, mechano-electric, and mechano-metabolic feedbacks in the heart. However, the majority of cultured cardiomyocytes exhibit impaired electrical, mechanical, biochemical, and metabolic functions. More specifically, the cells do not beat spontaneously (pacemaker cells) or beat at a rate far lower than their physiological counterparts and self-oscillate (atrial and ventricular cells) in culture. Thus, efforts are being invested in ensuring that cultured cardiomyocytes maintain the shape and function of freshly isolated cells. Elimination of contraction during culture has been shown to preserve the mechano-Ca2+, mechano-electric, and mechano-metabolic feedback loops of cultured cells. This review focuses on pacemaker cells, which reside in the sinoatrial node (SAN) and generate regular heartbeat through the initiation of the heart’s electrical, metabolic, and biochemical activities. In parallel, it places emphasis on atrial cells, which are responsible for bridging the electrical conductance from the SAN to the ventricle. The review provides a summary of the main mechanisms responsible for mechano-electrical, Ca2+, and metabolic feedback in pacemaker and atrial cells and of culture methods existing for both cell types. The work concludes with an explanation of how the elimination of mechano-electrical, mechano-Ca2+, and mechano-metabolic feedbacks during culture results in sustained cultured cell function.
Collapse
Affiliation(s)
- Noa Kirschner Peretz
- Biomedical Engineering Faculty, Technion Israel Institute of Technology, Haifa, Israel
| | - Sofia Segal
- Biomedical Engineering Faculty, Technion Israel Institute of Technology, Haifa, Israel
| | - Yael Yaniv
- Biomedical Engineering Faculty, Technion Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
36
|
Gilbert G, Demydenko K, Dries E, Puertas RD, Jin X, Sipido K, Roderick HL. Calcium Signaling in Cardiomyocyte Function. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035428. [PMID: 31308143 DOI: 10.1101/cshperspect.a035428] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Rhythmic increases in intracellular Ca2+ concentration underlie the contractile function of the heart. These heart muscle-wide changes in intracellular Ca2+ are induced and coordinated by electrical depolarization of the cardiomyocyte sarcolemma by the action potential. Originating at the sinoatrial node, conduction of this electrical signal throughout the heart ensures synchronization of individual myocytes into an effective cardiac pump. Ca2+ signaling pathways also regulate gene expression and cardiomyocyte growth during development and in pathology. These fundamental roles of Ca2+ in the heart are illustrated by the prevalence of altered Ca2+ homeostasis in cardiovascular diseases. Indeed, heart failure (an inability of the heart to support hemodynamic needs), rhythmic disturbances, and inappropriate cardiac growth all share an involvement of altered Ca2+ handling. The prevalence of these pathologies, contributing to a third of all deaths in the developed world as well as to substantial morbidity makes understanding the mechanisms of Ca2+ handling and dysregulation in cardiomyocytes of great importance.
Collapse
Affiliation(s)
- Guillaume Gilbert
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Kateryna Demydenko
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Eef Dries
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Rosa Doñate Puertas
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Xin Jin
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - Karin Sipido
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, BE3000 Leuven, Belgium
| |
Collapse
|
37
|
Miranda‐Silva D, Wüst RCI, Conceição G, Gonçalves‐Rodrigues P, Gonçalves N, Gonçalves A, Kuster DWD, Leite‐Moreira AF, Velden J, Sousa Beleza JM, Magalhães J, Stienen GJM, Falcão‐Pires I. Disturbed cardiac mitochondrial and cytosolic calcium handling in a metabolic risk-related rat model of heart failure with preserved ejection fraction. Acta Physiol (Oxf) 2020; 228:e13378. [PMID: 31520455 PMCID: PMC7064935 DOI: 10.1111/apha.13378] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022]
Abstract
AIM Calcium ions play a pivotal role in matching energy supply and demand in cardiac muscle. Mitochondrial calcium concentration is lower in animal models of heart failure with reduced ejection fraction (HFrEF), but limited information is available about mitochondrial calcium handling in heart failure with preserved ejection fraction (HFpEF). METHODS We assessed mitochondrial Ca2+ handling in intact cardiomyocytes from Zucker/fatty Spontaneously hypertensive F1 hybrid (ZSF1)-lean (control) and ZSF1-obese rats, a metabolic risk-related model of HFpEF. A mitochondrially targeted Ca2+ indicator (MitoCam) was expressed in cultured adult rat cardiomyocytes. Cytosolic and mitochondrial Ca2+ transients were measured at different stimulation frequencies. Mitochondrial respiration and swelling, and expression of key proteins were determined ex vivo. RESULTS At rest, mitochondrial Ca2+ concentration in ZSF1-obese was larger than in ZSF1-lean. The diastolic and systolic mitochondrial Ca2+ concentrations increased with stimulation frequency, but the steady-state levels were larger in ZSF1-obese. The half-widths of the contractile responses, the resting cytosolic Ca2+ concentration and the decay half-times of the cytosolic Ca2+ transients were higher in ZSF1-obese, likely because of a lower SERCA2a/phospholamban ratio. Mitochondrial respiration was lower, particularly with nicotinamide adenine dinucleotide (NADH) (complex I) substrates, and mitochondrial swelling was larger in ZSF1-obese. CONCLUSION The free mitochondrial calcium concentration is higher in HFpEF owing to alterations in mitochondrial and cytosolic Ca2+ handling. This coupling between cytosolic and mitochondrial Ca2+ levels may compensate for myocardial ATP supply in vivo under conditions of mild mitochondrial dysfunction. However, if mitochondrial Ca2+ concentration is sustainedly increased, it might trigger mitochondrial permeability transition pore opening.
Collapse
Affiliation(s)
- Daniela Miranda‐Silva
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Rob C. I. Wüst
- Department of Physiology Amsterdam UMC VUmc Amsterdam Cardiovascular Sciences Amsterdam the Netherlands
- Department of Human Movement Sciences Laboratory for Myology Faculty of Behavioural and Movement Sciences Amsterdam Movement Sciences Vrije Universiteit Amsterdam Amsterdam the Netherlands
| | - Glória Conceição
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Patrícia Gonçalves‐Rodrigues
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Nádia Gonçalves
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Alexandre Gonçalves
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Diederik W. D. Kuster
- Department of Physiology Amsterdam UMC VUmc Amsterdam Cardiovascular Sciences Amsterdam the Netherlands
| | - Adelino F. Leite‐Moreira
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| | - Jolanda Velden
- Department of Physiology Amsterdam UMC VUmc Amsterdam Cardiovascular Sciences Amsterdam the Netherlands
- Netherlands Heart Institute Utrecht the Netherlands
| | - Jorge M. Sousa Beleza
- LaMetEx—Laboratory of Metabolism and Exercise Faculty of Sport Cardiovascular Research Center - UniC, University of Porto Porto Portugal
| | - José Magalhães
- LaMetEx—Laboratory of Metabolism and Exercise Faculty of Sport Cardiovascular Research Center - UniC, University of Porto Porto Portugal
| | - Ger J. M. Stienen
- Department of Physiology Amsterdam UMC VUmc Amsterdam Cardiovascular Sciences Amsterdam the Netherlands
| | - Inês Falcão‐Pires
- Department of Surgery and Physiology Cardiovascular R & D center Faculty of Medicine of the University of Porto Porto Portugal
| |
Collapse
|
38
|
Lambert JP, Murray EK, Elrod JW. MCUB and mitochondrial calcium uptake - modeling, function, and therapeutic potential. Expert Opin Ther Targets 2020; 24:163-169. [PMID: 32093523 PMCID: PMC7078044 DOI: 10.1080/14728222.2020.1732926] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/18/2020] [Indexed: 01/06/2023]
Affiliation(s)
| | - Emma K Murray
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - John W Elrod
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
39
|
Eisner DA, Caldwell JL, Trafford AW, Hutchings DC. The Control of Diastolic Calcium in the Heart: Basic Mechanisms and Functional Implications. Circ Res 2020; 126:395-412. [PMID: 31999537 PMCID: PMC7004450 DOI: 10.1161/circresaha.119.315891] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Normal cardiac function requires that intracellular Ca2+ concentration be reduced to low levels in diastole so that the ventricle can relax and refill with blood. Heart failure is often associated with impaired cardiac relaxation. Little, however, is known about how diastolic intracellular Ca2+ concentration is regulated. This article first discusses the reasons for this ignorance before reviewing the basic mechanisms that control diastolic intracellular Ca2+ concentration. It then considers how the control of systolic and diastolic intracellular Ca2+ concentration is intimately connected. Finally, it discusses the changes that occur in heart failure and how these may result in heart failure with preserved versus reduced ejection fraction.
Collapse
Affiliation(s)
- David A Eisner
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Jessica L Caldwell
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Andrew W Trafford
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - David C Hutchings
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| |
Collapse
|
40
|
Gökerküçük EB, Tramier M, Bertolin G. Imaging Mitochondrial Functions: from Fluorescent Dyes to Genetically-Encoded Sensors. Genes (Basel) 2020; 11:E125. [PMID: 31979408 PMCID: PMC7073610 DOI: 10.3390/genes11020125] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondria are multifunctional organelles that are crucial to cell homeostasis. They constitute the major site of energy production for the cell, they are key players in signalling pathways using secondary messengers such as calcium, and they are involved in cell death and redox balance paradigms. Mitochondria quickly adapt their dynamics and biogenesis rates to meet the varying energy demands of the cells, both in normal and in pathological conditions. Therefore, understanding simultaneous changes in mitochondrial functions is crucial in developing mitochondria-based therapy options for complex pathological conditions such as cancer, neurological disorders, and metabolic syndromes. To this end, fluorescence microscopy coupled to live imaging represents a promising strategy to track these changes in real time. In this review, we will first describe the commonly available tools to follow three key mitochondrial functions using fluorescence microscopy: Calcium signalling, mitochondrial dynamics, and mitophagy. Then, we will focus on how the development of genetically-encoded fluorescent sensors became a milestone for the understanding of these mitochondrial functions. In particular, we will show how these tools allowed researchers to address several biochemical activities in living cells, and with high spatiotemporal resolution. With the ultimate goal of tracking multiple mitochondrial functions simultaneously, we will conclude by presenting future perspectives for the development of novel genetically-encoded fluorescent biosensors.
Collapse
Affiliation(s)
| | | | - Giulia Bertolin
- Univ Rennes, CNRS, IGDR [Institut de génétique et développement de Rennes] UMR 6290, F-35000 Rennes, France
| |
Collapse
|
41
|
Arrieta A, Blackwood EA, Stauffer WT, Glembotski CC. Integrating ER and Mitochondrial Proteostasis in the Healthy and Diseased Heart. Front Cardiovasc Med 2020; 6:193. [PMID: 32010709 PMCID: PMC6974444 DOI: 10.3389/fcvm.2019.00193] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
The integrity of the proteome in cardiac myocytes is critical for robust heart function. Proteome integrity in all cells is managed by protein homeostasis or proteostasis, which encompasses processes that maintain the balance of protein synthesis, folding, and degradation in ways that allow cells to adapt to conditions that present a potential challenge to viability (1). While there are processes in various cellular locations in cardiac myocytes that contribute to proteostasis, those in the cytosol, mitochondria and endoplasmic reticulum (ER) have dominant roles in maintaining cardiac contractile function. Cytosolic proteostasis has been reviewed elsewhere (2, 3); accordingly, this review focuses on proteostasis in the ER and mitochondria, and how they might influence each other and, thus, impact heart function in the settings of cardiac physiology and disease.
Collapse
Affiliation(s)
- Adrian Arrieta
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, CA, United States
| | - Erik A Blackwood
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, CA, United States
| | - Winston T Stauffer
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, CA, United States
| | - Christopher C Glembotski
- Department of Biology, San Diego State University Heart Institute, San Diego State University, San Diego, CA, United States
| |
Collapse
|
42
|
Song Z, Xie LH, Weiss JN, Qu Z. A Spatiotemporal Ventricular Myocyte Model Incorporating Mitochondrial Calcium Cycling. Biophys J 2019; 117:2349-2360. [PMID: 31623883 PMCID: PMC6990377 DOI: 10.1016/j.bpj.2019.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/19/2019] [Accepted: 09/09/2019] [Indexed: 12/27/2022] Open
Abstract
Intracellular calcium (Ca2+) cycling dynamics in cardiac myocytes are spatiotemporally generated by stochastic events arising from a spatially distributed network of coupled Ca2+ release units that interact with an intertwined mitochondrial network. In this study, we developed a spatiotemporal ventricular myocyte model that integrates mitochondria-related Ca2+ cycling components into our previously developed ventricular myocyte model consisting of a three-dimensional Ca2+ release unit network. Mathematical formulations of mitochondrial membrane potential, mitochondrial Ca2+ cycling, mitochondrial permeability transition pore stochastic opening and closing, intracellular reactive oxygen species signaling, and oxidized Ca2+/calmodulin-dependent protein kinase II signaling were incorporated into the model. We then used the model to simulate the effects of mitochondrial depolarization on mitochondrial Ca2+ cycling, Ca2+ spark frequency, and Ca2+ amplitude, which agree well with experimental data. We also simulated the effects of the strength of mitochondrial Ca2+ uniporters and their spatial localization on intracellular Ca2+ cycling properties, which substantially affected diastolic and systolic Ca2+ levels in the mitochondria but exhibited only a small effect on sarcoplasmic reticulum and cytosolic Ca2+ levels under normal conditions. We show that mitochondrial depolarization can cause Ca2+ waves and Ca2+ alternans, which agrees with previous experimental observations. We propose that this new, to our knowledge, spatiotemporal ventricular myocyte model, incorporating properties of mitochondrial Ca2+ cycling and reactive-oxygen-species-dependent signaling, will be useful for investigating the effects of mitochondria on intracellular Ca2+ cycling and action potential dynamics in ventricular myocytes.
Collapse
Affiliation(s)
- Zhen Song
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - James N Weiss
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Zhilin Qu
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Department of Biomathematics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
43
|
Tarasova NV, Vishnyakova PA, Logashina YA, Elchaninov AV. Mitochondrial Calcium Uniporter Structure and Function in Different Types of Muscle Tissues in Health and Disease. Int J Mol Sci 2019; 20:ijms20194823. [PMID: 31569359 PMCID: PMC6801532 DOI: 10.3390/ijms20194823] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/12/2019] [Accepted: 09/26/2019] [Indexed: 02/07/2023] Open
Abstract
Calcium ions (Ca2+) influx to mitochondrial matrix is crucial for the life of a cell. Mitochondrial calcium uniporter (mtCU) is a protein complex which consists of the pore-forming subunit (MCU) and several regulatory subunits. MtCU is the main contributor to inward Ca2+ currents through the inner mitochondrial membrane. Extensive investigations of mtCU involvement into normal and pathological molecular pathways started from the moment of discovery of its molecular components. A crucial role of mtCU in the control of these pathways is now recognized in both health and disease. In particular, impairments of mtCU function have been demonstrated for cardiovascular and skeletal muscle-associated pathologies. This review summarizes the current state of knowledge on mtCU structure, regulation, and function in different types of muscle tissues in health and disease.
Collapse
Affiliation(s)
- Nadezhda V Tarasova
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Trubetskaya str. 8, bld. 2, Moscow 119991, Russia.
| | - Polina A Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, Moscow 117997, Russia.
| | - Yulia A Logashina
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Trubetskaya str. 8, bld. 2, Moscow 119991, Russia.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Street 16/10, Moscow 117997, Russia.
| | - Andrey V Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, Moscow 117997, Russia.
- Scientific Research Institute of Human Morphology, 3 Tsurupa Street, Moscow 117418, Russia.
- Peoples' Friendship University of Russia, 6 Miklukho-Maklaya Street, Moscow 117198, Russia.
| |
Collapse
|
44
|
Abstract
In heart failure, alterations of Na+ and Ca2+ handling, energetic deficit, and oxidative stress in cardiac myocytes are important pathophysiological hallmarks. Mitochondria are central to these processes because they are the main source for ATP, but also reactive oxygen species (ROS), and their function is critically controlled by Ca2+ During physiological variations of workload, mitochondrial Ca2+ uptake is required to match energy supply to demand but also to keep the antioxidative capacity in a reduced state to prevent excessive emission of ROS. Mitochondria take up Ca2+ via the mitochondrial Ca2+ uniporter, which exists in a multiprotein complex whose molecular components were identified only recently. In heart failure, deterioration of cytosolic Ca2+ and Na+ handling hampers mitochondrial Ca2+ uptake and the ensuing Krebs cycle-induced regeneration of the reduced forms of NADH (nicotinamide adenine dinucleotide) and NADPH (nicotinamide adenine dinucleotide phosphate), giving rise to energetic deficit and oxidative stress. ROS emission from mitochondria can trigger further ROS release from neighboring mitochondria termed ROS-induced ROS release, and cross talk between different ROS sources provides a spatially confined cellular network of redox signaling. Although low levels of ROS may serve physiological roles, higher levels interfere with excitation-contraction coupling, induce maladaptive cardiac remodeling through redox-sensitive kinases, and cell death through mitochondrial permeability transition. Targeting the dysregulated interplay between excitation-contraction coupling and mitochondrial energetics may ameliorate the progression of heart failure.
Collapse
Affiliation(s)
- Edoardo Bertero
- From the Comprehensive Heart Failure Center, University Clinic Würzburg, Germany
| | - Christoph Maack
- From the Comprehensive Heart Failure Center, University Clinic Würzburg, Germany.
| |
Collapse
|
45
|
Lu X, Thai PN, Lu S, Pu J, Bers DM. Intrafibrillar and perinuclear mitochondrial heterogeneity in adult cardiac myocytes. J Mol Cell Cardiol 2019; 136:72-84. [PMID: 31491377 DOI: 10.1016/j.yjmcc.2019.08.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 08/12/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022]
Abstract
Mitochondria are involved in multiple cellular functions, in addition to their core role in energy metabolism. Mitochondria localized in different cellular locations may have different morphology, Ca2+ handling and biochemical properties and may interact differently with other intracellular structures, causing functional specificity. However, most prior studies have utilized isolated mitochondria, removed from their intracellular environment. Mitochondria in cardiac ventricular myocytes are highly organized, with a majority squeezed between the myofilaments in longitudinal chains (intrafibrillar mitochondria, IFM). There is another population of perinuclear mitochondria (PNM) around and between the two nuclei typical in myocytes. Here, we take advantage of live myocyte imaging to test for quantitative morphological and functional differences between IFM and PNM with respect to calcium fluxes, membrane potential, sensitivity to oxidative stress, shape and dynamics. Our findings show higher mitochondrial Ca2+ uptake and oxidative stress sensitivity for IFM vs. PNM, which may relate to higher local energy demand supporting the contractile machinery. In contrast to IFM which are remarkably static, PNM are relatively mobile, appear to participate readily in fission/fusion dynamics and appear to play a central role in mitochondrial genesis and turnover. We conclude that while IFM may be physiologically tuned to support local myofilament energy demands, PNM may be more critical in mitochondrial turnover and regulation of nuclear function and import/export. Thus, important functional differences are present in intrafibrillar vs. perinuclear mitochondrial subpopulations.
Collapse
Affiliation(s)
- Xiyuan Lu
- Division of Cardiology, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital School of Medicine, Shanghai Cancer Institute, Jiaotong University, Shanghai, China; Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | - Phung N Thai
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Shan Lu
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Jun Pu
- Division of Cardiology, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital School of Medicine, Shanghai Cancer Institute, Jiaotong University, Shanghai, China
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| |
Collapse
|
46
|
Fernandez-Sanz C, De la Fuente S, Sheu SS. Mitochondrial Ca 2+ concentrations in live cells: quantification methods and discrepancies. FEBS Lett 2019; 593:1528-1541. [PMID: 31058316 DOI: 10.1002/1873-3468.13427] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 12/11/2022]
Abstract
Intracellular Ca2+ signaling controls numerous cellular functions. Mitochondria respond to cytosolic Ca2+ changes by adapting mitochondrial functions and, in some cell types, shaping the spatiotemporal properties of the cytosolic Ca2+ signal. Numerous methods have been developed to specifically and quantitatively measure the mitochondrial-free Ca2+ concentrations ([Ca2+ ]m ), but there are still significant discrepancies in the calculated absolute values of [Ca2+ ]m in stimulated live cells. These discrepancies may be due to the distinct properties of the methods used to measure [Ca2+ ]m , the calcium-free/bound ratio, and the cell-type and stimulus-dependent Ca2+ dynamics. Critical processes happening in the mitochondria, such as ATP generation, ROS homeostasis, and mitochondrial permeability transition opening, depend directly on the [Ca2+ ]m values. Thus, precise determination of absolute [Ca2+ ]m values is imperative for understanding Ca2+ signaling. This review summarizes the reported calibrated [Ca2+ ]m values in many cell types and discusses the discrepancies among these values. Areas for future research are also proposed.
Collapse
Affiliation(s)
- Celia Fernandez-Sanz
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sergio De la Fuente
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
47
|
Hamilton S, Terentyev D. Altered Intracellular Calcium Homeostasis and Arrhythmogenesis in the Aged Heart. Int J Mol Sci 2019; 20:ijms20102386. [PMID: 31091723 PMCID: PMC6566636 DOI: 10.3390/ijms20102386] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/29/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022] Open
Abstract
Aging of the heart is associated with a blunted response to sympathetic stimulation, reduced contractility, and increased propensity for arrhythmias, with the risk of sudden cardiac death significantly increased in the elderly population. The altered cardiac structural and functional phenotype, as well as age-associated prevalent comorbidities including hypertension and atherosclerosis, predispose the heart to atrial fibrillation, heart failure, and ventricular tachyarrhythmias. At the cellular level, perturbations in mitochondrial function, excitation-contraction coupling, and calcium homeostasis contribute to this electrical and contractile dysfunction. Major determinants of cardiac contractility are the intracellular release of Ca2+ from the sarcoplasmic reticulum by the ryanodine receptors (RyR2), and the following sequestration of Ca2+ by the sarco/endoplasmic Ca2+-ATPase (SERCa2a). Activity of RyR2 and SERCa2a in myocytes is not only dependent on expression levels and interacting accessory proteins, but on fine-tuned regulation via post-translational modifications. In this paper, we review how aberrant changes in intracellular Ca2+ cycling via these proteins contributes to arrhythmogenesis in the aged heart.
Collapse
Affiliation(s)
- Shanna Hamilton
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Dmitry Terentyev
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
48
|
Cao JL, Adaniya SM, Cypress MW, Suzuki Y, Kusakari Y, Jhun BS, O-Uchi J. Role of mitochondrial Ca 2+ homeostasis in cardiac muscles. Arch Biochem Biophys 2019; 663:276-287. [PMID: 30684463 PMCID: PMC6469710 DOI: 10.1016/j.abb.2019.01.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/10/2019] [Accepted: 01/22/2019] [Indexed: 12/22/2022]
Abstract
Recent discoveries of the molecular identity of mitochondrial Ca2+ influx/efflux mechanisms have placed mitochondrial Ca2+ transport at center stage in views of cellular regulation in various cell-types/tissues. Indeed, mitochondria in cardiac muscles also possess the molecular components for efficient uptake and extraction of Ca2+. Over the last several years, multiple groups have taken advantage of newly available molecular information about these proteins and applied genetic tools to delineate the precise mechanisms for mitochondrial Ca2+ handling in cardiomyocytes and its contribution to excitation-contraction/metabolism coupling in the heart. Though mitochondrial Ca2+ has been proposed as one of the most crucial secondary messengers in controlling a cardiomyocyte's life and death, the detailed mechanisms of how mitochondrial Ca2+ regulates physiological mitochondrial and cellular functions in cardiac muscles, and how disorders of this mechanism lead to cardiac diseases remain unclear. In this review, we summarize the current controversies and discrepancies regarding cardiac mitochondrial Ca2+ signaling that remain in the field to provide a platform for future discussions and experiments to help close this gap.
Collapse
Affiliation(s)
- Jessica L Cao
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, USA; Department of Medicine, Division of Cardiology, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Stephanie M Adaniya
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, USA; Department of Medicine, Division of Cardiology, The Warren Alpert Medical School of Brown University, Providence, RI, USA; Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Michael W Cypress
- Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Yuta Suzuki
- Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Yoichiro Kusakari
- Department of Cell Physiology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Bong Sook Jhun
- Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Jin O-Uchi
- Lillehei Heart Institute, Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
49
|
Vicario M, Calì T. Measuring Ca 2+ Levels in Subcellular Compartments with Genetically Encoded GFP-Based Indicators. Methods Mol Biol 2019; 1925:31-42. [PMID: 30674014 DOI: 10.1007/978-1-4939-9018-4_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Ca2+ homeostasis is crucial for the entire life of eukaryotic cells from the beginning to the end. Mishandling in Ca2+ homeostasis is indeed linked with a large number of pathological conditions. Thus, the possibility to specifically monitor cellular calcium fluxes in different subcellular compartments represents a key tool to deeply understand the mechanisms involved in cellular dysfunctions. To cope with this need, several Ca2+ indicators have been developed allowing to accurately measure both basal Ca2+ concentration and agonist-induced Ca2+ signals in a wide spectrum of organelles. Among these, the genetically encoded GFP-based indicators are routinely used to measure Ca2+ transients thanks to their ability to change their spectral properties in response to Ca2+ binding. In this chapter, we will describe a protocol that utilizes the GCaMP6f probe targeted to mitochondria (4mtGCaMP) to measure mitochondrial calcium levels in resting conditions in HeLa cells. This method allows to easily and quickly register alterations of mitochondrial Ca2+ homeostasis in different cell populations and experimental settings, representing a precious tool to unravel the pathological pathways leading to pathogenic conditions.
Collapse
Affiliation(s)
- Mattia Vicario
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| |
Collapse
|
50
|
Hamilton S, Terentyeva R, Kim TY, Bronk P, Clements RT, O-Uchi J, Csordás G, Choi BR, Terentyev D. Pharmacological Modulation of Mitochondrial Ca 2+ Content Regulates Sarcoplasmic Reticulum Ca 2+ Release via Oxidation of the Ryanodine Receptor by Mitochondria-Derived Reactive Oxygen Species. Front Physiol 2018; 9:1831. [PMID: 30622478 PMCID: PMC6308295 DOI: 10.3389/fphys.2018.01831] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/06/2018] [Indexed: 01/09/2023] Open
Abstract
In a physiological setting, mitochondria increase oxidative phosphorylation during periods of stress to meet increased metabolic demand. This in part is mediated via enhanced mitochondrial Ca2+ uptake, an important regulator of cellular ATP homeostasis. In a pathophysiological setting pharmacological modulation of mitochondrial Ca2+ uptake or retention has been suggested as a therapeutic strategy to improve metabolic homeostasis or attenuate Ca2+-dependent arrhythmias in cardiac disease states. To explore the consequences of mitochondrial Ca2+ accumulation, we tested the effects of kaempferol, an activator of mitochondrial Ca2+ uniporter (MCU), CGP-37157, an inhibitor of mitochondrial Na+/Ca2+ exchanger, and MCU inhibitor Ru360 in rat ventricular myocytes (VMs) from control rats and rats with hypertrophy induced by thoracic aortic banding (TAB). In periodically paced VMs under β-adrenergic stimulation, treatment with kaempferol (10 μmol/L) or CGP-37157 (1 μmol/L) enhanced mitochondrial Ca2+ accumulation monitored by mitochondrial-targeted Ca2+ biosensor mtRCamp1h. Experiments with mitochondrial membrane potential-sensitive dye TMRM revealed this was accompanied by depolarization of the mitochondrial matrix. Using redox-sensitive OMM-HyPer and ERroGFP_iE biosensors, we found treatment with kaempferol or CGP-37157 increased the levels of reactive oxygen species (ROS) in mitochondria and the sarcoplasmic reticulum (SR), respectively. Confocal Ca2+ imaging showed that accelerated Ca2+ accumulation reduced Ca2+ transient amplitude and promoted generation of spontaneous Ca2+ waves in VMs paced under ISO, suggestive of abnormally high activity of the SR Ca2+ release channel ryanodine receptor (RyR). Western blot analyses showed increased RyR oxidation after treatment with kaempferol or CGP-37157 vs. controls. Furthermore, in freshly isolated TAB VMs, confocal Ca2+ imaging demonstrated that enhancement of mitochondrial Ca2+ accumulation further perturbed global Ca2+ handling, increasing the number of cells exhibiting spontaneous Ca2+ waves, shortening RyR refractoriness and decreasing SR Ca2+ content. In ex vivo optically mapped TAB hearts, kaempferol exacerbated proarrhythmic phenotype. On the contrary, incubation of cells with MCU inhibitor Ru360 (2 μmol/L, 30 min) normalized RyR oxidation state, improved intracellular Ca2+ homeostasis and reduced triggered activity in ex vivo TAB hearts. These findings suggest facilitation of mitochondrial Ca2+ uptake in cardiac disease can exacerbate proarrhythmic disturbances in Ca2+ homeostasis via ROS and enhanced activity of oxidized RyRs, while strategies to reduce mitochondrial Ca2+ accumulation can be protective.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Radmila Terentyeva
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Tae Yun Kim
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Peter Bronk
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Richard T. Clements
- Department of Surgery, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, United States
| | - Jin O-Uchi
- Lillehei Heart Institute University of Minnesota, Cancer and Cardiovascular Research Building, Minneapolis, MN, United States
| | - György Csordás
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bum-Rak Choi
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, United States
| |
Collapse
|