1
|
Medvedev RY, Afolabi SO, Turner DGP, Glukhov AV. Mechanisms of stretch-induced electro-anatomical remodeling and atrial arrhythmogenesis. J Mol Cell Cardiol 2024; 193:11-24. [PMID: 38797242 PMCID: PMC11260238 DOI: 10.1016/j.yjmcc.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Atrial fibrillation (AF) is the most common cardiac rhythm disorder, often occurring in the setting of atrial distension and elevated myocardialstretch. While various mechano-electrochemical signal transduction pathways have been linked to AF development and progression, the underlying molecular mechanisms remain poorly understood, hampering AF therapies. In this review, we describe different aspects of stretch-induced electro-anatomical remodeling as seen in animal models and in patients with AF. Specifically, we focus on cellular and molecular mechanisms that are responsible for mechano-electrochemical signal transduction and the development of ectopic beats triggering AF from pulmonary veins, the most common source of paroxysmal AF. Furthermore, we describe structural changes caused by stretch occurring before and shortly after the onset of AF as well as during AF progression, contributing to longstanding forms of AF. We also propose mechanical stretch as a new dimension to the concept "AF begets AF", in addition to underlying diseases. Finally, we discuss the mechanisms of these electro-anatomical alterations in a search for potential therapeutic strategies and the development of novel antiarrhythmic drugs targeted at the components of mechano-electrochemical signal transduction not only in cardiac myocytes, but also in cardiac non-myocyte cells.
Collapse
Affiliation(s)
- Roman Y Medvedev
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Saheed O Afolabi
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA; Department of Pharmacology and Therapeutics, University of Ilorin, Ilorin, Nigeria
| | - Daniel G P Turner
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Alexey V Glukhov
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
2
|
Meng M, Jiang Y, Wang Y, Huo R, Ma N, Shen X, Chang G. β-carotene targets IP3R/GRP75/VDAC1-MCU axis to renovate LPS-induced mitochondrial oxidative damage by regulating STIM1. Free Radic Biol Med 2023; 205:25-46. [PMID: 37270031 DOI: 10.1016/j.freeradbiomed.2023.05.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/03/2023] [Accepted: 05/18/2023] [Indexed: 06/05/2023]
Abstract
Endoplasmic reticulum (ER) and mitochondria are the main sites for the storage and regulation of Ca2+ homeostasis. An imbalance of Ca2+ homeostasis can cause ER stress and mitochondrial dysfunction, thereby inducing apoptosis. The store-operated calcium entry (SOCE) is the main channel for extracellular calcium influx. Mitochondria-associated endoplasmic reticulum (MAM) is an important agent for Ca2+ transfer from the ER to the mitochondria. Therefore, regulation of SOCE and MAMs has potential therapeutic value for disease prevention and treatment. In this study, bovine mammary epithelial cells (BMECs) and mice were used as models to explore the mechanisms of β-carotene to relieve ER stress and mitochondrial dysfunction. BAPTA-AM, EGTA (Ca2+ inhibitor), and BTP2 (SOCE channel inhibitor) alleviated ER stress and mitochondrial oxidative damage induced by increased intracellular Ca2+ levels after lipopolysaccharide (LPS) stimulation. Furthermore, inhibition of ER stress by 4-PBA (ER stress inhibitor), 2-APB (IP3R inhibitor), and ruthenium red (mitochondrial calcium uniporter (MCU) inhibitor) restored mitochondrial function by reducing mitochondrial ROS. Our data also confirm that β-carotene targeted STIM1 and IP3R channels to repair LPS-induced ER stress and mitochondrial disorders. Consistent with the in vitro study, in vito experiments in mice further showed that β-carotene attenuated LPS-induced ER stress and mitochondrial oxidative damage by inhibiting the expression of STIM1 and ORAI1, and reducing the level of Ca2+ in mouse mammary glands. Therefore, ER stress-mitochondrial oxidative damage mediated by the STIM1-ER-IP3R/GRP75/VDAC1-MCU axis plays an vital role in the development of mastitis. Our results provided novel ideas and therapeutic targets for the prevention and treatment of mastitis.
Collapse
Affiliation(s)
- Meijuan Meng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Yijin Jiang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Yan Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Ran Huo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Nana Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Xiangzhen Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Guangjun Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China.
| |
Collapse
|
3
|
Emrich SM, Yoast RE, Fike AJ, Bricker KN, Xin P, Zhang X, Rahman ZSM, Trebak M. The mitochondrial sodium/calcium exchanger NCLX (Slc8b1) in B lymphocytes. Cell Calcium 2022; 108:102667. [PMID: 36308855 DOI: 10.1016/j.ceca.2022.102667] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/20/2022] [Accepted: 10/18/2022] [Indexed: 01/25/2023]
Abstract
Antigen receptor stimulation triggers cytosolic Ca2+ signals, which activate transcriptional and metabolic programs critical for immune function. B-cell receptor (BCR) engagement causes rapid cytosolic Ca2+ rise through the ubiquitous store-operated calcium entry (SOCE) pathway. Slc8b1, which encodes the mitochondrial Na+/Ca2+ exchanger (NCLX), extrudes Ca2+ out of the mitochondria and maintains optimal SOCE activity. Inhibition of NCLX in DT40 and A20 B lymphocyte lines was recently shown to impair cytosolic Ca2+ transients in response to antigen-receptor stimulation, however the downstream functional consequences of this impairment remain unclear. Here, we generated Slc8b1 knockout A20 B-cell lines using CRISPR/Cas9 technology and B-cell specific Slc8b1 knockout mice. Surprisingly, while loss of Slc8b1 in B lymphocytes led to reduction in SOCE, it had a marginal effect on mitochondrial Ca2+ extrusion, suggesting that NCLX is not the major mitochondrial Ca2+ extrusion mechanism in B cells. Furthermore, endoplasmic reticulum (ER) Ca2+ content and rates of ER depletion and refilling remained unaltered in Slc8b1 knockout B cells. Slc8b1 deficiency increased mitochondrial production of oxidants, reduced mitochondrial bioenergetics and altered mitochondrial ultrastructure. B-cell specific Slc8b1 knockout mice showed reduced germinal center B cell responses following foreign antigen and pathogen driven immune responses. Our studies provide novel insights into the function of Slc8b1 in germinal center B cells and its contribution to B-cell signaling and effector function.
Collapse
Affiliation(s)
- Scott M Emrich
- Department of Cellular and Molecular Physiology, the Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Ryan E Yoast
- Department of Cellular and Molecular Physiology, the Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Adam J Fike
- Department of Microbiology and Immunology, the Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Kristen N Bricker
- Department of Microbiology and Immunology, the Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Ping Xin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA
| | - Xuexin Zhang
- Department of Cellular and Molecular Physiology, the Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Ziaur S M Rahman
- Department of Microbiology and Immunology, the Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, the Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA.
| |
Collapse
|
4
|
Peyronnet R, Desai A, Edelmann JC, Cameron BA, Emig R, Kohl P, Dean D. Simultaneous assessment of radial and axial myocyte mechanics by combining atomic force microscopy and carbon fibre techniques. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210326. [PMID: 36189808 PMCID: PMC9527909 DOI: 10.1098/rstb.2021.0326] [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: 03/31/2022] [Accepted: 07/21/2022] [Indexed: 11/12/2022] Open
Abstract
Cardiomyocytes sense and shape their mechanical environment, contributing to its dynamics by their passive and active mechanical properties. While axial forces generated by contracting cardiomyocytes have been amply investigated, the corresponding radial mechanics remain poorly characterized. Our aim is to simultaneously monitor passive and active forces, both axially and radially, in cardiomyocytes freshly isolated from adult mouse ventricles. To do so, we combine a carbon fibre (CF) set-up with a custom-made atomic force microscope (AFM). CF allows us to apply stretch and to record passive and active forces in the axial direction. The AFM, modified for frontal access to fit in CF, is used to characterize radial cell mechanics. We show that stretch increases the radial elastic modulus of cardiomyocytes. We further find that during contraction, cardiomyocytes generate radial forces that are reduced, but not abolished, when cells are forced to contract near isometrically. Radial forces may contribute to ventricular wall thickening during contraction, together with the dynamic re-orientation of cells and sheetlets in the myocardium. This new approach for characterizing cell mechanics allows one to obtain a more detailed picture of the balance of axial and radial mechanics in cardiomyocytes at rest, during stretch, and during contraction. 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)
- Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad Krozingen, University of Freiburg, 79110 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | - Breanne A. Cameron
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad Krozingen, University of Freiburg, 79110 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Ramona Emig
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad Krozingen, University of Freiburg, 79110 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad Krozingen, University of Freiburg, 79110 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
- National Heart and Lung Institute, Imperial College London, London, UK
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | | |
Collapse
|
5
|
Kamkin AG, Kamkina OV, Shim AL, Bilichenko A, Mitrokhin VM, Kazansky VE, Filatova TS, Abramochkin D, Mladenov MI. The role of activation of two different sGC binding sites by NO-dependent and NO-independent mechanisms in the regulation of SACs in rat ventricular cardiomyocytes. Physiol Rep 2022; 10:e15246. [PMID: 35384354 PMCID: PMC8981922 DOI: 10.14814/phy2.15246] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 04/18/2023] Open
Abstract
The mechanoelectrical feedback (MEF) mechanism in the heart that plays a significant role in the occurrence of arrhythmias, involves cation flux through cation nonselective stretch-activated channels (SACs). It is well known that nitric oxide (NO) can act as a regulator of MEF. Here we addressed the possibility of SAC's regulation along NO-dependent and NO-independent pathways, as well as the possibility of S-nitrosylation of SACs. In freshly isolated rat ventricular cardiomyocytes, using the patch-clamp method in whole-cell configuration, inward nonselective stretch-activated cation current ISAC was recorded through SACs, which occurs during dosed cell stretching. NO donor SNAP, α1-subunit of sGC activator BAY41-2272, sGC blocker ODQ, PKG blocker KT5823, PKG activator 8Br-cGMP, and S-nitrosylation blocker ascorbic acid, were employed. We concluded that the physiological concentration of NO in the cell is a necessary condition for the functioning of SACs. An increase in NO due to SNAP in an unstretched cell causes the appearance of a Gd3+ -sensitive nonselective cation current, an analog of ISAC , while in a stretched cell it eliminates ISAC . The NO-independent pathway of sGC activation of α subunit, triggered by BAY41-2272, is also important for the regulation of SACs. Since S-nitrosylation inhibitor completely abolishes ISAC , this mechanism occurs. The application of BAY41-2272 cannot induce ISAC in a nonstretched cell; however, the addition of SNAP on its background activates SACs, rather due to S-nitrosylation. ODQ eliminates ISAC , but SNAP added on the background of stretch increases ISAC in addition to ODQ. This may be a result of the lack of NO as a result of inhibition of NOS by metabolically modified ODQ. KT5823 reduces PKG activity and reduces SACs phosphorylation, leading to an increase in ISAC . 8Br-cGMP reduces ISAC by activating PKG and its phosphorylation. These results demonstrate a significant contribution of S-nitrosylation to the regulation of SACs.
Collapse
Affiliation(s)
- Andre G. Kamkin
- Department of PhysiologyPirogov Russian National Research Medical UniversityMoscowRussia
| | - Olga V. Kamkina
- Department of PhysiologyPirogov Russian National Research Medical UniversityMoscowRussia
| | - Andrey L. Shim
- Department of PhysiologyPirogov Russian National Research Medical UniversityMoscowRussia
| | - Andrey Bilichenko
- Department of PhysiologyPirogov Russian National Research Medical UniversityMoscowRussia
| | - Vadim M. Mitrokhin
- Department of PhysiologyPirogov Russian National Research Medical UniversityMoscowRussia
| | - Viktor E. Kazansky
- Department of PhysiologyPirogov Russian National Research Medical UniversityMoscowRussia
| | - Tatiana S. Filatova
- Department of PhysiologyPirogov Russian National Research Medical UniversityMoscowRussia
- Department of Human and Animal PhysiologyLomonosov Moscow State UniversityMoscowRussia
| | - Denis V. Abramochkin
- Department of PhysiologyPirogov Russian National Research Medical UniversityMoscowRussia
- Department of Human and Animal PhysiologyLomonosov Moscow State UniversityMoscowRussia
| | - Mitko I. Mladenov
- Department of PhysiologyPirogov Russian National Research Medical UniversityMoscowRussia
- Faculty of Natural Sciences and MathematicsInstitute of Biology, “Ss. Cyril and Methodius” UniversitySkopjeMacedonia
| |
Collapse
|
6
|
Quinn TA, Kohl P. Cardiac Mechano-Electric Coupling: Acute Effects of Mechanical Stimulation on Heart Rate and Rhythm. Physiol Rev 2020; 101:37-92. [PMID: 32380895 DOI: 10.1152/physrev.00036.2019] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The heart is vital for biological function in almost all chordates, including humans. It beats continually throughout our life, supplying the body with oxygen and nutrients while removing waste products. If it stops, so does life. The heartbeat involves precise coordination of the activity of billions of individual cells, as well as their swift and well-coordinated adaption to changes in physiological demand. Much of the vital control of cardiac function occurs at the level of individual cardiac muscle cells, including acute beat-by-beat feedback from the local mechanical environment to electrical activity (as opposed to longer term changes in gene expression and functional or structural remodeling). This process is known as mechano-electric coupling (MEC). In the current review, we present evidence for, and implications of, MEC in health and disease in human; summarize our understanding of MEC effects gained from whole animal, organ, tissue, and cell studies; identify potential molecular mediators of MEC responses; and demonstrate the power of computational modeling in developing a more comprehensive understanding of ‟what makes the heart tick.ˮ.
Collapse
Affiliation(s)
- T Alexander Quinn
- Department of Physiology and Biophysics and School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Medical Faculty of the University of Freiburg, Freiburg, Germany; and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Peter Kohl
- Department of Physiology and Biophysics and School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Medical Faculty of the University of Freiburg, Freiburg, Germany; and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Le QA, Kim JC, Kim KH, Van Vu AT, Woo SH. Distinct shear-induced Ca 2+ signaling in the left and right atrial myocytes: Role of P2 receptor context. J Mol Cell Cardiol 2020; 143:38-50. [PMID: 32305361 DOI: 10.1016/j.yjmcc.2020.04.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/05/2020] [Accepted: 04/11/2020] [Indexed: 11/18/2022]
Abstract
Atrial myocytes are continuously exposed to shear stress during cardiac cycles. Previous reports have shown that shear stress induces two different types of global Ca2+ signaling in atrial myocytes-longitudinal Ca2+ waves (L-waves) and action potential-involved transverse waves (T-waves), and suggested an underlying role of the autocrine activation of P2 receptors. We explored the correlations between ATP release and Ca2+ wave generation in atrial myocytes and investigated why the cells develop two Ca2+-wave types during the same shear force. We examined whether ATP release correlates with different shear-stress (~16 dyn/cm2)-mediated Ca2+ signaling by simultaneous measurement of local Ca2+ and ATP release in individual atrial myocytes using two-dimensional confocal imaging and sniffer patch techniques, respectively. Functional P2X7-receptor-expressing HEK293 cells were established as sniffer cells, which generated currents in real time in response to ATP released from a closely positioned atrial myocyte. Both shear-stress-induced L- and T-waves were preceded by sniffer currents with no difference in the current magnitude. Left atrial (LA) myocytes had two- to three-fold larger sniffer currents than right atrial (RA) cells, as was confirmed by ATP chemiluminescence assay. Shear-stress-induced ATP release was eliminated by connexin (Cx) 43 hemichannel inhibition using La3+, Gap19, or knock-down of Cx43 expression. The level of phosphorylated Cx43 at Ser386 (p-Cx43Ser368), but not total Cx43, was higher in LA versus RA myocytes. Most LA cells (~70%) developed L-waves, whereas most RA myocytes (~80%) presented T-waves. Shear-stress-induced T-waves were completely removed by inhibition of P2X4R, which were most abundant in rat atrial cells. Expression of P2X4R was higher in RA than LA myocytes, whereas expression of P2Y1R, the mediator of L-waves, was higher in LA than RA myocytes. ATP release mainly triggers L-waves in LA myocytes and T-waves in RA myocytes under the same shear force, partly because of the differential expression of P2Y1R and P2X4R between LA and RA myocytes. Higher ATP release in LA myocytes under shear stress may not contribute to determination of the wave pattern.
Collapse
Affiliation(s)
- Qui Anh Le
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, South Korea
| | - Joon-Chul Kim
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, South Korea
| | - Kyeong-Hee Kim
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, South Korea
| | - Anh Thi Van Vu
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, South Korea
| | - Sun-Hee Woo
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, South Korea.
| |
Collapse
|
8
|
Izu LT, Kohl P, Boyden PA, Miura M, Banyasz T, Chiamvimonvat N, Trayanova N, Bers DM, Chen-Izu Y. Mechano-electric and mechano-chemo-transduction in cardiomyocytes. J Physiol 2020; 598:1285-1305. [PMID: 31789427 PMCID: PMC7127983 DOI: 10.1113/jp276494] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiac excitation-contraction (E-C) coupling is influenced by (at least) three dynamic systems that couple and feedback to one another (see Abstract Figure). Here we review the mechanical effects on cardiomyocytes that include mechano-electro-transduction (commonly referred to as mechano-electric coupling, MEC) and mechano-chemo-transduction (MCT) mechanisms at cell and molecular levels which couple to Ca2+ -electro and E-C coupling reviewed elsewhere. These feedback loops from muscle contraction and mechano-transduction to the Ca2+ homeodynamics and to the electrical excitation are essential for understanding the E-C coupling dynamic system and arrhythmogenesis in mechanically loaded hearts. This white paper comprises two parts, each reflecting key aspects from the 2018 UC Davis symposium: MEC (how mechanical load influences electrical dynamics) and MCT (how mechanical load alters cell signalling and Ca2+ dynamics). Of course, such separation is artificial since Ca2+ dynamics profoundly affect ion channels and electrogenic transporters and vice versa. In time, these dynamic systems and their interactions must become fully integrated, and that should be a goal for a comprehensive understanding of how mechanical load influences cell signalling, Ca2+ homeodynamics and electrical dynamics. In this white paper we emphasize current understanding, consensus, controversies and the pressing issues for future investigations. Space constraints make it impossible to cover all relevant articles in the field, so we will focus on the topics discussed at the symposium.
Collapse
Affiliation(s)
- Leighton T. Izu
- Department of Pharmacology, University of California, Davis, CA 95616, USA
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre, and Faculty of Medicine, University of Freiburg, D-79110, Germany
| | | | - Masahito Miura
- Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Tamas Banyasz
- Department of Physiology, University of Debrecen, Debrecen, Hungary
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Cardiovascular Medicine, University of California, Davis, USA
| | - Natalia Trayanova
- Department of Department of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California, Davis, CA 95616, USA
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, CA 95616, USA
- Department of Internal Medicine, Cardiovascular Medicine, University of California, Davis, USA
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| |
Collapse
|
9
|
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
|
10
|
Kim JC, Son MJ, Woo SH. Regulation of cardiac calcium by mechanotransduction: Role of mitochondria. Arch Biochem Biophys 2018; 659:33-41. [DOI: 10.1016/j.abb.2018.09.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/28/2018] [Indexed: 12/27/2022]
|
11
|
Rog-Zielinska EA, O'Toole ET, Hoenger A, Kohl P. Mitochondrial Deformation During the Cardiac Mechanical Cycle. Anat Rec (Hoboken) 2018; 302:146-152. [PMID: 30302911 PMCID: PMC6312496 DOI: 10.1002/ar.23917] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/22/2018] [Accepted: 05/11/2018] [Indexed: 12/17/2022]
Abstract
Cardiomyocytes both cause and experience continual cyclic deformation. The exact effects of this deformation on the properties of intracellular organelles are not well characterized, although they are likely to be relevant for cardiomyocyte responses to active and passive changes in their mechanical environment. In the present study we provide three‐dimensional ultrastructural evidence for mechanically induced mitochondrial deformation in rabbit ventricular cardiomyocytes over a range of sarcomere lengths representing myocardial tissue stretch, an unloaded “slack” state, and contracture. We also show structural indications for interaction of mitochondria with one another, as well as with other intracellular elements such as microtubules, sarcoplasmic reticulum and T‐tubules. The data presented here help to contextualize recent reports on the mechanosensitivity and cell‐wide connectivity of the mitochondrial network and provide a structural framework that may aide interpretation of mechanically‐regulated molecular signaling in cardiac cells. Anat Rec, 302:146–152, 2019. © 2018 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- E A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - E T O'Toole
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, Colorado, USA
| | - A Hoenger
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, Colorado, USA
| | - P Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
12
|
Yang Y, Luo L, Cai X, Fang Y, Wang J, Chen G, Yang J, Zhou Q, Sun X, Cheng X, Yan H, Lu W, Hu C, Cao P. Nrf2 inhibits oxaliplatin-induced peripheral neuropathy via protection of mitochondrial function. Free Radic Biol Med 2018. [PMID: 29530794 DOI: 10.1016/j.freeradbiomed.2018.03.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Oxaliplatin-induced peripheral neuropathy (OIPN) is a severe, dose-limiting toxicity associated with cancer chemotherapy. The efficacy of antioxidant administration in OIPN is debatable, as the promising preliminary results obtained with a number of antioxidants have not been confirmed in larger clinical trials. Besides its antioxidant activity, the transcription factor, nuclear factor-erythroid 2 (NF-E2) p45-related factor 2 (Nrf2) plays a crucial role in the maintenance of mitochondrial homeostasis, and mitochondrial dysfunction is a key contributor to OIPN. Here, we have investigated the protective properties of Nrf2 in OIPN. Nrf2-/- mice displayed severe mechanical allodynia and cold sensitivity and thus experienced increased peripheral nervous system injury compared to Nrf2+/+ mice. Furthermore, Nrf2 knockout aggravated oxaliplatin-induced reactive oxygen species production, decreased the mitochondrial membrane potential, led to abnormal intracellular calcium levels, and induced cytochrome c-related apoptosis and overexpression of the TRP protein family. Sulforaphane-induced activation of the Nrf2 signaling pathway alleviated morphological alterations, mitochondrial dysfunction in dorsal root ganglion neurons, and nociceptive sensations in mice. Our findings reveal that Nrf2 may play a critical role in ameliorating OIPN, through protection of mitochondrial function by alleviating oxidative stress and inhibiting TRP protein family expression. This suggests that pharmacological or therapeutic activation of Nrf2 may be used to prevent or slow down the progression of OIPN.
Collapse
Affiliation(s)
- Yang Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210097, China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210097, China
| | - Xueting Cai
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Yuan Fang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Jiaqi Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Gang Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Jie Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Qian Zhou
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Xiaoyan Sun
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Xiaolan Cheng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Huaijiang Yan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Wuguang Lu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Chunping Hu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, #100 Shizi Street, Hongshan Road, Nanjing 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, Jiangsu, China.
| |
Collapse
|
13
|
Gambardella J, Sorriento D, Ciccarelli M, Del Giudice C, Fiordelisi A, Napolitano L, Trimarco B, Iaccarino G, Santulli G. Functional Role of Mitochondria in Arrhythmogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:191-202. [PMID: 28551788 DOI: 10.1007/978-3-319-55330-6_10] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Growing evidence indicate that mitochondria play a functional role in arrhythmogenesis. We report here the molecular mechanisms underlying the action of these highly dynamic organelles in the regulation of cell metabolism, action potential and, overall, heart excitability. In particular, we examine the role of cardiac mitochondria in linking metabolism and cell excitability. The importance of the main mitochondrial channels is evaluated as well, including the recently identified calcium uniporter. Promises and pitfalls of potential therapeutic strategies targeting mitochondrial pathways are also assessed.
Collapse
Affiliation(s)
- Jessica Gambardella
- Columbia University Medical Center, New York, NY, USA.,Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Carmine Del Giudice
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Luigi Napolitano
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Guido Iaccarino
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Gaetano Santulli
- Dept. of Biomedical Advanced Sciences, Federico II University, Naples, Italy. .,Columbia University Medical Center, New York Presbyterian Hospital-Manhattan, New York, NY, USA.
| |
Collapse
|
14
|
Quinn TA, Kohl P. Comparing maximum rate and sustainability of pacing by mechanical vs. electrical stimulation in the Langendorff-perfused rabbit heart. Europace 2017; 18:iv85-iv93. [PMID: 28011835 PMCID: PMC5400084 DOI: 10.1093/europace/euw354] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/01/2016] [Indexed: 01/04/2023] Open
Abstract
Aims Mechanical stimulation (MS) represents a readily available, non-invasive means of pacing the asystolic or bradycardic heart in patients, but benefits of MS at higher heart rates are unclear. Our aim was to assess the maximum rate and sustainability of excitation by MS vs. electrical stimulation (ES) in the isolated heart under normal physiological conditions. Methods and results Trains of local MS or ES at rates exceeding intrinsic sinus rhythm (overdrive pacing; lowest pacing rates 2.5±0.5 Hz) were applied to the same mid-left ventricular free-wall site on the epicardium of Langendorff-perfused rabbit hearts. Stimulation rates were progressively increased, with a recovery period of normal sinus rhythm between each stimulation period. Trains of MS caused repeated focal ventricular excitation from the site of stimulation. The maximum rate at which MS achieved 1:1 capture was lower than during ES (4.2±0.2 vs. 5.9±0.2 Hz, respectively). At all overdrive pacing rates for which repetitive MS was possible, 1:1 capture was reversibly lost after a finite number of cycles, even though same-site capture by ES remained possible. The number of MS cycles until loss of capture decreased with rising stimulation rate. If interspersed with ES, the number of MS to failure of capture was lower than for MS only. Conclusion In this study, we demonstrate that the maximum pacing rate at which MS can be sustained is lower than that for same-site ES in isolated heart, and that, in contrast to ES, the sustainability of successful 1:1 capture by MS is limited. The mechanism(s) of differences in MS vs. ES pacing ability, potentially important for emergency heart rhythm management, are currently unknown, thus warranting further investigation.
Collapse
Affiliation(s)
- T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, 5850 College St, Halifax, NS B3H 4R2, Canada
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Medical School of the University of Freiburg, Elsaesser Str 2Q, 79110 Freiburg, Germany.,National Heart and Lung Institute, Imperial College London, The Magdi Yacoub Institute, Hill End Road, UB9 6JH London, UK
| |
Collapse
|
15
|
MacDonald EA, Stoyek MR, Rose RA, Quinn TA. Intrinsic regulation of sinoatrial node function and the zebrafish as a model of stretch effects on pacemaking. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 130:198-211. [PMID: 28743586 DOI: 10.1016/j.pbiomolbio.2017.07.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/17/2017] [Accepted: 07/21/2017] [Indexed: 12/18/2022]
Abstract
Excitation of the heart occurs in a specialised region known as the sinoatrial node (SAN). Tight regulation of SAN function is essential for the maintenance of normal heart rhythm and the response to (patho-)physiological changes. The SAN is regulated by extrinsic (central nervous system) and intrinsic (neurons, peptides, mechanics) factors. The positive chronotropic response to stretch in particular is essential for beat-by-beat adaptation to changes in hemodynamic load. Yet, the mechanism of this stretch response is unknown, due in part to the lack of an appropriate experimental model for targeted investigations. We have been investigating the zebrafish as a model for the study of intrinsic regulation of SAN function. In this paper, we first briefly review current knowledge of the principal components of extrinsic and intrinsic SAN regulation, derived primarily from experiments in mammals, followed by a description of the zebrafish as a novel experimental model for studies of intrinsic SAN regulation. This mini-review is followed by an original investigation of the response of the zebrafish isolated SAN to controlled stretch. Stretch causes an immediate and continuous increase in beating rate in the zebrafish isolated SAN. This increase reaches a maximum part way through a period of sustained stretch, with the total change dependent on the magnitude and direction of stretch. This is comparable to what occurs in isolated SAN from most mammals (including human), suggesting that the zebrafish is a novel experimental model for the study of mechanisms involved in the intrinsic regulation of SAN function by mechanical effects.
Collapse
Affiliation(s)
- Eilidh A MacDonald
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Matthew R Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada; School of Biomedical Engineering, Dalhousie University, Halifax, Canada.
| |
Collapse
|
16
|
Kim JC, Son MJ, Wang J, Woo SH. Regulation of cardiac Ca 2+ and ion channels by shear mechanotransduction. Arch Pharm Res 2017; 40:783-795. [PMID: 28702845 DOI: 10.1007/s12272-017-0929-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 07/06/2017] [Indexed: 11/25/2022]
Abstract
Cardiac contraction is controlled by a Ca2+ signaling sequence that includes L-type Ca2+ current-gated opening of Ca2+ release channels (ryanodine receptors) in the sarcoplasmic reticulum (SR). Local Ca2+ signaling in the atrium differs from that in the ventricle because atrial myocytes lack transverse tubules and have more abundant corbular SR. Myocardium is subjected to a variety of forces with each contraction, such as stretch, shear stress, and afterload, and adapts to those mechanical stresses. These mechanical stimuli increase in heart failure, hypertension, and valvular heart diseases that are clinically implicated in atrial fibrillation and stroke. In the present review, we describe distinct responses of atrial and ventricular myocytes to shear stress and compare them with other mechanical responses in the context of local and global Ca2+ signaling and ion channel regulation. Recent evidence suggests that shear mechanotransduction in cardiac myocytes involves activation of gap junction hemichannels, purinergic signaling, and generation of mitochondrial reactive oxygen species. Significant alterations in Ca2+ signaling and ionic currents by shear stress may be implicated in the pathogenesis of cardiac arrhythmia and failure.
Collapse
Affiliation(s)
- Joon-Chul Kim
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 305-764, South Korea
| | - Min-Jeong Son
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 305-764, South Korea
| | - Jun Wang
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 305-764, South Korea
| | - Sun-Hee Woo
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 305-764, South Korea.
| |
Collapse
|
17
|
Altered Mitochondrial Metabolism and Mechanosensation in the Failing Heart: Focus on Intracellular Calcium Signaling. Int J Mol Sci 2017; 18:ijms18071487. [PMID: 28698526 PMCID: PMC5535977 DOI: 10.3390/ijms18071487] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 06/28/2017] [Accepted: 07/04/2017] [Indexed: 12/26/2022] Open
Abstract
The heart consists of millions of cells, namely cardiomyocytes, which are highly organized in terms of structure and function, at both macroscale and microscale levels. Such meticulous organization is imperative for assuring the physiological pump-function of the heart. One of the key players for the electrical and mechanical synchronization and contraction is the calcium ion via the well-known calcium-induced calcium release process. In cardiovascular diseases, the structural organization is lost, resulting in morphological, electrical, and metabolic remodeling owing the imbalance of the calcium handling and promoting heart failure and arrhythmias. Recently, attention has been focused on the role of mitochondria, which seem to jeopardize these events by misbalancing the calcium processes. In this review, we highlight our recent findings, especially the role of mitochondria (dys)function in failing cardiomyocytes with respect to the calcium machinery.
Collapse
|
18
|
Mechano-sensitivity of mitochondrial function in mouse cardiac myocytes. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 130:315-322. [PMID: 28668597 DOI: 10.1016/j.pbiomolbio.2017.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/19/2017] [Accepted: 05/23/2017] [Indexed: 10/19/2022]
Abstract
Mitochondria are an important source of reactive oxygen species (ROS). Although it has been reported that myocardial stretch increases cellular ROS production by activating nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2), referred to as X-ROS signalling, the involvement of mitochondria in X-ROS is not clear. Mitochondria are organelles that generate adenosine triphosphate (ATP) for cellular energy needs, which are mechanical-load-dependent. Therefore, it would not be surprising if these organelles had mechano-sensitive functions associated with stretch-induced ROS production. In the present study, we investigated the relation between X-ROS and mitochondrial stretch-sensitive responses in isolated mouse cardiac myocytes. The cells were subjected to 10% axial stretch using computer-controlled, piezo-manipulated carbon fibres attached to both cell ends. Cellular ROS production and mitochondrial membrane potential (Δψm) were assessed optically by confocal microscopy. The axial stretch increased ROS production and hyperpolarised Δψm. Treatment with a mitochondrial metabolic uncoupler, carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP), at 0.5 μM did not suppress stretch-induced ROS production, whereas treatment with a respiratory Complex III inhibitor, antimycin A (5 μM), blunted the response. Although NOX inhibition by apocynin abrogated the stretch-induced ROS production, it did not suppress stretch-induced hyperpolarisation of Δψm. These results suggest that stretch causes activation of the respiratory chain to hyperpolarise Δψm, followed by NOX activation, which increases ROS production.
Collapse
|
19
|
Schönleitner P, Schotten U, Antoons G. Mechanosensitivity of microdomain calcium signalling in the heart. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017. [PMID: 28648626 DOI: 10.1016/j.pbiomolbio.2017.06.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In cardiac myocytes, calcium (Ca2+) signalling is tightly controlled in dedicated microdomains. At the dyad, i.e. the narrow cleft between t-tubules and junctional sarcoplasmic reticulum (SR), many signalling pathways combine to control Ca2+-induced Ca2+ release during contraction. Local Ca2+ gradients also exist in regions where SR and mitochondria are in close contact to regulate energetic demands. Loss of microdomain structures, or dysregulation of local Ca2+ fluxes in cardiac disease, is often associated with oxidative stress, contractile dysfunction and arrhythmias. Ca2+ signalling at these microdomains is highly mechanosensitive. Recent work has demonstrated that increasing mechanical load triggers rapid local Ca2+ releases that are not reflected by changes in global Ca2+. Key mechanisms involve rapid mechanotransduction with reactive oxygen species or nitric oxide as primary signalling molecules targeting SR or mitochondria microdomains depending on the nature of the mechanical stimulus. This review summarizes the most recent insights in rapid Ca2+ microdomain mechanosensitivity and re-evaluates its (patho)physiological significance in the context of historical data on the macroscopic role of Ca2+ in acute force adaptation and mechanically-induced arrhythmias. We distinguish between preload and afterload mediated effects on local Ca2+ release, and highlight differences between atrial and ventricular myocytes. Finally, we provide an outlook for further investigation in chronic models of abnormal mechanics (eg post-myocardial infarction, atrial fibrillation), to identify the clinical significance of disturbed Ca2+ mechanosensitivity for arrhythmogenesis.
Collapse
Affiliation(s)
- Patrick Schönleitner
- Dept of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Uli Schotten
- Dept of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Gudrun Antoons
- Dept of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands.
| |
Collapse
|
20
|
A chemical chaperone improves muscle function in mice with a RyR1 mutation. Nat Commun 2017; 8:14659. [PMID: 28337975 PMCID: PMC5376670 DOI: 10.1038/ncomms14659] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 01/20/2017] [Indexed: 12/28/2022] Open
Abstract
Mutations in the RYR1 gene cause severe myopathies. Mice with an I4895T mutation in the type 1 ryanodine receptor/Ca2+ release channel (RyR1) display muscle weakness and atrophy, but the underlying mechanisms are unclear. Here we show that the I4895T mutation in RyR1 decreases the amplitude of the sarcoplasmic reticulum (SR) Ca2+ transient, resting cytosolic Ca2+ levels, muscle triadin content and calsequestrin (CSQ) localization to the junctional SR, and increases endoplasmic reticulum (ER) stress/unfolded protein response (UPR) and mitochondrial ROS production. Treatment of mice carrying the I4895T mutation with a chemical chaperone, sodium 4-phenylbutyrate (4PBA), reduces ER stress/UPR and improves muscle function, but does not restore SR Ca2+ transients in I4895T fibres to wild type levels, suggesting that decreased SR Ca2+ release is not the major driver of the myopathy. These findings suggest that 4PBA, an FDA-approved drug, has potential as a therapeutic intervention for RyR1 myopathies that are associated with ER stress. Mutations in the RyR1 channel cause core myopathies. Here the authors show that ER stress and the unfolded protein response underlie the pathology caused by a common RyR1 channel mutation, and show that treatment with a chemical chaperone restores muscle function in mice.
Collapse
|
21
|
Kim JC, Wang J, Son MJ, Woo SH. Shear stress enhances Ca 2+ sparks through Nox2-dependent mitochondrial reactive oxygen species generation in rat ventricular myocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1121-1131. [PMID: 28213332 DOI: 10.1016/j.bbamcr.2017.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/01/2017] [Accepted: 02/12/2017] [Indexed: 02/06/2023]
Abstract
Shear stress enhances diastolic and systolic Ca2+ concentration in ventricular myocytes. Here, using confocal Ca2+ imaging in rat ventricular myocytes, we assessed the effects of shear stress (~16dyn/cm2) on the frequency of spontaneous Ca2+ sparks and explored the mechanism underlying shear-mediated Ca2+ spark regulation. The frequency of Ca2+ sparks was immediately increased by shear stress (by ~80%), and increased further (by ~150%) during prolonged exposure (20s). The 2-D size and duration of individual sparks were increased by shear stimulation. Inhibition of nitric oxide synthase (NOS) only partially attenuated the prolonged shear-mediated enhancement in spark frequency. Pretreatment with antioxidants significantly attenuated the short- and long-term effects of shear on spark frequency. Microtubule or nicotinamide adenine dinucleotide phosphate oxidase 2 (Nox2) inhibition abolished the immediate shear-induced increase in spark frequency and suppressed the effects of prolonged exposure to shear stress by ~70%. Scavenging of mitochondrial reactive oxygen species (ROS) and mitochondrial uncoupling also abolished the effect of short-term shear on spark occurrence, and markedly reduced (by ~80%) the effects of prolonged shear. Mitochondrial ROS levels increased under shear; this was eliminated by blocking Nox2. Sarcoplasmic reticulum Ca2+ content was increased only by prolonged shear. Our data suggest that shear stress enhances ventricular spark frequency mainly via ROS generated from mitochondria through Nox2, and that NOS and higher sarcoplasmic reticulum Ca2+ concentrations may also contribute to the enhancement of Ca2+ sparks under shear stress. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Joon-Chul Kim
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 305-764, South Korea
| | - Jun Wang
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 305-764, South Korea
| | - Min-Jeong Son
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 305-764, South Korea
| | - Sun-Hee Woo
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 305-764, South Korea.
| |
Collapse
|
22
|
Miragoli M, Cabassi A. Mitochondrial Mechanosensor Microdomains in Cardiovascular Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:247-264. [PMID: 28551791 DOI: 10.1007/978-3-319-55330-6_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The cardiomyocytes populating the 'working myocardium' are highly organized and such organization ranges from macroscale (e.g. the geometrical rod shape) to microscale (dyad/t-tubules) domains. This meticulous level of organization is imperative for assuring the normal and physiological pump-function of the heart. In the pathological cardiac tissue, the domains-related architecture is partially lost, resulting in morphological, electrical and metabolic remodeling and promoting cardiovascular diseases including heart failure and arrhythmias. Indeed, arrhythmogenesis during heart failure is a major clinical problem. Arrhythmias have been extensively studied from an electrical etiology, but only recently, physiologists and scientists have focused their attention on cellular and subcellular mechanosensors. We and others have investigated whether the nanoscale mechanosensitive properties of cardiomyocytes from failing hearts have a bearing upon the initiation of abnormal electrical activity. This chapter highlights the recent findings in the field, especially the role of mitochondria function and alignment in failing cardiomyocytes interrogated via nanomechanical stimuli.
Collapse
Affiliation(s)
- Michele Miragoli
- Department of Medicine and Surgery, University of Parma, Parma, 43124, Italy. .,Humanitas Clinical and Research Center, Rozzano, MI, Italy.
| | - Aderville Cabassi
- Department of Medicine and Surgery, University of Parma, Parma, 43124, Italy
| |
Collapse
|
23
|
Yao H, Fan R, Zhao X, Zhao W, Liu W, Yang J, Sattar H, Zhao J, Zhang Z, Xu S. Selenoprotein W redox-regulated Ca2+ channels correlate with selenium deficiency-induced muscles Ca2+ leak. Oncotarget 2016; 7:57618-57632. [PMID: 27557522 PMCID: PMC5295377 DOI: 10.18632/oncotarget.11459] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 08/17/2016] [Indexed: 11/25/2022] Open
Abstract
Selenium (Se) deficiency induces Ca2+ leak and calcification in mammal skeletal muscles; however, the exact mechanism is still unclear. In the present study, both Se-deficient chicken muscle models and selenoprotein W (SelW) gene knockdown myoblast and embryo models were used to study the mechanism. The results showed that Se deficiency-induced typical muscular injuries accompanied with Ca2+ leak and oxidative stress (P < 0.05) injured the ultrastructure of the sarcoplasmic reticulum (SR) and mitochondria; decreased the levels of the Ca2+ channels, SERCA, SLC8A, CACNA1S, ORAI1, STIM1, TRPC1, and TRPC3 (P < 0.05); and increased the levels of Ca2+ channel PMCA (P < 0.05). Similarly, SelW knockdown also induced Ca2+ leak from the SR and cytoplasm; increased mitochondrial Ca2+ levels and oxidative stress; injured SR and mitochondrial ultrastructure; decreased levels of SLC8A, CACNA1S, ORA1, TRPC1, and TRPC3; and caused abnormal activities of Ca2+ channels in response to inhibitors in myoblasts and chicken embryos. Thus, both Se deficiency and SelW knockdown induced Ca2+ leak, oxidative stress, and Ca2+ channel reduction. In addition, Ca2+ levels and the expression of the Ca2+ channels, RyR1, SERCA, CACNA1S, TRPC1, and TRPC3 were recovered to normal levels by N-acetyl-L-cysteine (NAC) treatment compared with SelW knockdown cells. Thus, with regard to the decreased Ca2+ channels, SelW knockdown closely correlated Se deficiency with Ca2+ leak in muscles. The redox regulation role of SelW is crucial in Se deficiency-induced Ca2+ leak in muscles.
Collapse
Affiliation(s)
- Haidong Yao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Ruifeng Fan
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Xia Zhao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Wenchao Zhao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Wei Liu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Heilongjiang, P. R. China
| | - Jie Yang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Hamid Sattar
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Jinxin Zhao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Ziwei Zhang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Shiwen Xu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| |
Collapse
|
24
|
Son MJ, Kim JC, Kim SW, Chidipi B, Muniyandi J, Singh TD, So I, Subedi KP, Woo SH. Shear stress activates monovalent cation channel transient receptor potential melastatin subfamily 4 in rat atrial myocytes via type 2 inositol 1,4,5-trisphosphate receptors and Ca(2+) release. J Physiol 2016; 594:2985-3004. [PMID: 26751048 DOI: 10.1113/jp270887] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/06/2016] [Indexed: 12/22/2022] Open
Abstract
KEY POINTS During each contraction and haemodynamic disturbance, cardiac myocytes are subjected to fluid shear stress as a result of blood flow and the relative movement of sheets of myocytes. The present study aimed to characterize the shear stress-sensitive membrane current in atrial myocytes using the whole-cell patch clamp technique, combined with pressurized fluid flow, as well as pharmacological and genetic interventions of specific proteins. The data obtained suggest that shear stress indirectly activates the monovalent cation current carried by transient receptor potential melastatin subfamily 4 channels via type 2 inositol 1,4,5-trisphosphate receptor-mediated Ca(2+) release in subsarcolemmal domains of atrial myocytes. Ca(2+) -mediated interactions between these two proteins under shear stress may be an important mechanism by which atrial cells measure mechanical stress and translate it to alter their excitability. ABSTRACT Atrial myocytes are subjected to shear stress during the cardiac cycle under physiological or pathological conditions. The ionic currents regulated by shear stress remain poorly understood. We report the characteristics, molecular identity and activation mechanism of the shear stress-sensitive current (Ishear ) in rat atrial myocytes. A shear stress of ∼16 dyn cm(-2) was applied to single myocytes using a pressurized microflow system, and the current was measured by whole-cell patch clamp. In symmetrical CsCl solutions with minimal concentrations of internal EGTA, Ishear showed an outwardly rectifying current-voltage relationship (reversal at -2 mV). The current was conducted primarily (∼80%) by monovalent cations but not Ca(2+) . It was suppressed by intracellular Ca(2+) buffering at a fixed physiological level, inhibitors of transient receptor potential melastatin subfamily 4 (TRPM4), intracellular introduction of TRPM4 antibodies or knockdown of TRPM4 expression, suggesting that TRPM4 carries most of this current. A notable reduction in Ishear occurred upon inhibition of Ca(2+) release through the ryanodine receptors or inositol 1,4,5-trisphosphate receptors (IP3 R) and upon depletion of sarcoplasmic reticulum Ca(2+) . In type 2 IP3 R (IP3 R2) knockout atrial myocytes, Ishear was 10-20% of that in wild-type myocytes. Immunocytochemistry and proximity ligation assays revealed that TRPM4 and IP3 R2 were expressed at peripheral sites with co-localization, although they are not localized within 40 nm. Peripheral localization of TRPM4 was intact in IP3 R2 knockout cells. The data obtained in the present study suggest that shear stress activates TRPM4 current by triggering Ca(2+) release from the IP3 R2 in the peripheral domains of atrial myocytes.
Collapse
Affiliation(s)
- Min-Jeong Son
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, South Korea
| | - Joon-Chul Kim
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, South Korea
| | - Sung Woo Kim
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, South Korea
| | - Bojjibabu Chidipi
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, South Korea
| | - Jeyaraj Muniyandi
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, South Korea
| | - Thoudam Debraj Singh
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, South Korea
| | - Insuk So
- Department of Physiology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Krishna P Subedi
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, South Korea.,Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA
| | - Sun-Hee Woo
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, Yuseong-gu, Daejeon, South Korea
| |
Collapse
|
25
|
Scheitlin CG, Julian JA, Shanmughapriya S, Madesh M, Tsoukias NM, Alevriadou BR. Endothelial mitochondria regulate the intracellular Ca2+ response to fluid shear stress. Am J Physiol Cell Physiol 2016; 310:C479-90. [PMID: 26739489 DOI: 10.1152/ajpcell.00171.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 01/04/2016] [Indexed: 02/04/2023]
Abstract
Shear stress is known to stimulate an intracellular free calcium concentration ([Ca(2+)]i) response in vascular endothelial cells (ECs). [Ca(2+)]i is a key second messenger for signaling that leads to vasodilation and EC survival. Although it is accepted that the shear-induced [Ca(2+)]i response is, in part, due to Ca(2+) release from the endoplasmic reticulum (ER), the role of mitochondria (second largest Ca(2+) store) is unknown. We hypothesized that the mitochondria play a role in regulating [Ca(2+)]i in sheared ECs. Cultured ECs, loaded with a Ca(2+)-sensitive fluorophore, were exposed to physiological levels of shear stress. Shear stress elicited [Ca(2+)]i transients in a percentage of cells with a fraction of them displaying oscillations. Peak magnitudes, percentage of oscillating ECs, and oscillation frequencies depended on the shear level. [Ca(2+)]i transients/oscillations were present when experiments were conducted in Ca(2+)-free solution (plus lanthanum) but absent when ECs were treated with a phospholipase C inhibitor, suggesting that the ER inositol 1,4,5-trisphosphate receptor is responsible for the [Ca(2+)]i response. Either a mitochondrial uncoupler or an electron transport chain inhibitor, but not a mitochondrial ATP synthase inhibitor, prevented the occurrence of transients and especially inhibited the oscillations. Knockdown of the mitochondrial Ca(2+) uniporter also inhibited the shear-induced [Ca(2+)]i transients/oscillations compared with controls. Hence, EC mitochondria, through Ca(2+) uptake/release, regulate the temporal profile of shear-induced ER Ca(2+) release. [Ca(2+)]i oscillation frequencies detected were within the range for activation of mechanoresponsive kinases and transcription factors, suggesting that dysfunctional EC mitochondria may contribute to cardiovascular disease by deregulating the shear-induced [Ca(2+)]i response.
Collapse
Affiliation(s)
- Christopher G Scheitlin
- Departments of Biomedical Engineering and Internal Medicine, Division of Cardiovascular Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Justin A Julian
- Departments of Biomedical Engineering and Internal Medicine, Division of Cardiovascular Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Santhanam Shanmughapriya
- Department of Medical Genetics and Molecular Biochemistry and Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania; and
| | - Muniswamy Madesh
- Department of Medical Genetics and Molecular Biochemistry and Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania; and
| | - Nikolaos M Tsoukias
- Department of Biomedical Engineering, Florida International University, Miami, Florida
| | - B Rita Alevriadou
- Departments of Biomedical Engineering and Internal Medicine, Division of Cardiovascular Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio;
| |
Collapse
|
26
|
Miragoli M, Sanchez-Alonso JL, Bhargava A, Wright PT, Sikkel M, Schobesberger S, Diakonov I, Novak P, Castaldi A, Cattaneo P, Lyon AR, Lab MJ, Gorelik J. Microtubule-Dependent Mitochondria Alignment Regulates Calcium Release in Response to Nanomechanical Stimulus in Heart Myocytes. Cell Rep 2015; 14:140-151. [PMID: 26725114 PMCID: PMC4983655 DOI: 10.1016/j.celrep.2015.12.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 09/07/2015] [Accepted: 11/23/2015] [Indexed: 12/01/2022] Open
Abstract
Arrhythmogenesis during heart failure is a major clinical problem. Regional electrical gradients produce arrhythmias, and cellular ionic transmembrane gradients are its originators. We investigated whether the nanoscale mechanosensitive properties of cardiomyocytes from failing hearts have a bearing upon the initiation of abnormal electrical activity. Hydrojets through a nanopipette indent specific locations on the sarcolemma and initiate intracellular calcium release in both healthy and heart failure cardiomyocytes, as well as in human failing cardiomyocytes. In healthy cells, calcium is locally confined, whereas in failing cardiomyocytes, calcium propagates. Heart failure progressively stiffens the membrane and displaces sub-sarcolemmal mitochondria. Colchicine in healthy cells mimics the failing condition by stiffening the cells, disrupting microtubules, shifting mitochondria, and causing calcium release. Uncoupling the mitochondrial proton gradient abolished calcium initiation in both failing and colchicine-treated cells. We propose the disruption of microtubule-dependent mitochondrial mechanosensor microdomains as a mechanism for abnormal calcium release in failing heart. Nanomechanical pressure application changes mechanosensitivity in failing heart cells Microtubular network disorganization mediates the change in mechanosensitivity Mitochondria are displaced from their original location and trigger calcium release Uncoupling the mitochondrial proton gradient completely abolishes the phenomena
Collapse
Affiliation(s)
- Michele Miragoli
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK; Humanitas Clinical and Research Center, via Manzoni 56, Rozzano, 20090 Milan, Italy; Center of Excellence for Toxicological Research, INAIL exISPESL, University of Parma, via Gramsci 14, 43126 Parma, Italy.
| | - Jose L Sanchez-Alonso
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Anamika Bhargava
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK; Department of Biotechnology, Indian Institute of Technology Hyderabad, Ordnance Factory Estate, Yeddumailaram, 502205 Telangana, India
| | - Peter T Wright
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Markus Sikkel
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Sophie Schobesberger
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Ivan Diakonov
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK
| | - Pavel Novak
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK; School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London E1 4NS, UK
| | - Alessandra Castaldi
- Humanitas Clinical and Research Center, via Manzoni 56, Rozzano, 20090 Milan, Italy
| | - Paola Cattaneo
- Humanitas Clinical and Research Center, via Manzoni 56, Rozzano, 20090 Milan, Italy
| | - Alexander R Lyon
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK; NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital, London SW36NP, UK
| | - Max J Lab
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK.
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, 4th floor, Imperial Centre for Translational and Experimental Medicine, Hammersmith Campus Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
27
|
Zhang XH, Morad M. Calcium signaling in human stem cell-derived cardiomyocytes: Evidence from normal subjects and CPVT afflicted patients. Cell Calcium 2015; 59:98-107. [PMID: 26725479 DOI: 10.1016/j.ceca.2015.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/10/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
Derivation of cardiomyocyte cell lines from human fibroblasts (induced pluripotent stem cells, iPSCs) has made it possible not only to investigate the electrophysiological and Ca(2+) signaling properties of these cells, but also to determine the altered electrophysiological and Ca(2+)-signaling profiles of such cells lines derived from patients expressing mutation-inducing pathologies. This approach has the potential of generating in vitro human models of cardiovascular diseases where cellular pathology can be investigated in detail and possibly specific pharmacotherapy developed. Although this approach has been applied to a number of mutations in channel proteins that cause arrhythmias, there are only few detailed reports addressing Ca(2+) signaling pathologies beyond measurements of Ca(2+) transients in intact non-voltage clamped cells. Unfortunately, full understanding of Ca(2+) signaling pathologies remains elusive, not only because of the plethora of Ca(2+) signaling proteins defects that cause arrhythmias and cardiomyopathies, but also because detailed functional properties of Ca(2+) signaling proteins are difficult to obtain. Catecholaminergic polymorphic ventricular tachycardia (CPVT1) is a malignant inherited arrhythmogenic disorder predominantly caused by mutations in the cardiac ryanodine receptor (RyR2). Thus far over 150 mutations in RyR2 have been identified that appear to cause this arrhythmia, a number of which have been expressed and studied in transgenic mice or cell-line models. The development of human iPSC-technology makes it possible to create human heart cell-lines carrying these mutations, making detailed identification of Ca(2+) signaling defects and its specific pharmacotherapy possible. In this review we shall first briefly summarize the essential characteristics of the mammalian cardiac Ca(2+) signaling, then compare them to Ca(2+) signaling phenotypes of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CM) and to those of rat neonatal cardiomyocytes, and categorize the possible variance in Ca(2+) signaling defects caused by different CPVT-inducing mutations as expressed in hiPSC-CMs.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- Cardiac Signaling Center of USC, MUSC, & Clemson University, Charleston, SC 29425, USA
| | - Martin Morad
- Cardiac Signaling Center of USC, MUSC, & Clemson University, Charleston, SC 29425, USA.
| |
Collapse
|
28
|
Kim JC, Woo SH. Shear stress induces a longitudinal Ca(2+) wave via autocrine activation of P2Y1 purinergic signalling in rat atrial myocytes. J Physiol 2015; 593:5091-109. [PMID: 26377030 DOI: 10.1113/jp271016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 09/11/2015] [Indexed: 12/25/2022] Open
Abstract
Atrial myocytes are exposed to shear stress during the cardiac cycle and haemodynamic disturbance. In response, they generate a longitudinally propagating global Ca(2+) wave. Here, we investigated the cellular mechanisms underlying the shear stress-mediated Ca(2+) wave, using two-dimensional confocal Ca(2+) imaging combined with a pressurized microflow system in single rat atrial myocytes. Shear stress of ∼16 dyn cm(-2) for 8 s induced ∼1.2 aperiodic longitudinal Ca(2+) waves (∼79 μm s(-1)) with a delay of 0.2-3 s. Pharmacological blockade of ryanodine receptors (RyRs) or inositol 1,4,5-trisphosphate receptors (IP3 Rs) abolished shear stress-induced Ca(2+) wave generation. Furthermore, in atrial myocytes from type 2 IP3R (IP3R2) knock-out mice, shear stress failed to induce longitudinal Ca(2+) waves. The phospholipase C (PLC) inhibitor U73122, but not its inactive analogue U73343, abolished the shear-induced longitudinal Ca(2+) wave. However, pretreating atrial cells with blockers for stretch-activated channels, Na(+)-Ca(2+) exchanger, transient receptor potential melastatin subfamily 4, or nicotinamide adenine dinucleotide phosphate oxidase did not suppress wave generation under shear stress. The P2 purinoceptor inhibitor suramin, and the potent P2Y1 receptor antagonist MRS 2179, both suppressed the Ca(2+) wave, whereas the P2X receptor antagonist, iso-PPADS, did not alter it. Suppression of gap junction hemichannels permeable to ATP or extracellular application of ATP-metabolizing apyrase inhibited the wave. Removal of external Ca(2+) to enhance hemichannel opening facilitated the wave generation. Our data suggest that longitudinally propagating, regenerative Ca(2+) release through RyRs is triggered by P2Y1-PLC-IP3R2 signalling that is activated by gap junction hemichannel-mediated ATP release in atrial myocytes under shear stress.
Collapse
Affiliation(s)
- Joon-Chul Kim
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 305-764, South Korea
| | - Sun-Hee Woo
- Laboratory of Physiology, College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 305-764, South Korea
| |
Collapse
|
29
|
Pechánová O, Varga ZV, Cebová M, Giricz Z, Pacher P, Ferdinandy P. Cardiac NO signalling in the metabolic syndrome. Br J Pharmacol 2015; 172:1415-33. [PMID: 25297560 PMCID: PMC4369254 DOI: 10.1111/bph.12960] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 09/09/2014] [Accepted: 09/28/2014] [Indexed: 02/06/2023] Open
Abstract
It is well documented that metabolic syndrome (i.e. a group of risk factors, such as abdominal obesity, elevated blood pressure, elevated fasting plasma glucose, high serum triglycerides and low cholesterol level in high-density lipoprotein), which raises the risk for heart disease and diabetes, is associated with increased reactive oxygen and nitrogen species (ROS/RNS) generation. ROS/RNS can modulate cardiac NO signalling and trigger various adaptive changes in NOS and antioxidant enzyme expressions/activities. While initially these changes may represent protective mechanisms in metabolic syndrome, later with more prolonged oxidative, nitrosative and nitrative stress, these are often exhausted, eventually favouring myocardial RNS generation and decreased NO bioavailability. The increased oxidative and nitrative stress also impairs the NO-soluble guanylate cyclase (sGC) signalling pathway, limiting the ability of NO to exert its fundamental signalling roles in the heart. Enhanced ROS/RNS generation in the presence of risk factors also facilitates activation of redox-dependent transcriptional factors such as NF-κB, promoting myocardial expression of various pro-inflammatory mediators, and eventually the development of cardiac dysfunction and remodelling. While the dysregulation of NO signalling may interfere with the therapeutic efficacy of conventional drugs used in the management of metabolic syndrome, the modulation of NO signalling may also be responsible for the therapeutic benefits of already proven or recently developed treatment approaches, such as ACE inhibitors, certain β-blockers, and sGC activators. Better understanding of the above-mentioned pathological processes may ultimately lead to more successful therapeutic approaches to overcome metabolic syndrome and its pathological consequences in cardiac NO signalling.
Collapse
Affiliation(s)
- O Pechánová
- Institute of Normal and Pathological Physiology and Centre of Excellence for Regulatory Role of Nitric Oxide in Civilization Diseases, Slovak Academy of SciencesBratislava, Slovak Republic
- Faculty of Natural Sciences, Comenius UniversityBratislava, Slovak Republic
| | - Z V Varga
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis UniversityBudapest, Hungary
| | - M Cebová
- Institute of Normal and Pathological Physiology and Centre of Excellence for Regulatory Role of Nitric Oxide in Civilization Diseases, Slovak Academy of SciencesBratislava, Slovak Republic
| | - Z Giricz
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis UniversityBudapest, Hungary
| | - P Pacher
- Laboratory of Physiological Studies, National Institutes of Health/NIAAABethesda, MD, USA
| | - P Ferdinandy
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis UniversityBudapest, Hungary
- Pharmahungary GroupSzeged, Hungary
| |
Collapse
|
30
|
Hsiao YF, Pan HJ, Tung YC, Chen CC, Lee CH. Effects of hydraulic pressure on cardiomyoblasts in a microfluidic device. BIOMICROFLUIDICS 2015; 9:024111. [PMID: 25945137 PMCID: PMC4393411 DOI: 10.1063/1.4917080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/27/2015] [Indexed: 05/05/2023]
Abstract
We employed a microfluidic device to study the effects of hydraulic pressure on cardiomyoblast H9c2. The 170 mm Hg pressure increased the cellular area and the expression of atrial natriuretic peptide. With the same device, we demonstrated that the effects of hydraulic pressure on the cardiomyoblast could be reduced by the inhibitor of focal adhesion kinase. This mechanical-chemical antagonism could lead to a potential therapeutic strategy of hypertension-induced cardiac hypertrophy.
Collapse
Affiliation(s)
| | - Huei-Jyuan Pan
- Research Center for Applied Sciences , Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Chung Tung
- Research Center for Applied Sciences , Academia Sinica, Taipei 11529, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences , Academia Sinica, Taipei 11529, Taiwan
| | | |
Collapse
|
31
|
Living cardiac tissue slices: an organotypic pseudo two-dimensional model for cardiac biophysics research. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:314-27. [PMID: 25124067 DOI: 10.1016/j.pbiomolbio.2014.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/02/2014] [Indexed: 11/24/2022]
Abstract
Living cardiac tissue slices, a pseudo two-dimensional (2D) preparation, have received less attention than isolated single cells, cell cultures, or Langendorff-perfused hearts in cardiac biophysics research. This is, in part, due to difficulties associated with sectioning cardiac tissue to obtain live slices. With moderate complexity, native cell-types, and well-preserved cell-cell electrical and mechanical interconnections, cardiac tissue slices have several advantages for studying cardiac electrophysiology. The trans-membrane potential (Vm) has, thus far, mainly been explored using multi-electrode arrays. Here, we combine tissue slices with optical mapping to monitor Vm and intracellular Ca(2+) concentration ([Ca(2+)]i). This combination opens up the possibility of studying the effects of experimental interventions upon action potential (AP) and calcium transient (CaT) dynamics in 2D, and with relatively high spatio-temporal resolution. As an intervention, we conducted proof-of-principle application of stretch. Mechanical stimulation of cardiac preparations is well-established for membrane patches, single cells and whole heart preparations. For cardiac tissue slices, it is possible to apply stretch perpendicular or parallel to the dominant orientation of cells, while keeping the preparation in a constant focal plane for fluorescent imaging of in-slice functional dynamics. Slice-to-slice comparison furthermore allows one to assess transmural differences in ventricular tissue responses to mechanical challenges. We developed and tested application of axial stretch to cardiac tissue slices, using a manually-controlled stretching device, and recorded Vm and [Ca(2+)]i by optical mapping before, during, and after application of stretch. Living cardiac tissue slices, exposed to axial stretch, show an initial shortening in both AP and CaT duration upon stretch application, followed in most cases by a gradual prolongation of AP and CaT duration during stretch maintained for up to 50 min. After release of sustained stretch, AP duration (APD) and CaT duration reverted to shorter values. Living cardiac tissue slices are a promising experimental model for the study of cardiac mechano-electric interactions. The methodology described here can be refined to achieve more accurate control over stretch amplitude and timing (e.g. using a computer-controlled motorised stage, or by synchronising electrical and mechanical events) and through monitoring of regional tissue deformation (e.g. by adding motion tracking).
Collapse
|
32
|
Cardiac mechano-electric coupling research: Fifty years of progress and scientific innovation. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:71-5. [DOI: 10.1016/j.pbiomolbio.2014.06.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/19/2014] [Indexed: 12/22/2022]
|
33
|
Fernández-Sada E, Silva-Platas C, Villegas CA, Rivero SL, Willis BC, García N, Garza JR, Oropeza-Almazán Y, Valverde CA, Mazzocchi G, Zazueta C, Torre-Amione G, García-Rivas G. Cardiac responses to β-adrenoceptor stimulation is partly dependent on mitochondrial calcium uniporter activity. Br J Pharmacol 2014; 171:4207-21. [PMID: 24628066 DOI: 10.1111/bph.12684] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 02/06/2014] [Accepted: 03/01/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Despite the importance of mitochondrial Ca(2+) to metabolic regulation and cell physiology, little is known about the mechanisms that regulate Ca(2+) entry into the mitochondria. Accordingly, we established a system to determine the role of the mitochondrial Ca(2+) uniporter in an isolated heart model, at baseline and during increased workload following β-adrenoceptor stimulation. EXPERIMENTAL APPROACH Cardiac contractility, oxygen consumption and intracellular Ca(2+) transients were measured in ex vivo perfused murine hearts. Ru360 and spermine were used to modify mitochondrial Ca(2+) uniporter activity. Changes in mitochondrial Ca(2+) content and energetic phosphate metabolite levels were determined. KEY RESULTS The addition of Ru360 , a selective inhibitor of the mitochondrial Ca(2+) uniporter, induced progressively and sustained negative inotropic effects that were dose-dependent with an EC50 of 7 μM. Treatment with spermine, a uniporter agonist, showed a positive inotropic effect that was blocked by Ru360 . Inotropic stimulation with isoprenaline elevated oxygen consumption (2.7-fold), Ca(2+) -dependent activation of pyruvate dehydrogenase (5-fold) and mitochondrial Ca(2+) content (2.5-fold). However, in Ru360 -treated hearts, this parameter was attenuated. In addition, β-adrenoceptor stimulation in the presence of Ru360 did not affect intracellular Ca(2+) handling, PKA or Ca(2+) /calmodulin-dependent PK signalling. CONCLUSIONS AND IMPLICATIONS Inhibition of the mitochondrial Ca(2+) uniporter decreases β-adrenoceptor response, uncoupling between workload and production of energetic metabolites. Our results support the hypothesis that the coupling of workload and energy supply is partly dependent on mitochondrial Ca(2+) uniporter activity.
Collapse
Affiliation(s)
- E Fernández-Sada
- Cátedra de Cardiología y Medicina Vascular, Escuela de Medicina, Tecnológico de Monterrey, Monterrey, Nuevo León, México
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Diversity of mitochondrial Ca²⁺ signaling in rat neonatal cardiomyocytes: evidence from a genetically directed Ca²⁺ probe, mitycam-E31Q. Cell Calcium 2014; 56:133-46. [PMID: 24994483 DOI: 10.1016/j.ceca.2014.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 06/03/2014] [Accepted: 06/07/2014] [Indexed: 11/24/2022]
Abstract
I(Ca)-gated Ca(2+) release (CICR) from the cardiac SR is the main mechanism mediating the rise of cytosolic Ca(2+), but the extent to which mitochondria contribute to the overall Ca(2+) signaling remains controversial. To examine the possible role of mitochondria in Ca(2+) signaling, we developed a low affinity mitochondrial Ca(2+) probe, mitycam-E31Q (300-500 MOI, 48-72h) and used it in conjunction with Fura-2AM to obtain simultaneous TIRF images of mitochondrial and cytosolic Ca(2+) in cultured neonatal rat cardiomyocytes. Mitycam-E31Q staining of adult feline cardiomyocytes showed the typical mitochondrial longitudinal fluorescent bandings similar to that of TMRE staining, while neonatal rat cardiomyocytes had a disorganized tubular or punctuate appearance. Caffeine puffs produced rapid increases in cytosolic Ca(2+) while simultaneously measured global mitycam-E31Q signals decreased more slowly (increased mitochondrial Ca(2+)) before decaying to baseline levels. Similar, but oscillating mitycam-E31Q signals were seen in spontaneously pacing cells. Withdrawal of Na(+) increased global cytosolic and mitochondrial Ca(2+) signals in one population of mitochondria, but unexpectedly decreased it (release of Ca(2+)) in another mitochondrial population. Such mitochondrial Ca(2+) release signals were seen not only during long lasting Na(+) withdrawal, but also when Ca(2+) loaded cells were exposed to caffeine-puffs, and during spontaneous rhythmic beating. Thus, mitochondrial Ca(2+) transients appear to activate with a delay following the cytosolic rise of Ca(2+) and show diversity in subpopulations of mitochondria that could contribute to the plasticity of mitochondrial Ca(2+) signaling.
Collapse
|
35
|
Hatano A, Okada JI, Washio T, Hisada T, Sugiura S. Mitochondrial colocalization with Ca2+ release sites is crucial to cardiac metabolism. Biophys J 2013; 104:496-504. [PMID: 23442872 DOI: 10.1016/j.bpj.2012.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 11/22/2012] [Accepted: 12/04/2012] [Indexed: 10/27/2022] Open
Abstract
In cardiomyocyte subcellular structures, colocalization of mitochondria with Ca2+ release sites is implicated in regulation of cardiac energetics by facilitating Ca2+ influx into mitochondria to modulate the tricarboxylic acid (TCA) cycle. However, current experimental techniques limit detailed examination of this regulatory mechanism. Earlier, we developed a three-dimensional (3D) finite-element cardiomyocyte model featuring a subcellular structure that integrates excitation-contraction coupling and energy metabolism. Here, using this model, we examined the influence of distance between mitochondria and Ca2+ release sites by comparing a normal (50-nm) distance model and a large (200-nm) distance model (LD). The influence of distance was minimal under a low pacing rate (0.25 Hz), but under a higher pacing rate (2 Hz), lower levels of mitochondrial Ca2+ and NADH, elevated phosphate, and suppressed force generation became apparent in the LD model. Such differences became greater when functional impairments (reduced TCA cycle activity, uncoupling effect, and failing excitation-contraction coupling) were additionally imposed. We concluded that juxtaposition of the mitochondria and the Ca2+ release sites is crucial for rapid signal transmission to maintain cardiac-energy balance. The idealized 3D model of cardiac excitation-contraction and metabolism is a powerful tool to study cardiac energetics.
Collapse
Affiliation(s)
- Asuka Hatano
- Department of Frontier Science, The University of Tokyo, Kashiwa, Chiba, Japan.
| | | | | | | | | |
Collapse
|
36
|
Arnáiz-Cot JJ, Damon BJ, Zhang XH, Cleemann L, Yamaguchi N, Meissner G, Morad M. Cardiac calcium signalling pathologies associated with defective calmodulin regulation of type 2 ryanodine receptor. J Physiol 2013; 591:4287-99. [PMID: 23836685 DOI: 10.1113/jphysiol.2013.256123] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cardiac ryanodine receptor (RyR2) is a homotetramer of 560 kDa polypeptides regulated by calmodulin (CaM), which decreases its open probability at diastolic and systolic Ca(2+) concentrations. Point mutations in the CaM-binding domain of RyR2 (W3587A/L3591D/F3603A, RyR2(ADA)) in mice result in severe cardiac hypertrophy, poor left ventricle contraction and death by postnatal day 16, suggesting that CaM inhibition of RyR2 is required for normal cardiac function. Here, we report on Ca(2+) signalling properties of enzymatically isolated, Fluo-4 dialysed whole cell clamped cardiac myocytes from 10-15-day-old wild-type (WT) and homozygous Ryr2(ADA/ADA) mice. Spontaneously occurring Ca(2+) spark frequency, measured at -80 mV, was 14-fold lower in mutant compared to WT myocytes. ICa, though significantly smaller in mutant myocytes, triggered Ca(2+) transients that were of comparable size to those of WT myocytes, but with slower activation and decay kinetics. Caffeine-triggered Ca(2+) transients were about three times larger in mutant myocytes, generating three- to four-fold bigger Na(+)-Ca(2+) exchanger NCX currents (INCX). Mutant myocytes often exhibited Ca(2+) transients of variable size and duration that were accompanied by similarly alternating and slowly activating INCX. The data suggest that RyR2(ADA) mutation produces significant reduction in ICa density and ICa-triggered Ca(2+) release gain, longer but infrequently occurring Ca(2+) sparks, larger sarcoplasmic reticulum Ca(2+) loads, and spontaneous Ca(2+) releases accompanied by activation of large and potentially arrhythmogenic inward INCX.
Collapse
Affiliation(s)
- Juan José Arnáiz-Cot
- M. Morad: Cardiac Signaling Center, 173 Ashley Ave, Bioengineering Building, Room 306, Charleston, SC 29403, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Zhang XH, Haviland S, Wei H, Sarić T, Fatima A, Hescheler J, Cleemann L, Morad M. Ca2+ signaling in human induced pluripotent stem cell-derived cardiomyocytes (iPS-CM) from normal and catecholaminergic polymorphic ventricular tachycardia (CPVT)-afflicted subjects. Cell Calcium 2013; 54:57-70. [PMID: 23684427 DOI: 10.1016/j.ceca.2013.04.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/10/2013] [Accepted: 04/12/2013] [Indexed: 12/16/2022]
Abstract
Derivation of cardiomyocytes from induced pluripotent stem cells (iPS-CMs) allowed us to probe the Ca(2+)-signaling parameters of human iPS-CMs from healthy- and catecholaminergic polymorphic ventricular tachycardia (CPVT1)-afflicted individuals carrying a novel point mutation p.F2483I in ryanodine receptors (RyR2). iPS-CMs were dissociated on day 30-40 of differentiation and patch-clamped within 3-6 days. Calcium currents (ICa) averaged ∼8pA/pF in control and mutant iPS-CMs. ICa-induced Ca(2+)-transients in control and mutant cells had bell-shaped voltage-dependence similar to that of ICa, consistent with Ca(2+)-induced Ca(2+)-release (CICR) mechanism. The ratio of ICa-activated to caffeine-triggered Ca(2+)-transients was ∼0.3 in both cell types. Caffeine-induced Ca(2+)-transients generated significantly smaller Na(+)-Ca(2+) exchanger current (INCX) in mutant cells, reflecting their smaller Ca(2+)-stores. The gain of CICR was voltage-dependent as in adult cardiomyocytes. Adrenergic agonists enhanced ICa, but differentially altered the CICR gain, diastolic Ca(2+), and Ca(2+)-sparks in mutant cells. The mutant cells, when Ca(2+)-overloaded, showed longer and wandering Ca(2+)-sparks that activated adjoining release sites, had larger CICR gain at -30mV yet smaller Ca(2+)-stores. We conclude that control and mutant iPS-CMs express the adult cardiomyocyte Ca(2+)-signaling phenotype. RyR2 F2483I mutant myocytes have aberrant unitary Ca(2+)-signaling, smaller Ca(2+)-stores, higher CICR gains, and sensitized adrenergic regulation, consistent with functionally altered Ca(2+)-release profile of CPVT syndrome.
Collapse
Affiliation(s)
- X-H Zhang
- Cardiac Signaling Center of USC, MUSC, & Clemson University, Charleston, SC 29403, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhang H, Gomez AM, Wang X, Yan Y, Zheng M, Cheng H. ROS regulation of microdomain Ca(2+) signalling at the dyads. Cardiovasc Res 2013; 98:248-58. [PMID: 23455546 DOI: 10.1093/cvr/cvt050] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Reactive oxygen species (ROS) are emerging as centre-stage players in cardiac functional regulation. ROS and Ca(2+) signals converge at dyads, the structural and functional units of cardiac excitation-contraction coupling. These two prominent signalling systems are intertwined with ROS modulation of the entire Ca(2+)-signalling network, and vice versa. While constitutively generated homoeostatic ROS are important in setting the redox potential of the intracellular milieu, dynamic signalling ROS shape microdomain and global Ca(2+) signals on both the beat-to-beat and greater time scales. However, ROS effects are complex and subtle, characterized by multiphasic and bidirectional Ca(2+) responses; and sustained oxidative stress may lead to compromised contractility and arrhythmogenicity. These new understandings should be leveraged to harness ROS for their beneficial roles while avoiding deleterious effects in the heart.
Collapse
Affiliation(s)
- Huiliang Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | | | | | | | | | | |
Collapse
|
39
|
Rosa AO, Yamaguchi N, Morad M. Mechanical regulation of native and the recombinant calcium channel. Cell Calcium 2013; 53:264-74. [PMID: 23357406 DOI: 10.1016/j.ceca.2012.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 12/25/2012] [Indexed: 11/30/2022]
Abstract
L-type calcium channels are modulated by a host of mechanisms that include voltage, calcium ions (Ca(2+) dependent inactivation and facilitation), cytosolic proteins (CAM, CAMKII, PKA, PKC, etc.), and oxygen radicals. Here we describe yet another Ca(2+) channel regulatory mechanism that is induced by pressure-flow (PF) forces of ∼25dyn/cm(2) producing 35-60% inhibition of channel current. Only brief periods (300ms) of such PF pulses were required to suppress reversibly the current. Recombinant Ca(2+) channels (α1c77/β2a/α2δ and α1c77/β1/α2δ), expressed in HEK293 cells, were similarly suppressed by PF pulses. To examine whether Ca(2+) released by PF pulses triggered from different sub-cellular compartments (SR, ER, mitochondria) underlies the inhibitory effect of PF on the channel current, pharmacological agents and ionic substitutions were employed to probe this possibility. No significant difference in effectiveness of PF pulses to suppress ICa or IBa (used to inhibit CICR) was found between control cells and those exposed to U73122 and 2-APB (PLC and IP3R pathway modulators), thapsigargin and BAPTA (SERCA2a modulator), dinitrophenol, FCCP and Ru360 (mitochondrial inhibitors), l-NAME (NOS inhibitor signaling), cAMP and Pertussis toxin (Gi protein modulator). We concluded that the rapid and reversible modulation of the Ca(2+) channel by PF pulses is independent of intracellular release of Ca(2+) and Ca(2+) dependent inactivation of the channel and may represent direct mechanical regulatory effect on the channel protein in addition to previously reported Ca(2+)-release or entry dependent mechanism.
Collapse
Affiliation(s)
- Angelo O Rosa
- Cardiac Signaling Center, University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
40
|
Dedkova EN, Blatter LA. Calcium signaling in cardiac mitochondria. J Mol Cell Cardiol 2013; 58:125-33. [PMID: 23306007 DOI: 10.1016/j.yjmcc.2012.12.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 12/01/2012] [Accepted: 12/28/2012] [Indexed: 01/02/2023]
Abstract
Mitochondrial Ca signaling contributes to the regulation of cellular energy metabolism, and mitochondria participate in cardiac excitation-contraction coupling (ECC) through their ability to store Ca, shape the cytosolic Ca signals and generate ATP required for contraction. The mitochondrial inner membrane is equipped with an elaborate system of channels and transporters for Ca uptake and extrusion that allows for the decoding of cytosolic Ca signals, and the storage of Ca in the mitochondrial matrix compartment. Controversy, however remains whether the fast cytosolic Ca transients underlying ECC in the beating heart are transmitted rapidly into the matrix compartment or slowly integrated by the mitochondrial Ca transport machinery. This review summarizes established and novel findings on cardiac mitochondrial Ca transport and buffering, and discusses the evidence either supporting or arguing against the idea that Ca can be taken up rapidly by mitochondria during ECC.
Collapse
Affiliation(s)
- Elena N Dedkova
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, IL 60612, USA
| | | |
Collapse
|
41
|
|
42
|
Kohl P, Bollensdorff C, Morad M. Progress in Biophysics and Molecular Biology of the Beating Heart. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:151-3. [DOI: 10.1016/j.pbiomolbio.2012.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 08/09/2012] [Indexed: 12/14/2022]
|
43
|
Quinn TA, Kohl P. Mechano-sensitivity of cardiac pacemaker function: pathophysiological relevance, experimental implications, and conceptual integration with other mechanisms of rhythmicity. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:257-68. [PMID: 23046620 PMCID: PMC3526794 DOI: 10.1016/j.pbiomolbio.2012.08.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 08/09/2012] [Indexed: 12/11/2022]
Abstract
Cardiac pacemaker cells exhibit spontaneous, rhythmic electrical excitation, termed automaticity. This automatic initiation of action potentials requires spontaneous diastolic depolarisation, whose rate determines normal rhythm generation in the heart. Pacemaker mechanisms have been split recently into: (i) cyclic changes in trans-sarcolemmal ion flows (termed the ‘membrane-clock’), and (ii) rhythmic intracellular calcium cycling (the ‘calcium-clock’). These two ‘clocks’ undoubtedly interact, as trans-sarcolemmal currents involved in pacemaking include calcium-carrying mechanisms, while intracellular calcium cycling requires trans-sarcolemmal ion flux as the mechanism by which it affects membrane potential. The split into separate ‘clocks’ is, therefore, somewhat arbitrary. Nonetheless, the ‘clock’ metaphor has been conceptually stimulating, in particular since there is evidence to support the view that either ‘clock’ could be sufficient in principle to set the rate of pacemaker activation. Of course, the same has also been shown for sub-sets of ‘membrane-clock’ ion currents, illustrating the redundancy of mechanisms involved in maintaining such basic functionality as the heartbeat, a theme that is common for vital physiological systems. Following the conceptual path of identifying individual groups of sub-mechanisms, it is important to remember that the heart is able to adapt pacemaker rate to changes in haemodynamic load, even after isolation or transplantation, and on a beat-by-beat basis. Neither the ‘membrane-’ nor the ‘calcium-clock’ do, as such, inherently account for this rapid adaptation to circulatory demand (cellular Ca2+ balance changes over multiple beats, while variation of sarcolemmal ion channel presence takes even longer). This suggests that a third set of mechanisms must be involved in setting the pace. These mechanisms are characterised by their sensitivity to the cyclically changing mechanical environment, and – in analogy to the above terminology – this might be considered a ‘mechanics-clock’. In this review, we discuss possible roles of mechano-sensitive mechanisms for the entrainment of membrane current dynamics and calcium-handling. This can occur directly via stretch-activation of mechano-sensitive ion channels in the sarcolemma and/or in intracellular membrane compartments, as well as by modulation of ‘standard’ components of the ‘membrane-’ or ‘calcium-clock’. Together, these mechanisms allow rapid adaptation to changes in haemodynamic load, on a beat-by-beat basis. Additional relevance arises from the fact that mechano-sensitivity of pacemaking may help to explain pacemaker dysfunction in mechanically over- or under-loaded tissue. As the combined contributions of the various underlying oscillatory mechanisms are integrated at the pacemaker cell level into a single output – a train of pacemaker action potentials – we will not adhere to a metaphor that implies separate time-keeping units (‘clocks’), and rather focus on cardiac pacemaking as the result of interactions of a set of coupled oscillators, whose individual contributions vary depending on the pathophysiological context. We conclude by considering the utility and limitations of viewing the pacemaker as a coupled system of voltage-, calcium-, and mechanics-modulated oscillators that, by integrating a multitude of inputs, offers the high level of functional redundancy that is vitally important for cardiac automaticity.
Collapse
Affiliation(s)
- T Alexander Quinn
- National Heart and Lung Institute, Imperial College London, London, UK.
| | | |
Collapse
|
44
|
Hatano A, Okada JI, Washio T, Hisada T, Sugiura S. A three-dimensional simulation model of cardiomyocyte integrating excitation-contraction coupling and metabolism. Biophys J 2012; 101:2601-10. [PMID: 22261047 DOI: 10.1016/j.bpj.2011.10.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 09/15/2011] [Accepted: 10/11/2011] [Indexed: 11/30/2022] Open
Abstract
Recent studies have revealed that Ca(2+) not only regulates the contraction of cardiomyocytes, but can also function as a signaling agent to stimulate ATP production by the mitochondria. However, the spatiotemporal resolution of current experimental techniques limits our investigative capacity to understand this phenomenon. Here, we created a detailed three-dimensional (3D) cardiomyocyte model to study the subcellular regulatory mechanisms of myocardial energetics. The 3D cardiomyocyte model was based on the finite-element method, with detailed subcellular structures reproduced, and it included all elementary processes involved in cardiomyocyte electrophysiology, contraction, and ATP metabolism localized to specific loci. The simulation results were found to be reproducible and consistent with experimental data regarding the spatiotemporal pattern of cytosolic, intrasarcoplasmic-reticulum, and mitochondrial changes in Ca(2+); as well as changes in metabolite levels. Detailed analysis suggested that although the observed large cytosolic Ca(2+) gradient facilitated uptake by the mitochondrial Ca(2+) uniporter to produce cyclic changes in mitochondrial Ca(2+) near the Z-line region, the average mitochondrial Ca(2+) changes slowly. We also confirmed the importance of the creatine phosphate shuttle in cardiac energy regulation. In summary, our 3D model provides a powerful tool for the study of cardiac function by overcoming some of the spatiotemporal limitations of current experimental approaches.
Collapse
Affiliation(s)
- Asuka Hatano
- Department of Frontier Science, The University of Tokyo, Kashiwa, Chiba, Japan.
| | | | | | | | | |
Collapse
|
45
|
Johnson JD, Bround MJ, White SA, Luciani DS. Nanospaces between endoplasmic reticulum and mitochondria as control centres of pancreatic β-cell metabolism and survival. PROTOPLASMA 2012; 249 Suppl 1:S49-S58. [PMID: 22105567 DOI: 10.1007/s00709-011-0349-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 11/07/2011] [Indexed: 05/31/2023]
Abstract
Nanometre-scale spaces between organelles represent focused nodes for signal transduction and the control of cellular decisions. The endoplasmic reticulum (ER) and the mitochondria form dynamic quasi-synaptic interaction nanodomains in all cell types examined, but the functional role of these junctions in cellular metabolism and cell survival remains to be fully understood. In this paper, we review recent evidence that ER Ca(2+) channels, such as the RyR and IP(3)R, can signal specifically across this nanodomain to the adjacent mitochondria to pace basal metabolism, with focus on the pancreatic β-cell. Blocking these signals in the basal state leads to a form of programmed cell death associated with reduced ATP and the induction of calpain-10 and hypoxia-inducible factors. On the other hand, the hyperactivity of this signalling domain plays a deleterious role during classical forms of apoptosis. Thus, the nanospace between ER and mitochondria represents a critical rheostat controlling both metabolism and programmed cell death. Many aspects of the mechanisms underlying this control system remain to be uncovered, and new nanotechnologies are required understand these domains at a molecular level.
Collapse
Affiliation(s)
- James D Johnson
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada.
| | | | | | | |
Collapse
|
46
|
Chan DD, Van Dyke WS, Bahls M, Connell SD, Critser P, Kelleher JE, Kramer MA, Pearce SM, Sharma S, Neu CP. Mechanostasis in apoptosis and medicine. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2011; 106:517-24. [PMID: 21846479 DOI: 10.1016/j.pbiomolbio.2011.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 08/02/2011] [Indexed: 10/17/2022]
Abstract
Mechanostasis describes a complex and dynamic process where cells maintain equilibrium in response to mechanical forces. Normal physiological loading modes and magnitudes contribute to cell proliferation, tissue growth, differentiation and development. However, cell responses to abnormal forces include compensatory apoptotic mechanisms that may contribute to the development of tissue disease and pathological conditions. Mechanotransduction mechanisms tightly regulate the cell response through discrete signaling pathways. Here, we provide an overview of links between pro- and anti-apoptotic signaling and mechanotransduction signaling pathways, and identify potential clinical applications for treatments of disease by exploiting mechanically-linked apoptotic pathways.
Collapse
Affiliation(s)
- D D Chan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Movafagh S, Cleemann L, Morad M. Regulation of cardiac Ca(2+) channel by extracellular Na(+). Cell Calcium 2011; 49:162-73. [PMID: 21349579 DOI: 10.1016/j.ceca.2011.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 01/20/2011] [Indexed: 11/25/2022]
Abstract
Hyponatremia is a predictor of poor cardiovascular outcomes during acute myocardial infarction and in the setting of preexisting heart failure [1]. There are no definitive mechanisms as to how hyponatremia suppresses cardiac function. In this report we provide evidence for direct down-regulation of Ca(2+) channel current in response to low serum Na(+). In voltage-clamped rat ventricular myocytes or HEK 293 cells expressing the L-type Ca(2+) channel, a 15mM drop in extracellular Na(+) suppressed the Ca(2+) current by ∼15%; with maximal suppression of ∼30% when Na(+) levels were reduced to 100mM or less. The suppressive effects of low Na(+) on I(Ca), in part, depended on the substituting monovalent species (Li(+), Cs(+), TEA(+)), but were independent of phosphorylation state of the channel and possible influx of Ca(2+) on Na(+)/Ca(2+) exchanger. Acidification sensitized the Ca(2+) channel current to Na(+) withdrawal. Collectively our data suggest that Na(+) and H(+) may interact with regulatory site(s) at the outer recesses of the Ca(2+) channel pore thereby directly modulating the electro-diffusion of the permeating divalents (Ca(2+), Ba(2+)).
Collapse
Affiliation(s)
- Shahrzad Movafagh
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | |
Collapse
|
48
|
Neumann JT, Diaz-Sylvester PL, Fleischer S, Copello JA. CGP-37157 inhibits the sarcoplasmic reticulum Ca²+ ATPase and activates ryanodine receptor channels in striated muscle. Mol Pharmacol 2010; 79:141-7. [PMID: 20923851 DOI: 10.1124/mol.110.067165] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
7-Chloro-5-(2-chlorophenyl)-1,5-dihydro-4,1-benzothiazepin-2(3H)-one [CGP-37157 (CGP)], a benzothiazepine derivative of clonazepam, is commonly used as a blocker of the mitochondrial Na+/Ca²+ exchanger. However, evidence suggests that CGP could also affect other targets, such as L-type Ca²+ channels and plasmalemma Na+/Ca²+ exchanger. Here, we tested the possibility of a direct modulation of ryanodine receptor channels (RyRs) and/or sarco/endoplasmic reticulum Ca²+-stimulated ATPase (SERCA) by CGP. In the presence of ruthenium red (inhibitor of RyRs), CGP decreased SERCA-mediated Ca²+ uptake of cardiac and skeletal sarcoplasmic reticulum (SR) microsomes (IC₅₀ values of 6.6 and 9.9 μM, respectively). The CGP effects on SERCA activity correlated with a decreased V(max) of ATPase activity of SERCA-enriched skeletal SR fractions. CGP (≥ 5 μM) also increased RyR-mediated Ca²+ leak from skeletal SR microsomes. Planar bilayer studies confirmed that both cardiac and skeletal RyRs are directly activated by CGP (EC(50) values of 9.4 and 12.0 μM, respectively). In summary, we found that CGP inhibits SERCA and activates RyR channels. Hence, the action of CGP on cellular Ca²+ homeostasis reported in the literature of cardiac, skeletal muscle, and other nonmuscle systems requires further analysis to take into account the contribution of all CGP-sensitive Ca²+ transporters.
Collapse
Affiliation(s)
- Jake T Neumann
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | | | | | | |
Collapse
|
49
|
Abstract
Despite a high prevalence of sudden cardiac death throughout the world, the mechanisms that lead to ventricular arrhythmias are not fully understood. Over the last 20 years, a growing body of evidence indicates that cardiac mitochondria are involved in the genesis of arrhythmia. In this review, we have attempted to describe the role that mitochondria play in altering the heart's electrical function by introducing heterogeneity into the cardiac action potential. Specifically, we have focused on how the energetic status of the mitochondrial network can alter sarcolemmal potassium fluxes through ATP-sensitive potassium channels, creating a 'metabolic sink' for depolarizing wave-fronts and introducing conditions that favour catastrophic arrhythmia. Mechanisms by which mitochondria depolarize under conditions of oxidative stress are characterized, and the contributions of several mitochondrial ion channels to mitochondrial depolarization are presented. The inner membrane anion channel in particular opens upstream of other inner membrane channels during metabolic stress, and may be an effective target to prevent the metabolic oscillations that create action potential lability. Finally, we discuss therapeutic strategies that prevent arrhythmias by preserving mitochondrial membrane potential in the face of oxidative stress, supporting the notion that treatments aimed at cardiac mitochondria have significant potential in attenuating electrical dysfunction in the heart.
Collapse
Affiliation(s)
- David A Brown
- Department of Physiology, Brody School of Medicine and the East Carolina Heart Institute, East Carolina University, Room 6N-98, 600 Moye Blvd, Greenville, NC 27834, USA.
| | | |
Collapse
|
50
|
Pozzan T, Rudolf R. Measurements of mitochondrial calcium in vivo. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:1317-23. [DOI: 10.1016/j.bbabio.2008.11.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Revised: 11/20/2008] [Accepted: 11/21/2008] [Indexed: 12/21/2022]
|