1
|
Schreiber J, Rotard L, Tourneur Y, Lafoux A, Berthier C, Allard B, Huchet C, Jacquemond V. Reduced voltage-activated Ca2+ release flux in muscle fibers from a rat model of Duchenne dystrophy. J Gen Physiol 2025; 157:e202413588. [PMID: 39718509 DOI: 10.1085/jgp.202413588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/14/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024] Open
Abstract
The potential pathogenic role of disturbed Ca2+ homeostasis in Duchenne muscular dystrophy (DMD) remains a complex, unsettled issue. We used muscle fibers isolated from 3-mo-old DMDmdx rats to further investigate the case. Most DMDmdx fibers exhibited no sign of trophic or morphology distinction as compared with WT fibers and mitochondria and t-tubule membrane networks also showed no stringent discrepancy. Under voltage clamp, values for holding current were similar in the two groups, whereas values for capacitance were larger in DMDmdx fibers, suggestive of enhanced amount of t-tubule membrane. The Ca2+ current density across the channel carried by the EC coupling voltage sensor (CaV1.1) was unchanged. The maximum rate of voltage-activated sarcoplasmic reticulum (SR) Ca2+ release was reduced by 25% in the DMDmdx fibers, with no change in voltage dependency. Imaging resting Ca2+ revealed rare spontaneous local SR Ca2+ release events with no sign of elevated activity in DMDmdx fibers. Under current clamp, DMDmdx fibers generated similar trains of action potentials as WT fibers. Results suggest that reduced peak amplitude of SR Ca2+ release is an inherent feature of this DMD model, likely contributing to muscle weakness. This occurs despite a preserved amount of releasable Ca2+ and with no change in excitability, CaV1.1 channel activity, and SR Ca2+ release at rest. Although we cannot exclude that fibers from the 3-mo-old animals do not yet display a fully developed disease phenotype, results provide limited support for pathomechanistic concepts frequently associated with DMD such as membrane fragility, excessive Ca2+ entry, or enhanced SR Ca2+ leak.
Collapse
Affiliation(s)
- Jonathan Schreiber
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Ludivine Rotard
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Yves Tourneur
- UFPE Department Nutrição, Cidade Universitária, Recife, Brazil
| | - Aude Lafoux
- Therassay Platform, CAPACITES, Nantes Université , Nantes, France
| | - Christine Berthier
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Bruno Allard
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Corinne Huchet
- Therassay Platform, CAPACITES, Nantes Université , Nantes, France
- Nantes Gene Therapy Laboratory, Nantes Université, INSERM UMR TARGET 1089, Nantes, France
| | - Vincent Jacquemond
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| |
Collapse
|
2
|
Pelizzari S, Heiss MC, Fernández-Quintero ML, El Ghaleb Y, Liedl KR, Tuluc P, Campiglio M, Flucher BE. Ca V1.1 voltage-sensing domain III exclusively controls skeletal muscle excitation-contraction coupling. Nat Commun 2024; 15:7440. [PMID: 39198449 PMCID: PMC11358481 DOI: 10.1038/s41467-024-51809-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Skeletal muscle contractions are initiated by action potentials, which are sensed by the voltage-gated calcium channel (CaV1.1) and are conformationally coupled to calcium release from intracellular stores. Notably, CaV1.1 contains four separate voltage-sensing domains (VSDs), which activate channel gating and excitation-contraction (EC-) coupling at different voltages and with distinct kinetics. Here we show that a single VSD of CaV1.1 controls skeletal muscle EC-coupling. Whereas mutations in VSDs I, II and IV affect the current properties but not EC-coupling, only mutations in VSD III alter the voltage-dependence of depolarization-induced calcium release. Molecular dynamics simulations reveal comprehensive, non-canonical state transitions of VSD III in response to membrane depolarization. Identifying the voltage sensor that activates EC-coupling and detecting its unique conformational changes opens the door to unraveling the downstream events linking VSD III motion to the opening of the calcium release channel, and thus resolving the signal transduction mechanism of skeletal muscle EC-coupling.
Collapse
Affiliation(s)
- Simone Pelizzari
- Institute of Physiology, Department of Physiology and Medical Biophysics, Medical University Innsbruck, 6020, Innsbruck, Austria
| | - Martin C Heiss
- Institute of Physiology, Department of Physiology and Medical Biophysics, Medical University Innsbruck, 6020, Innsbruck, Austria
| | | | - Yousra El Ghaleb
- Institute of Physiology, Department of Physiology and Medical Biophysics, Medical University Innsbruck, 6020, Innsbruck, Austria
| | - Klaus R Liedl
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Marta Campiglio
- Institute of Physiology, Department of Physiology and Medical Biophysics, Medical University Innsbruck, 6020, Innsbruck, Austria
| | - Bernhard E Flucher
- Institute of Physiology, Department of Physiology and Medical Biophysics, Medical University Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
3
|
Gaitán-Peñas H, Pérez-Rius C, Muhaisen A, Castellanos A, Errasti-Murugarren E, Barrallo-Gimeno A, Alcaraz-Pérez F, Estévez R. Characterization of ClC-1 chloride channels in zebrafish: a new model to study myotonia. J Physiol 2024; 602:3975-3994. [PMID: 39031529 DOI: 10.1113/jp286530] [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: 03/08/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024] Open
Abstract
The function of the chloride channel ClC-1 is crucial for the control of muscle excitability. Thus, reduction of ClC-1 functions by CLCN1 mutations leads to myotonia congenita. Many different animal models have contributed to understanding the myotonia pathophysiology. However, these models do not allow in vivo screening of potentially therapeutic drugs, as the zebrafish model does. In this work, we identified and characterized the two zebrafish orthologues (clc-1a and clc-1b) of the ClC-1 channel. Both channels are mostly expressed in the skeletal muscle as revealed by RT-PCR, western blot, and electrophysiological recordings of myotubes, and clc-1a is predominantly expressed in adult stages. Characterization in Xenopus oocytes shows that the zebrafish channels display similar anion selectivity and voltage dependence to their human counterparts. However, they show reduced sensitivity to the inhibitor 9-anthracenecarboxylic acid (9-AC), and acidic pH inverts the voltage dependence of activation. Reduction of clc-1a/b expression hampers spontaneous and mechanically stimulated movement, which could be reverted by expression of human ClC-1 but not by some ClC-1 containing myotonia mutations. Treatment of clc-1-depleted zebrafish with mexiletine, a typical drug used in human myotonia, improves the motor behaviour. Our work extends the repertoire of ClC channels to evolutionary structure-function studies and proposes the zebrafish clcn1 crispant model as a simple tool to find novel therapies for myotonia. KEY POINTS: We have identified two orthologues of ClC-1 in zebrafish (clc-1a and clc-1b) which are mostly expressed in skeletal muscle at different developmental stages. Functional characterization of the activity of these channels reveals many similitudes with their mammalian counterparts, although they are less sensitive to 9-AC and acidic pH inverts their voltage dependence of gating. Reduction of clc-1a/b expression hampers spontaneous and mechanically stimulated movement which could be reverted by expression of human ClC-1. Myotonia-like symptoms caused by clc-1a/b depletion can be reverted by mexiletine, suggesting that this model could be used to find novel therapies for myotonia.
Collapse
Affiliation(s)
- Héctor Gaitán-Peñas
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona-IDIBELL, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Carla Pérez-Rius
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona-IDIBELL, Barcelona, Spain
| | - Ashraf Muhaisen
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona-IDIBELL, Barcelona, Spain
| | - Aida Castellanos
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona-IDIBELL, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Ekaitz Errasti-Murugarren
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona-IDIBELL, Barcelona, Spain
| | - Alejandro Barrallo-Gimeno
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona-IDIBELL, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Francisca Alcaraz-Pérez
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Department of Surgery, Telomerase, Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Raúl Estévez
- Physiology Unit, Department of Physiological Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona-IDIBELL, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| |
Collapse
|
4
|
Yoo Y, Yeon M, Kim WK, Shin HB, Lee SM, Yoon MS, Ro H, Seo YK. Age-dependent loss of Crls1 causes myopathy and skeletal muscle regeneration failure. Exp Mol Med 2024; 56:922-934. [PMID: 38556544 PMCID: PMC11059380 DOI: 10.1038/s12276-024-01199-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 04/02/2024] Open
Abstract
Skeletal muscle aging results in the gradual suppression of myogenesis, leading to muscle mass loss. However, the specific role of cardiolipin in myogenesis has not been determined. This study investigated the crucial role of mitochondrial cardiolipin and cardiolipin synthase 1 (Crls1) in age-related muscle deterioration and myogenesis. Our findings demonstrated that cardiolipin and Crls1 are downregulated in aged skeletal muscle. Moreover, the knockdown of Crls1 in myoblasts reduced mitochondrial mass, activity, and OXPHOS complex IV expression and disrupted the structure of the mitochondrial cristae. AAV9-shCrls1-mediated downregulation of Crls1 impaired muscle regeneration in a mouse model of cardiotoxin (CTX)-induced muscle damage, whereas AAV9-mCrls1-mediated Crls1 overexpression improved regeneration. Overall, our results highlight that the age-dependent decrease in CRLS1 expression contributes to muscle loss by diminishing mitochondrial quality in skeletal muscle myoblasts. Hence, modulating CRLS1 expression is a promising therapeutic strategy for mitigating muscle deterioration associated with aging, suggesting potential avenues for developing interventions to improve overall muscle health and quality of life in elderly individuals.
Collapse
Affiliation(s)
- Youngbum Yoo
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - MyeongHoon Yeon
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Won-Kyung Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Korea
| | - Hyeon-Bin Shin
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Korea
| | - Seung-Min Lee
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Mee-Sup Yoon
- Department of Molecular Medicine, College of Medicine, Gachon University College of Medicine, Incheon, 21999, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Young-Kyo Seo
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Korea.
- School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
5
|
Kaura V, Hopkins P. Recent advances in skeletal muscle physiology. BJA Educ 2024; 24:84-90. [PMID: 38375493 PMCID: PMC10874741 DOI: 10.1016/j.bjae.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 02/21/2024] Open
Affiliation(s)
- V. Kaura
- Leeds Institute of Medical Research at St James's, University of Leeds, UK
| | - P.M. Hopkins
- Leeds Institute of Medical Research at St James's, University of Leeds, UK
| |
Collapse
|
6
|
Endo Y, Groom L, Wang SM, Pannia E, Griffiths NW, Van Gennip JLM, Ciruna B, Laporte J, Dirksen RT, Dowling JJ. Two zebrafish cacna1s loss-of-function variants provide models of mild and severe CACNA1S-related myopathy. Hum Mol Genet 2024; 33:254-269. [PMID: 37930228 PMCID: PMC10800018 DOI: 10.1093/hmg/ddad178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
CACNA1S-related myopathy, due to pathogenic variants in the CACNA1S gene, is a recently described congenital muscle disease. Disease associated variants result in loss of gene expression and/or reduction of Cav1.1 protein stability. There is an incomplete understanding of the underlying disease pathomechanisms and no effective therapies are currently available. A barrier to the study of this myopathy is the lack of a suitable animal model that phenocopies key aspects of the disease. To address this barrier, we generated knockouts of the two zebrafish CACNA1S paralogs, cacna1sa and cacna1sb. Double knockout fish exhibit severe weakness and early death, and are characterized by the absence of Cav1.1 α1 subunit expression, abnormal triad structure, and impaired excitation-contraction coupling, thus mirroring the severe form of human CACNA1S-related myopathy. A double mutant (cacna1sa homozygous, cacna1sb heterozygote) exhibits normal development, but displays reduced body size, abnormal facial structure, and cores on muscle pathologic examination, thus phenocopying the mild form of human CACNA1S-related myopathy. In summary, we generated and characterized the first cacna1s zebrafish loss-of-function mutants, and show them to be faithful models of severe and mild forms of human CACNA1S-related myopathy suitable for future mechanistic studies and therapy development.
Collapse
Affiliation(s)
- Yukari Endo
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Linda Groom
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642, United States
| | - Sabrina M Wang
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Emanuela Pannia
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Zebrafish Genetics and Disease Models Core Facility, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Nigel W Griffiths
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Jenica L M Van Gennip
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Brian Ciruna
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, Cnrs UMR7104, Université de Strasbourg, 1 Rue Laurent Fries, Illkirch 67400, France
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642, United States
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
- Division of Neurology, Hospital for Sick Children, 555 University Ave, Toronto, ON M5G 1X8, Canada
- Department of Paediatrics, University of Toronto, 555 University Ave, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
7
|
Cisco LA, Sipple MT, Edwards KM, Thornton CA, Lueck JD. Verapamil mitigates chloride and calcium bi-channelopathy in a myotonic dystrophy mouse model. J Clin Invest 2024; 134:e173576. [PMID: 38165038 PMCID: PMC10760957 DOI: 10.1172/jci173576] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024] Open
Abstract
Myotonic dystrophy type 1 (DM1) involves misregulated alternative splicing for specific genes. We used exon or nucleotide deletion to mimic altered splicing of genes central to muscle excitation-contraction coupling in mice. Mice with forced skipping of exon 29 in the CaV1.1 calcium channel combined with loss of ClC-1 chloride channel function displayed markedly reduced lifespan, whereas other combinations of splicing mimics did not affect survival. The Ca2+/Cl- bi-channelopathy mice exhibited myotonia, weakness, and impairment of mobility and respiration. Chronic administration of the calcium channel blocker verapamil rescued survival and improved force generation, myotonia, and respiratory function. These results suggest that Ca2+/Cl- bi-channelopathy contributes to muscle impairment in DM1 and is potentially mitigated by common clinically available calcium channel blockers.
Collapse
Affiliation(s)
| | | | | | - Charles A. Thornton
- Department of Neurology
- Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - John D. Lueck
- Department of Pharmacology and Physiology
- Department of Neurology
- Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
8
|
Terrell K, Choi S, Choi S. Calcium's Role and Signaling in Aging Muscle, Cellular Senescence, and Mineral Interactions. Int J Mol Sci 2023; 24:17034. [PMID: 38069357 PMCID: PMC10706910 DOI: 10.3390/ijms242317034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Calcium research, since its pivotal discovery in the early 1800s through the heating of limestone, has led to the identification of its multi-functional roles. These include its functions as a reducing agent in chemical processes, structural properties in shells and bones, and significant role in cells relating to this review: cellular signaling. Calcium signaling involves the movement of calcium ions within or between cells, which can affect the electrochemical gradients between intra- and extracellular membranes, ligand binding, enzyme activity, and other mechanisms that determine cell fate. Calcium signaling in muscle, as elucidated by the sliding filament model, plays a significant role in muscle contraction. However, as organisms age, alterations occur within muscle tissue. These changes include sarcopenia, loss of neuromuscular junctions, and changes in mineral concentration, all of which have implications for calcium's role. Additionally, a field of study that has gained recent attention, cellular senescence, is associated with aging and disturbed calcium homeostasis, and is thought to affect sarcopenia progression. Changes seen in calcium upon aging may also be influenced by its crosstalk with other minerals such as iron and zinc. This review investigates the role of calcium signaling in aging muscle and cellular senescence. We also aim to elucidate the interactions among calcium, iron, and zinc across various cells and conditions, ultimately deepening our understanding of calcium signaling in muscle aging.
Collapse
Affiliation(s)
| | | | - Sangyong Choi
- Department of Nutritional Sciences, College of Agriculture, Health, and Natural Resources, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
9
|
Bibollet H, Kramer A, Bannister RA, Hernández-Ochoa EO. Advances in Ca V1.1 gating: New insights into permeation and voltage-sensing mechanisms. Channels (Austin) 2023; 17:2167569. [PMID: 36642864 PMCID: PMC9851209 DOI: 10.1080/19336950.2023.2167569] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/09/2023] [Indexed: 01/17/2023] Open
Abstract
The CaV1.1 voltage-gated Ca2+ channel carries L-type Ca2+ current and is the voltage-sensor for excitation-contraction (EC) coupling in skeletal muscle. Significant breakthroughs in the EC coupling field have often been close on the heels of technological advancement. In particular, CaV1.1 was the first voltage-gated Ca2+ channel to be cloned, the first ion channel to have its gating current measured and the first ion channel to have an effectively null animal model. Though these innovations have provided invaluable information regarding how CaV1.1 detects changes in membrane potential and transmits intra- and inter-molecular signals which cause opening of the channel pore and support Ca2+ release from the sarcoplasmic reticulum remain elusive. Here, we review current perspectives on this topic including the recent application of functional site-directed fluorometry.
Collapse
Affiliation(s)
- Hugo Bibollet
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Audra Kramer
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Roger A. Bannister
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erick O. Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
O’Connor TN, Zhao N, Orciuoli HM, Brasile A, Pietrangelo L, He M, Groom L, Leigh J, Mahamed Z, Liang C, Malik S, Protasi F, Dirksen RT. Voluntary wheel running mitigates disease in an Orai1 gain-of-function mouse model of tubular aggregate myopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.559036. [PMID: 37808709 PMCID: PMC10557777 DOI: 10.1101/2023.09.29.559036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Tubular aggregate myopathy (TAM) is an inherited skeletal muscle disease associated with progressive muscle weakness, cramps, and myalgia. Tubular aggregates (TAs) are regular arrays of highly ordered and densely packed SR straight-tubes in muscle biopsies; the extensive presence of TAs represent a key histopathological hallmark of this disease in TAM patients. TAM is caused by gain-of-function mutations in proteins that coordinate store-operated Ca2+ entry (SOCE): STIM1 Ca2+ sensor proteins in the sarcoplasmic reticulum (SR) and Ca2+-permeable ORAI1 channels in the surface membrane. We have previously shown that voluntary wheel running (VWR) prevents formation of TAs in aging mice. Here, we assessed the therapeutic potential of endurance exercise (in the form of VWR) in mitigating the functional and structural alterations in a knock-in mouse model of TAM (Orai1G100S/+ or GS mice) based on a gain-of-function mutation in the ORAI1 pore. WT and GS mice were singly-housed for six months (from two to eight months of age) with either free-spinning or locked low profile wheels. Six months of VWR exercise significantly increased soleus peak tetanic specific force production, normalized FDB fiber Ca2+ store content, and markedly reduced TAs in EDL muscle from GS mice. Six months of VWR exercise normalized the expression of mitochondrial proteins found to be altered in soleus muscle of sedentary GS mice in conjunction with a signature of increased protein translation and biosynthetic processes. Parallel proteomic analyses of EDL muscles from sedentary WT and GS mice revealed changes in a tight network of pathways involved in formation of supramolecular complexes, which were also normalized following six months of VWR. In summary, sustained voluntary endurance exercise improved slow twitch muscle function, reduced the presence of TAs in fast twitch muscle, and normalized the muscle proteome of GS mice consistent with protective adaptions in proteostasis, mitochondrial structure/function, and formation of supramolecular complexes.
Collapse
Affiliation(s)
- Thomas N. O’Connor
- Department of Biomedical Genetics, Genetics and Genomics Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Nan Zhao
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Haley M. Orciuoli
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biology, Biological Sciences, University of Rochester, Rochester, NY, USA
| | - Alice Brasile
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| | - Miao He
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Linda Groom
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jennifer Leigh
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Zahra Mahamed
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Chen Liang
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Feliciano Protasi
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| | - Robert T. Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
11
|
Calle-Ciborro B, Espin-Jaime T, Santos FJ, Gomez-Martin A, Jardin I, Pozo MJ, Rosado JA, Camello PJ, Camello-Almaraz C. Secretion of Interleukin 6 in Human Skeletal Muscle Cultures Depends on Ca 2+ Signalling. BIOLOGY 2023; 12:968. [PMID: 37508398 PMCID: PMC10376320 DOI: 10.3390/biology12070968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023]
Abstract
The systemic effects of physical activity are mediated by the release of IL-6 and other myokines from contracting muscle. Although the release of IL-6 from muscle has been extensively studied, the information on the cellular mechanisms is fragmentary and scarce, especially regarding the role of Ca2+ signals. The aim of this study was to characterize the role of the main components of Ca2+ signals in human skeletal muscle cells during IL-6 secretion stimulated by the Ca2+ mobilizing agonist ATP. Primary cultures were prepared from surgical samples, fluorescence microscopy was used to evaluate the Ca2+ signals and the stimulated release of IL-6 into the medium was determined using ELISA. Intracellular calcium chelator Bapta, low extracellular calcium and the Ca2+ channels blocker La3+ reduced the ATP-stimulated, but not the basal secretion. Secretion was inhibited by blockers of L-type (nifedipine, verapamil), T-type (NNC55-0396) and Orai1 (Synta66) Ca2+ channels and by silencing Orai1 expression. The same effect was achieved with inhibitors of ryanodine receptors (ryanodine, dantrolene) and IP3 receptors (xestospongin C, 2-APB, caffeine). Inhibitors of calmodulin (calmidazolium) and calcineurin (FK506) also decreased secretion. IL-6 transcription in response to ATP was not affected by Bapta or by the T channel blocker. Our results prove that ATP-stimulated IL-6 secretion is mediated at the post-transcriptional level by Ca2+ signals, including the mobilization of calcium stores, the activation of store-operated Ca2+ entry, and the subsequent activation of voltage-operated Ca2+ channels and calmodulin/calcineurin pathways.
Collapse
Affiliation(s)
- Blanca Calle-Ciborro
- Department of Physiology, Instituto de Biomarcadores Patológicos Moleculares y Metabólicos, Universidad de Extremadura, 10003 Cáceres, Spain
| | - Teresa Espin-Jaime
- Faculty of Medicine, Hospital Universitario, Universidad de Extremadura, 06006 Badajoz, Spain
| | | | - Ana Gomez-Martin
- Department of Nursing, Faculty of Nursing and Occupational Therapy, Universidad de Extremadura, 10003 Cáceres, Spain
| | - Isaac Jardin
- Department of Physiology, Instituto de Biomarcadores Patológicos Moleculares y Metabólicos, Universidad de Extremadura, 10003 Cáceres, Spain
| | - Maria J Pozo
- Department of Physiology, Instituto de Biomarcadores Patológicos Moleculares y Metabólicos, Universidad de Extremadura, 10003 Cáceres, Spain
| | - Juan A Rosado
- Department of Physiology, Instituto de Biomarcadores Patológicos Moleculares y Metabólicos, Universidad de Extremadura, 10003 Cáceres, Spain
| | - Pedro J Camello
- Department of Physiology, Instituto de Biomarcadores Patológicos Moleculares y Metabólicos, Universidad de Extremadura, 10003 Cáceres, Spain
| | - Cristina Camello-Almaraz
- Department of Physiology, Instituto de Biomarcadores Patológicos Moleculares y Metabólicos, Universidad de Extremadura, 10003 Cáceres, Spain
| |
Collapse
|
12
|
McClenaghan C, Mukadam MA, Roeglin J, Tryon RC, Grabner M, Dayal A, Meyer GA, Nichols CG. Skeletal muscle delimited myopathy and verapamil toxicity in SUR2 mutant mouse models of AIMS. EMBO Mol Med 2023; 15:e16883. [PMID: 37154692 PMCID: PMC10245035 DOI: 10.15252/emmm.202216883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
ABCC9-related intellectual disability and myopathy syndrome (AIMS) arises from loss-of-function (LoF) mutations in the ABCC9 gene, which encodes the SUR2 subunit of ATP-sensitive potassium (KATP ) channels. KATP channels are found throughout the cardiovascular system and skeletal muscle and couple cellular metabolism to excitability. AIMS individuals show fatigability, muscle spasms, and cardiac dysfunction. We found reduced exercise performance in mouse models of AIMS harboring premature stop codons in ABCC9. Given the roles of KATP channels in all muscles, we sought to determine how myopathy arises using tissue-selective suppression of KATP and found that LoF in skeletal muscle, specifically, underlies myopathy. In isolated muscle, SUR2 LoF results in abnormal generation of unstimulated forces, potentially explaining painful spasms in AIMS. We sought to determine whether excessive Ca2+ influx through CaV 1.1 channels was responsible for myopathology but found that the Ca2+ channel blocker verapamil unexpectedly resulted in premature death of AIMS mice and that rendering CaV 1.1 channels nonpermeable by mutation failed to reverse pathology; results which caution against the use of calcium channel blockers in AIMS.
Collapse
Affiliation(s)
- Conor McClenaghan
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
- Center for Advanced Biotechnology and Medicine, and Departments of Pharmacology and Medicine, Robert Wood Johnson Medical SchoolRutgers UniversityPiscatawayNJUSA
| | - Maya A Mukadam
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| | - Jacob Roeglin
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| | - Robert C Tryon
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| | - Manfred Grabner
- Department of PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Anamika Dayal
- Department of PharmacologyMedical University of InnsbruckInnsbruckAustria
| | - Gretchen A Meyer
- Program in Physical Therapy, Departments of Orthopaedic Surgery, Neurology and Biomedical EngineeringWashington University School of MedicineSt. LouisMOUSA
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, and Department of Cell Biology and PhysiologyWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
13
|
Cisco LA, Sipple MT, Edwards KM, Thornton CA, Lueck JD. Combinatorial chloride and calcium channelopathy in myotonic dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.542752. [PMID: 37398406 PMCID: PMC10312834 DOI: 10.1101/2023.05.29.542752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Myotonic dystrophy type 1 (DM1) involves misregulated alternative splicing for specific genes. We used exon or nucleotide deletion to mimic altered splicing of genes central to muscle excitation-contraction coupling processes in mice. Mice with forced-skipping of exon 29 in CaV1.1 calcium channel combined with loss of ClC-1 chloride channel function showed a markedly reduced lifespan, whereas other combinations of splicing mimics did not affect survival. The Ca2+/Cl- bi-channelopathy mice exhibited myotonia, weakness, and impairment of mobility and respiration. Chronic administration of the calcium channel blocker verapamil rescued survival and improved force generation, myotonia, and respiratory function. These results suggest that Ca2+/Cl- bi-channelopathy contributes to muscle impairment in DM1 and is potentially mitigated by common clinically available calcium channel blockers.
Collapse
Affiliation(s)
- Lily A. Cisco
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Matthew T. Sipple
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Katherine M. Edwards
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Charles A. Thornton
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, Rochester, NY 14642, USA
- Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, Rochester, NY 14642, USA
| | - John D. Lueck
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, Rochester, NY 14642, USA
- Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, Rochester, NY 14642, USA
| |
Collapse
|
14
|
Bibollet H, Nguyen EL, Miranda DR, Ward CW, Voss AA, Schneider MF, Hernández‐Ochoa EO. Voltage sensor current, SR Ca 2+ release, and Ca 2+ channel current during trains of action potential-like depolarizations of skeletal muscle fibers. Physiol Rep 2023; 11:e15675. [PMID: 37147904 PMCID: PMC10163276 DOI: 10.14814/phy2.15675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 05/07/2023] Open
Abstract
In skeletal muscle, CaV 1.1 serves as the voltage sensor for both excitation-contraction coupling (ECC) and L-type Ca2+ channel activation. We have recently adapted the technique of action potential (AP) voltage clamp (APVC) to monitor the current generated by the movement of intramembrane voltage sensors (IQ ) during single imposed transverse tubular AP-like depolarization waveforms (IQAP ). We now extend this procedure to monitoring IQAP , and Ca2+ currents during trains of tubular AP-like waveforms in adult murine skeletal muscle fibers, and compare them with the trajectories of APs and AP-induced Ca2+ release measured in other fibers using field stimulation and optical probes. The AP waveform remains relatively constant during brief trains (<1 sec) for propagating APs in non-V clamped fibers. Trains of 10 AP-like depolarizations at 10 Hz (900 ms), 50 Hz (180 ms), or 100 Hz (90 ms) did not alter IQAP amplitude or kinetics, consistent with previous findings in isolated muscle fibers where negligible charge immobilization occurred during 100 ms step depolarizations. Using field stimulation, Ca2+ release did exhibit a considerable decline from pulse to pulse during the train, also consistent with previous findings, indicating that the decline of Ca2+ release during a short train of APs is not correlated to modification of charge movement. Ca2+ currents during single or 10 Hz trains of AP-like depolarizations were hardly detectable, were minimal during 50 Hz trains, and became more evident during 100 Hz trains in some fibers. Our results verify predictions on the behavior of the ECC machinery in response to AP-like depolarizations and provide a direct demonstration that Ca2+ currents elicited by single AP-like waveforms are negligible, but can become more prominent in some fibers during short high-frequency train stimulation that elicits maximal isometric force.
Collapse
Affiliation(s)
- Hugo Bibollet
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Elton L. Nguyen
- Department of Biological SciencesWright State UniversityDaytonOhioUSA
| | - Daniel R. Miranda
- Department of Biological SciencesWright State UniversityDaytonOhioUSA
| | - Christopher W. Ward
- Department of OrthopedicsUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Andrew A. Voss
- Department of Biological SciencesWright State UniversityDaytonOhioUSA
| | - Martin F. Schneider
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Erick O. Hernández‐Ochoa
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
15
|
Romare M, Elcadi GH, Johansson E, Tsaklis P. Relative Neuroadaptive Effect of Resistance Training along the Descending Neuroaxis in Older Adults. Brain Sci 2023; 13:brainsci13040679. [PMID: 37190644 DOI: 10.3390/brainsci13040679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/01/2023] [Accepted: 04/16/2023] [Indexed: 05/17/2023] Open
Abstract
Age-related decline in voluntary force production represents one of the main contributors to the onset of physical disability in older adults and is argued to stem from adverse musculoskeletal alterations and changes along the descending neuroaxis. The neural contribution of the above is possibly indicated by disproportionate losses in voluntary activation (VA) compared to muscle mass. For young adults, resistance training (RT) induces muscular and neural adaptations over several levels of the central nervous system, contributing to increased physical performance. However, less is known about the relative neuroadaptive contribution of RT in older adults. The aim of this review was to outline the current state of the literature regarding where and to what extent neural adaptations occur along the descending neuroaxis in response to RT in older adults. We performed a literature search in PubMed, Google Scholar and Scopus. A total of 63 articles met the primary inclusion criteria and following quality analysis (PEDro) 23 articles were included. Overall, neuroadaptations in older adults seemingly favor top-down adaptations, where the preceding changes of neural drive from superior levels affect the neural output of lower levels, following RT. Moreover, older adults appear more predisposed to neural rather than morphological adaptations compared to young adults, a potentially important implication for the improved maintenance of neuromuscular function during aging.
Collapse
Affiliation(s)
- Mattias Romare
- ErgoMech-Lab, Department of Physical Education and Sport Science, University of Thessaly, 42100 Trikala, Greece
| | - Guilherme H Elcadi
- ErgoMech-Lab, Department of Physical Education and Sport Science, University of Thessaly, 42100 Trikala, Greece
- Division of Ergonomics, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 14157 Huddinge, Sweden
| | - Elin Johansson
- Pain in Motion Research Group, Departments of Human Physiology and Rehabilitation Sciences, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, PC 1050 Brussel, Belgium
| | - Panagiotis Tsaklis
- ErgoMech-Lab, Department of Physical Education and Sport Science, University of Thessaly, 42100 Trikala, Greece
- Centre of Orthopaedics and Regenerative Medicine, C.O.R.E.-C.I.R.I., Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
- Department of Molecular Medicine and Surgery, Karolinska Institute, SE-171 76 Solna, Sweden
| |
Collapse
|
16
|
Campiglio M, Dyrda A, Tuinte WE, Török E. Ca V1.1 Calcium Channel Signaling Complexes in Excitation-Contraction Coupling: Insights from Channelopathies. Handb Exp Pharmacol 2023; 279:3-39. [PMID: 36592225 DOI: 10.1007/164_2022_627] [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] [Indexed: 01/03/2023]
Abstract
In skeletal muscle, excitation-contraction (EC) coupling relies on the mechanical coupling between two ion channels: the L-type voltage-gated calcium channel (CaV1.1), located in the sarcolemma and functioning as the voltage sensor of EC coupling, and the ryanodine receptor 1 (RyR1), located on the sarcoplasmic reticulum serving as the calcium release channel. To this day, the molecular mechanism by which these two ion channels are linked remains elusive. However, recently, skeletal muscle EC coupling could be reconstituted in heterologous cells, revealing that only four proteins are essential for this process: CaV1.1, RyR1, and the cytosolic proteins CaVβ1a and STAC3. Due to the crucial role of these proteins in skeletal muscle EC coupling, any mutation that affects any one of these proteins can have devastating consequences, resulting in congenital myopathies and other pathologies.Here, we summarize the current knowledge concerning these four essential proteins and discuss the pathophysiology of the CaV1.1, RyR1, and STAC3-related skeletal muscle diseases with an emphasis on the molecular mechanisms. Being part of the same signalosome, mutations in different proteins often result in congenital myopathies with similar symptoms or even in the same disease.
Collapse
Affiliation(s)
- Marta Campiglio
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria.
| | - Agnieszka Dyrda
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Wietske E Tuinte
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Enikő Török
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
17
|
Morera C, Kim J, Paredes-Redondo A, Nobles M, Rybin D, Moccia R, Kowala A, Meng J, Garren S, Liu P, Morgan JE, Muntoni F, Christoforou N, Owens J, Tinker A, Lin YY. CRISPR-mediated correction of skeletal muscle Ca 2+ handling in a novel DMD patient-derived pluripotent stem cell model. Neuromuscul Disord 2022; 32:908-922. [PMID: 36418198 DOI: 10.1016/j.nmd.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 11/05/2022]
Abstract
Mutations in the dystrophin gene cause the most common and currently incurable Duchenne muscular dystrophy (DMD) characterized by progressive muscle wasting. Although abnormal Ca2+ handling is a pathological feature of DMD, mechanisms underlying defective Ca2+ homeostasis remain unclear. Here we generate a novel DMD patient-derived pluripotent stem cell (PSC) model of skeletal muscle with an isogenic control using clustered regularly interspaced short palindromic repeat (CRISPR)-mediated precise gene correction. Transcriptome analysis identifies dysregulated gene sets in the absence of dystrophin, including genes involved in Ca2+ handling, excitation-contraction coupling and muscle contraction. Specifically, analysis of intracellular Ca2+ transients and mathematical modeling of Ca2+ dynamics reveal significantly reduced cytosolic Ca2+ clearance rates in DMD-PSC derived myotubes. Pharmacological assays demonstrate Ca2+ flux in myotubes is determined by both intracellular and extracellular sources. DMD-PSC derived myotubes display significantly reduced velocity of contractility. Compared with a non-isogenic wildtype PSC line, these pathophysiological defects could be rescued by CRISPR-mediated precise gene correction. Our study provides new insights into abnormal Ca2+ homeostasis in DMD and suggests that Ca2+ signaling pathways amenable to pharmacological modulation are potential therapeutic targets. Importantly, we have established a human physiology-relevant in vitro model enabling rapid pre-clinical testing of potential therapies for DMD.
Collapse
Affiliation(s)
- Cristina Morera
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom
| | - Jihee Kim
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom
| | - Amaia Paredes-Redondo
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom; Centre for Predictive in vitro Model, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| | - Muriel Nobles
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Denis Rybin
- Rare Disease Research Unit, Pfizer, 610 Main Street, Cambridge, MA 02139, USA
| | - Robert Moccia
- Rare Disease Research Unit, Pfizer, 610 Main Street, Cambridge, MA 02139, USA
| | - Anna Kowala
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom; Centre for Predictive in vitro Model, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| | - Jinhong Meng
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom
| | - Seth Garren
- Rare Disease Research Unit, Pfizer, 610 Main Street, Cambridge, MA 02139, USA
| | - Pentao Liu
- School of Biomedical Sciences, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, China
| | - Jennifer E Morgan
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom; NIHR Biomedical Research Centre at Great Ormond Street Hospital, Great Ormond Street, London, United Kingdom
| | - Francesco Muntoni
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom; NIHR Biomedical Research Centre at Great Ormond Street Hospital, Great Ormond Street, London, United Kingdom
| | | | - Jane Owens
- Rare Disease Research Unit, Pfizer, 610 Main Street, Cambridge, MA 02139, USA
| | - Andrew Tinker
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Yung-Yao Lin
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom; Centre for Predictive in vitro Model, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom.
| |
Collapse
|
18
|
Tuinte WE, Török E, Mahlknecht I, Tuluc P, Flucher BE, Campiglio M. STAC3 determines the slow activation kinetics of Ca V 1.1 currents and inhibits its voltage-dependent inactivation. J Cell Physiol 2022; 237:4197-4214. [PMID: 36161458 DOI: 10.1002/jcp.30870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/23/2022] [Indexed: 11/06/2022]
Abstract
The skeletal muscle CaV 1.1 channel functions as the voltage-sensor of excitation-contraction (EC) coupling. Recently, the adaptor protein STAC3 was found to be essential for both CaV 1.1 functional expression and EC coupling. Interestingly, STAC proteins were also reported to inhibit calcium-dependent inactivation (CDI) of L-type calcium channels (LTCC), an important negative feedback mechanism in calcium signaling. The same could not be demonstrated for CaV 1.1, as STAC3 is required for its functional expression. However, upon strong membrane depolarization, CaV 1.1 conducts calcium currents characterized by very slow kinetics of activation and inactivation. Therefore, we hypothesized that the negligible inactivation observed in CaV 1.1 currents reflects the inhibitory effect of STAC3. Here, we inserted a triple mutation in the linker region of STAC3 (ETLAAA), as the analogous mutation abolished the inhibitory effect of STAC2 on CDI of CaV 1.3 currents. When coexpressed in CaV 1.1/STAC3 double knockout myotubes, the mutant STAC3-ETLAAA failed to colocalize with CaV 1.1 in the sarcoplasmic reticulum/membrane junctions. However, combined patch-clamp and calcium recording experiments revealed that STAC3-ETLAAA supports CaV 1.1 functional expression and EC coupling, although at a reduced extent compared to wild-type STAC3. Importantly, STAC3-ETLAAA coexpression dramatically accelerated the kinetics of activation and inactivation of CaV 1.1 currents, suggesting that STAC3 determines the slow CaV 1.1 currents kinetics. To examine if STAC3 specifically inhibits the CDI of CaV 1.1 currents, we performed patch-clamp recordings using calcium and barium as charge carriers in HEK cells. While CaV 1.1 displayed negligible CDI with STAC3, this did not increase in the presence of STAC3-ETLAAA. On the contrary, our data demonstrate that STAC3 specifically inhibits the voltage-dependent inactivation (VDI) of CaV 1.1 currents. Altogether, these results designate STAC3 as a crucial determinant for the slow activation kinetics of CaV 1.1 currents and implicate STAC proteins as modulators of both components of inactivation of LTCC.
Collapse
Affiliation(s)
- Wietske E Tuinte
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Enikő Török
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Irene Mahlknecht
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Bernhard E Flucher
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Marta Campiglio
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
19
|
Perni S, Beam K. Junctophilins 1, 2, and 3 all support voltage-induced Ca2+ release despite considerable divergence. J Gen Physiol 2022; 154:212989. [PMID: 35089322 PMCID: PMC9488633 DOI: 10.1085/jgp.202113024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022] Open
Abstract
In skeletal muscle, depolarization of the plasma membrane (PM) causes conformational changes of the calcium channel CaV1.1 that then activate RYR1 to release calcium from the SR. Being independent of extracellular calcium entry, this process is termed voltage-induced calcium release. In skeletal muscle, junctophilins (JPHs) 1 and 2 form the SR–PM junctions at which voltage-induced calcium release occurs. Previous work demonstrated that JPH2 is able to recapitulate voltage-induced calcium release when expressed in HEK293 cells together with CaV1.1, β1a, Stac3, and RYR1. However, it is unknown whether JPH1 and the more distantly related neuronal JPH3 and JPH4 might also function in this manner, a question of interest because different JPH isoforms diverge in their interactions with RYR1. Here, we show that, like JPH2, JPH1 and JPH3, coexpressed with CaV1.1, β1a, Stac3, and RYR1 in HEK293 cells, cause colocalization of CaV1.1 and RYR1 at ER–PM junctions. Furthermore, potassium depolarization elicited cytoplasmic calcium transients in cells in which WT CaV1.1 was replaced with the calcium impermeant mutant CaV1.1(N617D), indicating that JPH1, JPH2, and JPH3 can all support voltage-induced calcium release, despite sequence divergence and differences in interaction with RYR1. Conversely, JPH4-induced ER–PM junctions contain CaV1.1 but not RYR1, and cells expressing JPH4 are unable to produce depolarization-induced calcium transients. Thus, JPHs seem to act primarily to form ER–PM junctions and to recruit the necessary signaling proteins to these junctions but appear not to be directly involved in the functional interactions between these proteins.
Collapse
Affiliation(s)
- Stefano Perni
- Department of Physiology and Biophysics, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Kurt Beam
- Department of Physiology and Biophysics, University of Colorado, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
20
|
Bolaños P, Calderón JC. Excitation-contraction coupling in mammalian skeletal muscle: Blending old and last-decade research. Front Physiol 2022; 13:989796. [PMID: 36117698 PMCID: PMC9478590 DOI: 10.3389/fphys.2022.989796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The excitation–contraction coupling (ECC) in skeletal muscle refers to the Ca2+-mediated link between the membrane excitation and the mechanical contraction. The initiation and propagation of an action potential through the membranous system of the sarcolemma and the tubular network lead to the activation of the Ca2+-release units (CRU): tightly coupled dihydropyridine and ryanodine (RyR) receptors. The RyR gating allows a rapid, massive, and highly regulated release of Ca2+ from the sarcoplasmic reticulum (SR). The release from triadic places generates a sarcomeric gradient of Ca2+ concentrations ([Ca2+]) depending on the distance of a subcellular region from the CRU. Upon release, the diffusing Ca2+ has multiple fates: binds to troponin C thus activating the contractile machinery, binds to classical sarcoplasmic Ca2+ buffers such as parvalbumin, adenosine triphosphate and, experimentally, fluorescent dyes, enters the mitochondria and the SR, or is recycled through the Na+/Ca2+ exchanger and store-operated Ca2+ entry (SOCE) mechanisms. To commemorate the 7th decade after being coined, we comprehensively and critically reviewed “old”, historical landmarks and well-established concepts, and blended them with recent advances to have a complete, quantitative-focused landscape of the ECC. We discuss the: 1) elucidation of the CRU structures at near-atomic resolution and its implications for functional coupling; 2) reliable quantification of peak sarcoplasmic [Ca2+] using fast, low affinity Ca2+ dyes and the relative contributions of the Ca2+-binding mechanisms to the whole concert of Ca2+ fluxes inside the fibre; 3) articulation of this novel quantitative information with the unveiled structural details of the molecular machinery involved in mitochondrial Ca2+ handing to understand how and how much Ca2+ enters the mitochondria; 4) presence of the SOCE machinery and its different modes of activation, which awaits understanding of its magnitude and relevance in situ; 5) pharmacology of the ECC, and 6) emerging topics such as the use and potential applications of super-resolution and induced pluripotent stem cells (iPSC) in ECC. Blending the old with the new works better!
Collapse
Affiliation(s)
- Pura Bolaños
- Laboratory of Cellular Physiology, Centre of Biophysics and Biochemistry, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela
| | - Juan C. Calderón
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellín, Colombia
- *Correspondence: Juan C. Calderón,
| |
Collapse
|
21
|
Melzer W. From α1s splicing to γ1 function: A new twist in subunit modulation of the skeletal muscle L-type Ca2+ channel. J Gen Physiol 2022; 154:213270. [PMID: 35674662 PMCID: PMC9184848 DOI: 10.1085/jgp.202213182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Melzer discusses a recent JGP study showing that alternative splicing of the skeletal muscle L-type calcium channel impacts on a modulatory effect of its γ subunit.
Collapse
Affiliation(s)
- Werner Melzer
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
22
|
The distal C terminus of the dihydropyridine receptor β 1a subunit is essential for tetrad formation in skeletal muscle. Proc Natl Acad Sci U S A 2022; 119:e2201136119. [PMID: 35507876 PMCID: PMC9171810 DOI: 10.1073/pnas.2201136119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
SignificanceVertebrate skeletal muscle excitation-contraction coupling (ECC) is based on Ca2+-influx-independent interchannel cross-talk between DHPR and RyR1. The skeletal muscle DHPR complex consists of the main, voltage-sensing, and pore-forming α1S subunit, the auxiliary β1a, α2δ-1, γ1 subunits, and Stac3. The DHPRβ1a subunit plays an essential role in full triad targeting of DHPRα1S, voltage sensing, and tetrad formation (grouping of four DHPRs)-the three prerequisites for skeletal muscle ECC. Hence, a lack of DHPRβ1a results in a lethal phenotype in both β1-null mice and zebrafish. Here, we identified the nonconserved, distal C terminus of DHPRβ1a as playing a pivotal role in the formation of DHPR tetrads, and thus allosteric DHPR-RyR1 coupling, essential for proper skeletal muscle ECC.
Collapse
|
23
|
Sirt6 reprograms myofibers to oxidative type through CREB-dependent Sox6 suppression. Nat Commun 2022; 13:1808. [PMID: 35379817 PMCID: PMC8980083 DOI: 10.1038/s41467-022-29472-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/17/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractExpanding the exercise capacity of skeletal muscle is an emerging strategy to combat obesity-related metabolic diseases and this can be achieved by shifting skeletal muscle fibers toward slow-twitch oxidative type. Here, we report that Sirt6, an anti-aging histone deacetylase, is critical in regulating myofiber configuration toward oxidative type and that Sirt6 activator can be an exercise mimetic. Genetic inactivation of Sirt6 in skeletal muscle reduced while its transgenic overexpression increased mitochondrial oxidative capacity and exercise performance in mice. Mechanistically, we show that Sirt6 downregulated Sox6, a key repressor of slow fiber specific gene, by increasing the transcription of CREB. Sirt6 expression is elevated in chronically exercised humans, and mice treated with an activator of Sirt6 showed an increase in exercise endurance as compared to exercise-trained controls. Thus, the current study identifies Sirt6 as a molecular target for reprogramming myofiber composition toward the oxidative type and for improving muscle performance.
Collapse
|
24
|
Ion Channel Partnerships: Odd and Not-So-Odd Couples Controlling Neuronal Ion Channel Function. Int J Mol Sci 2022; 23:ijms23041953. [PMID: 35216068 PMCID: PMC8878034 DOI: 10.3390/ijms23041953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
The concerted function of the large number of ion channels expressed in excitable cells, including brain neurons, shapes diverse signaling events by controlling the electrical properties of membranes. It has long been recognized that specific groups of ion channels are functionally coupled in mediating ionic fluxes that impact membrane potential, and that these changes in membrane potential impact ion channel gating. Recent studies have identified distinct sets of ion channels that can also physically and functionally associate to regulate the function of either ion channel partner beyond that afforded by changes in membrane potential alone. Here, we review canonical examples of such ion channel partnerships, in which a Ca2+ channel is partnered with a Ca2+-activated K+ channel to provide a dedicated route for efficient coupling of Ca2+ influx to K+ channel activation. We also highlight examples of non-canonical ion channel partnerships between Ca2+ channels and voltage-gated K+ channels that are not intrinsically Ca2+ sensitive, but whose partnership nonetheless yields enhanced regulation of one or the other ion channel partner. We also discuss how these ion channel partnerships can be shaped by the subcellular compartments in which they are found and provide perspectives on how recent advances in techniques to identify proteins in close proximity to one another in native cells may lead to an expanded knowledge of other ion channel partnerships.
Collapse
|
25
|
Jaque-Fernández F, Jorquera G, Troc-Gajardo J, Pietri-Rouxel F, Gentil C, Buvinic S, Allard B, Jaimovich E, Jacquemond V, Casas M. Pannexin-1 and CaV1.1 show reciprocal interaction during excitation-contraction and excitation-transcription coupling in skeletal muscle. J Gen Physiol 2021; 153:212695. [PMID: 34636893 PMCID: PMC8515650 DOI: 10.1085/jgp.202012635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/24/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023] Open
Abstract
One of the most important functions of skeletal muscle is to respond to nerve stimuli by contracting. This function ensures body movement but also participates in other important physiological roles, like regulation of glucose homeostasis. Muscle activity is closely regulated to adapt to different demands and shows a plasticity that relies on both transcriptional activity and nerve stimuli. These two processes, both dependent on depolarization of the plasma membrane, have so far been regarded as separated and independent processes due to a lack of evidence of common protein partners or molecular mechanisms. In this study, we reveal intimate functional interactions between the process of excitation-induced contraction and the process of excitation-induced transcriptional activity in skeletal muscle. We show that the plasma membrane voltage-sensing protein CaV1.1 and the ATP-releasing channel Pannexin-1 (Panx1) regulate each other in a reciprocal manner, playing roles in both processes. Specifically, knockdown of CaV1.1 produces chronically elevated extracellular ATP concentrations at rest, consistent with disruption of the normal control of Panx1 activity. Conversely, knockdown of Panx1 affects not only activation of transcription but also CaV1.1 function on the control of muscle fiber contraction. Altogether, our results establish the presence of bidirectional functional regulations between the molecular machineries involved in the control of contraction and transcription induced by membrane depolarization of adult muscle fibers. Our results are important for an integrative understanding of skeletal muscle function and may impact our understanding of several neuromuscular diseases.
Collapse
Affiliation(s)
- Francisco Jaque-Fernández
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Gonzalo Jorquera
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jennifer Troc-Gajardo
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - France Pietri-Rouxel
- Université Pierre et Marie Curie, Université Paris 06, Institut National de la Santé et de la Recherche Médicale/Centre National de la Recherche Scientifique/Institut de Myologie/Centre de Recherche en Myologie, Groupement hospitalier universitaire Pitié Salpêtrière, Paris, France
| | - Christel Gentil
- Université Pierre et Marie Curie, Université Paris 06, Institut National de la Santé et de la Recherche Médicale/Centre National de la Recherche Scientifique/Institut de Myologie/Centre de Recherche en Myologie, Groupement hospitalier universitaire Pitié Salpêtrière, Paris, France
| | - Sonja Buvinic
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Bruno Allard
- Université Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR-5310, Institut National de la Santé et de la Recherche Médicale U-1217, Institut NeuroMyoGène, Lyon, France
| | - Enrique Jaimovich
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Vincent Jacquemond
- Université Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR-5310, Institut National de la Santé et de la Recherche Médicale U-1217, Institut NeuroMyoGène, Lyon, France
| | - Mariana Casas
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
26
|
Voltage sensor movements of Ca V1.1 during an action potential in skeletal muscle fibers. Proc Natl Acad Sci U S A 2021; 118:2026116118. [PMID: 34583989 DOI: 10.1073/pnas.2026116118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 11/18/2022] Open
Abstract
The skeletal muscle L-type Ca2+ channel (CaV1.1) works primarily as a voltage sensor for skeletal muscle action potential (AP)-evoked Ca2+ release. CaV1.1 contains four distinct voltage-sensing domains (VSDs), yet the contribution of each VSD to AP-evoked Ca2+ release remains unknown. To investigate the role of VSDs in excitation-contraction coupling (ECC), we encoded cysteine substitutions on each S4 voltage-sensing segment of CaV1.1, expressed each construct via in vivo gene transfer electroporation, and used in cellulo AP fluorometry to track the movement of each CaV1.1 VSD in skeletal muscle fibers. We first provide electrical measurements of CaV1.1 voltage sensor charge movement in response to an AP waveform. Then we characterize the fluorescently labeled channels' VSD fluorescence signal responses to an AP and compare them with the waveforms of the electrically measured charge movement, the optically measured free myoplasmic Ca2+, and the calculated rate of Ca2+ release from the sarcoplasmic reticulum for an AP, the physiological signal for skeletal muscle fiber activation. A considerable fraction of the fluorescence signal for each VSD occurred after the time of peak Ca2+ release, and even more occurred after the earlier peak of electrically measured charge movement during an AP, and thus could not directly reflect activation of Ca2+ release or charge movement, respectively. However, a sizable fraction of the fluorometric signals for VSDs I, II, and IV, but not VSDIII, overlap the rising phase of charge moved, and even more for Ca2+ release, and thus could be involved in voltage sensor rearrangements or Ca2+ release activation.
Collapse
|
27
|
Woll KA, Van Petegem F. Calcium Release Channels: Structure and Function of IP3 Receptors and Ryanodine Receptors. Physiol Rev 2021; 102:209-268. [PMID: 34280054 DOI: 10.1152/physrev.00033.2020] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ca2+-release channels are giant membrane proteins that control the release of Ca2+ from the endoplasmic and sarcoplasmic reticulum. The two members, ryanodine receptors (RyRs) and inositol-1,4,5-trisphosphate Receptors (IP3Rs), are evolutionarily related and are both activated by cytosolic Ca2+. They share a common architecture, but RyRs have evolved additional modules in the cytosolic region. Their massive size allows for the regulation by tens of proteins and small molecules, which can affect the opening and closing of the channels. In addition to Ca2+, other major triggers include IP3 for the IP3Rs, and depolarization of the plasma membrane for a particular RyR subtype. Their size has made them popular targets for study via electron microscopic methods, with current structures culminating near 3Å. The available structures have provided many new mechanistic insights int the binding of auxiliary proteins and small molecules, how these can regulate channel opening, and the mechanisms of disease-associated mutations. They also help scrutinize previously proposed binding sites, as some of these are now incompatible with the structures. Many questions remain around the structural effects of post-translational modifications, additional binding partners, and the higher-order complexes these channels can make in situ. This review summarizes our current knowledge about the structures of Ca2+-release channels and how this informs on their function.
Collapse
Affiliation(s)
- Kellie A Woll
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
28
|
Miranda DR, Voss AA, Bannister RA. Into the spotlight: RGK proteins in skeletal muscle. Cell Calcium 2021; 98:102439. [PMID: 34261001 DOI: 10.1016/j.ceca.2021.102439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 10/20/2022]
Abstract
The RGK (Rad, Rem, Rem2 and Gem/Kir) family of small GTPases are potent endogenous inhibitors of voltage-gated Ca2+ channels (VGCCs). While the impact of RGK proteins on cardiac physiology has been investigated extensively, much less is known regarding their influence on skeletal muscle biology. Thus, the purpose of this article is to establish a basis for future investigation into the role of RGK proteins in regulating the skeletal muscle excitation-contraction (EC) coupling complex via modulation of the L-type CaV1.1 VGCC. The pathological consequences of elevated muscle RGK protein expression in Type II Diabetes, Amyotrophic Lateral Sclerosis (ALS), Duchenne's Muscular Dystrophy and traumatic nerve injury are also discussed.
Collapse
Affiliation(s)
- Daniel R Miranda
- Department of Biological Sciences, College of Science and Mathematics, Wright State University, 235A Biological Sciences, 3640 Colonel Glenn Highway, Dayton, OH 45435, USA
| | - Andrew A Voss
- Department of Biological Sciences, College of Science and Mathematics, Wright State University, 235A Biological Sciences, 3640 Colonel Glenn Highway, Dayton, OH 45435, USA.
| | - Roger A Bannister
- Departments of Pathology and Biochemistry & Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
29
|
Hostrup M, Cairns SP, Bangsbo J. Muscle Ionic Shifts During Exercise: Implications for Fatigue and Exercise Performance. Compr Physiol 2021; 11:1895-1959. [PMID: 34190344 DOI: 10.1002/cphy.c190024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Exercise causes major shifts in multiple ions (e.g., K+ , Na+ , H+ , lactate- , Ca2+ , and Cl- ) during muscle activity that contributes to development of muscle fatigue. Sarcolemmal processes can be impaired by the trans-sarcolemmal rundown of ion gradients for K+ , Na+ , and Ca2+ during fatiguing exercise, while changes in gradients for Cl- and Cl- conductance may exert either protective or detrimental effects on fatigue. Myocellular H+ accumulation may also contribute to fatigue development by lowering glycolytic rate and has been shown to act synergistically with inorganic phosphate (Pi) to compromise cross-bridge function. In addition, sarcoplasmic reticulum Ca2+ release function is severely affected by fatiguing exercise. Skeletal muscle has a multitude of ion transport systems that counter exercise-related ionic shifts of which the Na+ /K+ -ATPase is of major importance. Metabolic perturbations occurring during exercise can exacerbate trans-sarcolemmal ionic shifts, in particular for K+ and Cl- , respectively via metabolic regulation of the ATP-sensitive K+ channel (KATP ) and the chloride channel isoform 1 (ClC-1). Ion transport systems are highly adaptable to exercise training resulting in an enhanced ability to counter ionic disturbances to delay fatigue and improve exercise performance. In this article, we discuss (i) the ionic shifts occurring during exercise, (ii) the role of ion transport systems in skeletal muscle for ionic regulation, (iii) how ionic disturbances affect sarcolemmal processes and muscle fatigue, (iv) how metabolic perturbations exacerbate ionic shifts during exercise, and (v) how pharmacological manipulation and exercise training regulate ion transport systems to influence exercise performance in humans. © 2021 American Physiological Society. Compr Physiol 11:1895-1959, 2021.
Collapse
Affiliation(s)
- Morten Hostrup
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Simeon Peter Cairns
- SPRINZ, School of Sport and Recreation, Auckland University of Technology, Auckland, New Zealand.,Health and Rehabilitation Research Institute, Auckland University of Technology, Auckland, New Zealand
| | - Jens Bangsbo
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Dayal A, Fernández-Quintero ML, Liedl KR, Grabner M. Pore mutation N617D in the skeletal muscle DHPR blocks Ca 2+ influx due to atypical high-affinity Ca 2+ binding. eLife 2021; 10:63435. [PMID: 34061024 PMCID: PMC8184209 DOI: 10.7554/elife.63435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 05/28/2021] [Indexed: 11/13/2022] Open
Abstract
Skeletal muscle excitation-contraction (EC) coupling roots in Ca2+-influx-independent inter-channel signaling between the sarcolemmal dihydropyridine receptor (DHPR) and the ryanodine receptor (RyR1) in the sarcoplasmic reticulum. Although DHPR Ca2+ influx is irrelevant for EC coupling, its putative role in other muscle-physiological and developmental pathways was recently examined using two distinct genetically engineered mouse models carrying Ca2+ non-conducting DHPRs: DHPR(N617D) (Dayal et al., 2017) and DHPR(E1014K) (Lee et al., 2015). Surprisingly, despite complete block of DHPR Ca2+-conductance, histological, biochemical, and physiological results obtained from these two models were contradictory. Here, we characterize the permeability and selectivity properties and henceforth the mechanism of Ca2+ non-conductance of DHPR(N617). Our results reveal that only mutant DHPR(N617D) with atypical high-affinity Ca2+ pore-binding is tight for physiologically relevant monovalent cations like Na+ and K+. Consequently, we propose a molecular model of cooperativity between two ion selectivity rings formed by negatively charged residues in the DHPR pore region.
Collapse
Affiliation(s)
- Anamika Dayal
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Klaus R Liedl
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Manfred Grabner
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
31
|
Gonçalves LDS, Sales LP, Saito TR, Campos JC, Fernandes AL, Natali J, Jensen L, Arnold A, Ramalho L, Bechara LRG, Esteca MV, Correa I, Sant'Anna D, Ceroni A, Michelini LC, Gualano B, Teodoro W, Carvalho VH, Vargas BS, Medeiros MHG, Baptista IL, Irigoyen MC, Sale C, Ferreira JCB, Artioli GG. Histidine dipeptides are key regulators of excitation-contraction coupling in cardiac muscle: Evidence from a novel CARNS1 knockout rat model. Redox Biol 2021; 44:102016. [PMID: 34038814 PMCID: PMC8144739 DOI: 10.1016/j.redox.2021.102016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 12/04/2022] Open
Abstract
Histidine-containing dipeptides (HCDs) are abundantly expressed in striated muscles. Although important properties have been ascribed to HCDs, including H+ buffering, regulation of Ca2+ transients and protection against oxidative stress, it remains unknown whether they play relevant functions in vivo. To investigate the in vivo roles of HCDs, we developed the first carnosine synthase knockout (CARNS1−/−) rat strain to investigate the impact of an absence of HCDs on skeletal and cardiac muscle function. Male wild-type (WT) and knockout rats (4 months-old) were used. Skeletal muscle function was assessed by an exercise tolerance test, contractile function in situ and muscle buffering capacity in vitro. Cardiac function was assessed in vivo by echocardiography and cardiac electrical activity by electrocardiography. Cardiomyocyte contractile function was assessed in isolated cardiomyocytes by measuring sarcomere contractility, along with the determination of Ca2+ transient. Markers of oxidative stress, mitochondrial function and expression of proteins were also evaluated in cardiac muscle. Animals were supplemented with carnosine (1.8% in drinking water for 12 weeks) in an attempt to rescue tissue HCDs levels and function. CARNS1−/− resulted in the complete absence of carnosine and anserine, but it did not affect exercise capacity, skeletal muscle force production, fatigability or buffering capacity in vitro, indicating that these are not essential for pH regulation and function in skeletal muscle. In cardiac muscle, however, CARNS1−/− resulted in a significant impairment of contractile function, which was confirmed both in vivo and ex vivo in isolated sarcomeres. Impaired systolic and diastolic dysfunction were accompanied by reduced intracellular Ca2+ peaks and slowed Ca2+ removal, but not by increased markers of oxidative stress or impaired mitochondrial respiration. No relevant increases in muscle carnosine content were observed after carnosine supplementation. Results show that a primary function of HCDs in cardiac muscle is the regulation of Ca2+ handling and excitation-contraction coupling.
Collapse
Affiliation(s)
- Lívia de Souza Gonçalves
- Applied Physiology & Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina, Divisão de Reumatologia, Universidade de São Paulo, SP, Brazil; Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Brazil
| | - Lucas Peixoto Sales
- Applied Physiology & Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina, Divisão de Reumatologia, Universidade de São Paulo, SP, Brazil; Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Brazil
| | - Tiemi Raquel Saito
- Applied Physiology & Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina, Divisão de Reumatologia, Universidade de São Paulo, SP, Brazil; Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Brazil
| | | | - Alan Lins Fernandes
- Applied Physiology & Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina, Divisão de Reumatologia, Universidade de São Paulo, SP, Brazil; Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Brazil
| | - José Natali
- Applied Physiology & Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina, Divisão de Reumatologia, Universidade de São Paulo, SP, Brazil
| | - Leonardo Jensen
- Laboratório de Hipertensão do Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Brazil
| | - Alexandre Arnold
- Laboratório de Hipertensão do Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Brazil
| | - Lisley Ramalho
- Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | | | - Marcos Vinicius Esteca
- Laboratory of Cell and Tissue Biology, Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Brazil
| | - Isis Correa
- Laboratório de Hipertensão do Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Brazil
| | - Diogo Sant'Anna
- Laboratório de Hipertensão do Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Brazil
| | - Alexandre Ceroni
- Departamento de Fisiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | | | - Bruno Gualano
- Applied Physiology & Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina, Divisão de Reumatologia, Universidade de São Paulo, SP, Brazil; Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Brazil
| | - Walcy Teodoro
- Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Brazil
| | | | | | | | - Igor Luchini Baptista
- Laboratory of Cell and Tissue Biology, Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Brazil
| | - Maria Cláudia Irigoyen
- Laboratório de Hipertensão do Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Brazil
| | - Craig Sale
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, Nottingham Trent University, UK
| | | | - Guilherme Giannini Artioli
- Applied Physiology & Nutrition Research Group, School of Physical Education and Sport, Faculdade de Medicina, Divisão de Reumatologia, Universidade de São Paulo, SP, Brazil; Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, Brazil.
| |
Collapse
|
32
|
Myers JH, Denman K, DuPont C, Hawash AA, Novak KR, Koesters A, Grabner M, Dayal A, Voss AA, Rich MM. The mechanism underlying transient weakness in myotonia congenita. eLife 2021; 10:e65691. [PMID: 33904400 PMCID: PMC8079152 DOI: 10.7554/elife.65691] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/24/2021] [Indexed: 12/23/2022] Open
Abstract
In addition to the hallmark muscle stiffness, patients with recessive myotonia congenita (Becker disease) experience debilitating bouts of transient weakness that remain poorly understood despite years of study. We performed intracellular recordings from muscle of both genetic and pharmacologic mouse models of Becker disease to identify the mechanism underlying transient weakness. Our recordings reveal transient depolarizations (plateau potentials) of the membrane potential to -25 to -35 mV in the genetic and pharmacologic models of Becker disease. Both Na+ and Ca2+ currents contribute to plateau potentials. Na+ persistent inward current (NaPIC) through NaV1.4 channels is the key trigger of plateau potentials and current through CaV1.1 Ca2+ channels contributes to the duration of the plateau. Inhibiting NaPIC with ranolazine prevents the development of plateau potentials and eliminates transient weakness in vivo. These data suggest that targeting NaPIC may be an effective treatment to prevent transient weakness in myotonia congenita.
Collapse
Affiliation(s)
- Jessica H Myers
- Department of Neuroscience, Cell Biology and Physiology, Wright State UniversityDaytonUnited States
| | - Kirsten Denman
- Department of Neuroscience, Cell Biology and Physiology, Wright State UniversityDaytonUnited States
| | - Chris DuPont
- Department of Neuroscience, Cell Biology and Physiology, Wright State UniversityDaytonUnited States
| | - Ahmed A Hawash
- Department of Dermatology & Cutaneous Surgery, University of MiamiMiamiUnited States
| | | | - Andrew Koesters
- Naval Medical Research Unit, Wright Patterson Air Force BaseDaytonUnited States
| | - Manfred Grabner
- Department of Pharmacology, Medical University of InnsbruckInnsbruckAustria
| | - Anamika Dayal
- Department of Pharmacology, Medical University of InnsbruckInnsbruckAustria
| | - Andrew A Voss
- Department of Biology, Wright State UniversityDaytonUnited States
| | - Mark M Rich
- Department of Neuroscience, Cell Biology and Physiology, Wright State UniversityDaytonUnited States
| |
Collapse
|
33
|
Nagasaka T, Hata T, Shindo K, Adachi Y, Takeuchi M, Saito K, Takiyama Y. Morphological Alterations of the Sarcotubular System in Permanent Myopathy of Hereditary Hypokalemic Periodic Paralysis with a Mutation in the CACNA1S Gene. J Neuropathol Exp Neurol 2021; 79:1276-1292. [PMID: 33184660 DOI: 10.1093/jnen/nlaa098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We investigated the immunohistochemical localization of several proteins related to excitation-contraction coupling and ultrastructural alterations of the sarcotubular system in biopsied muscles from a father and a daughter in a family with permanent myopathy with hypokalemic periodic paralysis (PMPP) due to a mutation in calcium channel CACNA1S; p. R1239H hetero. Immunostaining for L-type calcium channels (LCaC) showed linear hyper-stained regions indicating proliferation of longitudinal t-tubules. The margin of vacuoles was positive for ryanodine receptor, LCaC, calsequestrin (CASQ) 1, CASQ 2, SR/ER Ca2+-ATPase (SERCA) 1, SERCA2, dysferlin, dystrophin, α-actinin, LC3, and LAMP 1. Electron microscopy indicated that the vacuoles mainly originated from the sarcoplasmic reticulum (SR). These findings indicate impairment of the muscle contraction system related to Ca2+ dynamics, remodeling of t-tubules and muscle fiber repair. We speculate that PMPP in patients with a CACNA1S mutation might start with abnormal SR function due to impaired LCaC. Subsequent induction of muscular contractile abnormalities and the vacuoles formed by fused SR in the repair process including autophagy might result in permanent myopathy. Our findings may facilitate prediction of the pathomechanisms of PMPP seen on morphological observation.
Collapse
Affiliation(s)
- Takamura Nagasaka
- Department of Neurology, Faculty of Medicine, University of Yamanashi, Chuou-city, Yamanashi, Japan
| | - Takanori Hata
- Department of Neurology, Faculty of Medicine, University of Yamanashi, Chuou-city, Yamanashi, Japan
| | - Kazumasa Shindo
- Department of Neurology, Faculty of Medicine, University of Yamanashi, Chuou-city, Yamanashi, Japan
| | - Yoshiki Adachi
- Department of Neurology, Matsue Medical Center, National Hospital Organization, Shimane, Japan
| | | | - Kayoko Saito
- Institute of Medical Genetics, Tokyo Women's University, Tokyo, Japan
| | - Yoshihisa Takiyama
- Department of Neurology, Faculty of Medicine, University of Yamanashi, Chuou-city, Yamanashi, Japan
| |
Collapse
|
34
|
Balderas-Villalobos J, Steele TWE, Eltit JM. Physiological and Pathological Relevance of Selective and Nonselective Ca 2+ Channels in Skeletal and Cardiac Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:225-247. [PMID: 35138617 PMCID: PMC10683374 DOI: 10.1007/978-981-16-4254-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Contraction of the striated muscle is fundamental for human existence. The action of voluntary skeletal muscle enables activities such as breathing, establishing body posture, and diverse body movements. Additionally, highly precise motion empowers communication, artistic expression, and other activities that define everyday human life. The involuntary contraction of striated muscle is the core function of the heart and is essential for blood flow. Several ion channels are important in the transduction of action potentials to cytosolic Ca2+ signals that enable muscle contraction; however, other ion channels are involved in the progression of muscle pathologies that can impair normal life or threaten it. This chapter describes types of selective and nonselective Ca2+ permeable ion channels expressed in the striated muscle, their participation in different aspects of muscle excitation and contraction, and their relevance to the progression of some pathological states.
Collapse
Affiliation(s)
- Jaime Balderas-Villalobos
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tyler W E Steele
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
35
|
Cserne Szappanos H, Vincze J, Bodnár D, Dienes B, Schneider MF, Csernoch L, Szentesi P. High Time Resolution Analysis of Voltage-Dependent and Voltage-Independent Calcium Sparks in Frog Skeletal Muscle Fibers. Front Physiol 2020; 11:599822. [PMID: 33384612 PMCID: PMC7769825 DOI: 10.3389/fphys.2020.599822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/25/2020] [Indexed: 11/29/2022] Open
Abstract
In amphibian skeletal muscle calcium (Ca2+) sparks occur both as voltage-dependent and voltage-independent ligand-activated release events. However, whether their properties and their origin show similarities are still in debate. Elevated K+, constant Cl- content solutions were used to initiate small depolarizations of the resting membrane potential to activate dihydropyridine receptors (DHPR) and caffeine to open ryanodine receptors (RyR) on intact fibers. The properties of Ca2+ sparks observed under control conditions were compared to those measured on depolarized cells and those after caffeine treatment. Calcium sparks were recorded on intact frog skeletal muscle fibers using high time resolution confocal microscopy (x-y scan: 30 Hz). Sparks were elicited by 1 mmol/l caffeine or subthreshold depolarization to different membrane potentials. Both treatments increased the frequency of sparks and altered their morphology. Images were analyzed by custom-made computer programs. Both the amplitude (in ΔF/F0; 0.259 ± 0.001 vs. 0.164 ± 0.001; n = 24942 and 43326, respectively; mean ± SE, p < 0.001) and the full width at half maximum (FWHM, in μm; parallel with fiber axis: 2.34 ± 0.01 vs. 1.92 ± 0.01, p < 0.001; perpendicular to fiber axis: 2.08 ± 0.01 vs. 1.68 ± 0.01, p < 0.001) of sparks was significantly greater after caffeine treatment than on depolarized cells. 9.8% of the sparks detected on depolarized fibers and about one third of the caffeine activated sparks (29.7%) overlapped with another one on the previous frame on x-y scans. Centre of overlapping sparks travelled significantly longer distances between consecutive frames after caffeine treatment then after depolarization (in μm; 1.66 ± 0.01 vs. 0.95 ± 0.01, p < 0.001). Our results suggest that the two types of ryanodine receptors, the junctional RyRs controlled by DHPRs and the parajunctional RyRs are activated independently, using alternate ways, with the possibility of cooperation between neighboring release channels.
Collapse
Affiliation(s)
- Henrietta Cserne Szappanos
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Vincze
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dóra Bodnár
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Martin F. Schneider
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
36
|
Melzer W. ECC meets CEU-New focus on the backdoor for calcium ions in skeletal muscle cells. J Gen Physiol 2020; 152:152046. [PMID: 32851409 PMCID: PMC7537343 DOI: 10.1085/jgp.202012679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In this issue, Michelucci et al. report the existence of specific sites acting as Ca2+ entry units (CEUs) in fast skeletal muscle of mice lacking calsequestrin (CASQ1), the major Ca2+ binding protein of the SR. The CEU provides constitutive and store-operated Ca2+ entry (SOCE) and resistance to force decline resulting from SR Ca2+ depletion during repetitive muscle activity.
Collapse
Affiliation(s)
- Werner Melzer
- Institute of Applied Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
37
|
FABP3-mediated membrane lipid saturation alters fluidity and induces ER stress in skeletal muscle with aging. Nat Commun 2020; 11:5661. [PMID: 33168829 PMCID: PMC7653047 DOI: 10.1038/s41467-020-19501-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/14/2020] [Indexed: 12/19/2022] Open
Abstract
Sarcopenia is characterized by decreased skeletal muscle mass and function with age. Aged muscles have altered lipid compositions; however, the role and regulation of lipids are unknown. Here we report that FABP3 is upregulated in aged skeletal muscles, disrupting homeostasis via lipid remodeling. Lipidomic analyses reveal that FABP3 overexpression in young muscles alters the membrane lipid composition to that of aged muscle by decreasing polyunsaturated phospholipid acyl chains, while increasing sphingomyelin and lysophosphatidylcholine. FABP3-dependent membrane lipid remodeling causes ER stress via the PERK-eIF2α pathway and inhibits protein synthesis, limiting muscle recovery after immobilization. FABP3 knockdown induces a young-like lipid composition in aged muscles, reduces ER stress, and improves protein synthesis and muscle recovery. Further, FABP3 reduces membrane fluidity and knockdown increases fluidity in vitro, potentially causing ER stress. Therefore, FABP3 drives membrane lipid composition-mediated ER stress to regulate muscle homeostasis during aging and is a valuable target for sarcopenia. Ageing leads to a loss of muscle mass and strength, called sarcopenia. Here, the authors show that fatty acid binding protein 3 (FABP3), a lipid chaperone, drives age-dependent lipidome remodeling in skeletal muscle and deteriorates muscle mass and contractility by modulating membrane fluidity and ER stress signaling.
Collapse
|
38
|
Idoux R, Fuster C, Jacquemond V, Dayal A, Grabner M, Charnet P, Allard B. Divalent cations permeation in a Ca 2+ non-conducting skeletal muscle dihydropyridine receptor mouse model. Cell Calcium 2020; 91:102256. [PMID: 32866694 DOI: 10.1016/j.ceca.2020.102256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 11/22/2022]
Abstract
In response to excitation of skeletal muscle fibers, trains of action potentials induce changes in the configuration of the dihydropyridine receptor (DHPR) anchored in the tubular membrane which opens the Ca2+ release channel in the sarcoplasmic reticulum membrane. The DHPR also functions as a voltage-gated Ca2+ channel that conducts L-type Ca2+ currents routinely recorded in mammalian muscle fibers, which role was debated for more than four decades. Recently, to allow a closer look into the role of DHPR Ca2+ influx in mammalian muscle, a knock-in (ki) mouse model (ncDHPR) carrying mutation N617D (adjacent to domain II selectivity filter E) in the DHPRα1S subunit abolishing Ca2+ permeation through the channel was generated [Dayal et al., 2017]. In the present study, the Mn2+ quenching technique was initially intended to be used on voltage-clamped muscle fibers from this mouse to determine whether Ca2+ influx through a pathway distinct from DHPR may occur to compensate for the absence of DHPR Ca2+ influx. Surprisingly, while N617D DHPR muscle fibers of the ki mouse do not conduct Ca2+, Mn2+ entry and subsequent quenching did occur because Mn2+ was able to permeate and produce L-type currents through N617D DHPR. N617D DHPR was also found to conduct Ba2+ and Ba2+ currents were strongly blocked by external Ca2+. Ba2+ permeation was smaller, current kinetics slower and Ca2+ block more potent than in wild-type DHPR. These results indicate that residue N617 when replaced by the negatively charged residue D is suitably located at entrance of the pore to trap external Ca2+ impeding in this way permeation. Because Ba2+ binds with lower affinity to D, Ba2+ currents occur, but with reduced amplitudes as compared to Ba2+ currents through wild-type channels. We conclude that mutations located outside the selectivity filter influence channel permeation and possibly channel gating in a fully differentiated skeletal muscle environment.
Collapse
Affiliation(s)
- Romane Idoux
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| | - Clarisse Fuster
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| | - Vincent Jacquemond
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| | - Anamika Dayal
- Department of Genetics and Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1, A-6020, Innsbruck, Austria
| | - Manfred Grabner
- Department of Genetics and Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1, A-6020, Innsbruck, Austria
| | - Pierre Charnet
- Université de Montpellier, Institut des Biomolécules Max Mousseron, CNRS UMR-5247, Place Eugène Bataillon, 34090 Montpellier, France
| | - Bruno Allard
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France.
| |
Collapse
|
39
|
Hsu IU, Linsley JW, Reid LE, Hume RI, Leflein A, Kuwada JY. Dstac Regulates Excitation-Contraction Coupling in Drosophila Body Wall Muscles. Front Physiol 2020; 11:573723. [PMID: 33123029 PMCID: PMC7573238 DOI: 10.3389/fphys.2020.573723] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Stac3 regulates excitation-contraction coupling (EC coupling) in vertebrate skeletal muscles by regulating the L-type voltage-gated calcium channel (Cav channel). Recently a stac-like gene, Dstac, was identified in Drosophila and found to be expressed by both a subset of neurons and muscles. Here, we show that Dstac and Dmca1D, the Drosophila L-type Cav channel, are necessary for normal locomotion by larvae. Immunolabeling with specific antibodies against Dstac and Dmca1D found that Dstac and Dmca1D are expressed by larval body-wall muscles. Furthermore, Ca2+ imaging of muscles of Dstac and Dmca1D deficient larvae found that Dstac and Dmca1D are required for excitation-contraction coupling. Finally, Dstac appears to be required for normal expression levels of Dmca1D in body-wall muscles. These results suggest that Dstac regulates Dmca1D during EC coupling and thus muscle contraction.
Collapse
Affiliation(s)
- I-Uen Hsu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Jeremy W Linsley
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States.,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, United States
| | - Lilly E Reid
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Richard I Hume
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Ari Leflein
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - John Y Kuwada
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States.,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
40
|
Shin YJ, Kwon ES, Lee SM, Kim SK, Min KW, Lim JY, Lee B, Kang JS, Kwak JY, Son YH, Choi JY, Yang YR, Kim S, Kim YS, Jang HC, Suh Y, Yoon JH, Lee KP, Kwon KS. A subset of microRNAs in the Dlk1-Dio3 cluster regulates age-associated muscle atrophy by targeting Atrogin-1. J Cachexia Sarcopenia Muscle 2020; 11:1336-1350. [PMID: 32495509 PMCID: PMC7567143 DOI: 10.1002/jcsm.12578] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 03/16/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The microRNAs (miRNAs) down-regulated in aged mouse skeletal muscle were mainly clustered within the delta-like homologue 1 and the type III iodothyronine deiodinase (Dlk1-Dio3) genomic region. Although clustered miRNAs are coexpressed and regulate multiple targets in a specific signalling pathway, the function of miRNAs in the Dlk1-Dio3 cluster in muscle aging is largely unknown. We aimed to ascertain whether these miRNAs play a common role to regulate age-related muscle atrophy. METHODS To examine anti-atrophic effect of miRNAs, we individually transfected 42 miRNA mimics in fully differentiated myotubes and analysed their diameters. The luciferase reporter assay using target 3' untranslated region (UTR) and RNA pull-down assay were employed to ascertain the target predicted by the TargetScan algorithm. To investigate the therapeutic potential of the miRNAs in vivo, we generated adeno-associated virus (AAV) serotype 9 expressing green fluorescent protein (GFP) (AAV9-GFP) bearing miR-376c-3p and infected it into the tibialis anterior muscle of old mice. We performed morphometric analysis and measured ex vivo isometric force using a force transducer. Human gluteus maximus muscle tissues (ages ranging from 25 to 80 years) were used to investigate expression levels of the conserved miRNAs in the Dlk1-Dio3 cluster. RESULTS We found that the majority of miRNAs (33 out of 42 tested) in the cluster induced anti-atrophic phenotypes in fully differentiated myotubes with increasing their diameters. Eighteen of these miRNAs, eight of which are conserved in humans, harboured predicted binding sites in the 3' UTR of muscle atrophy gene-1 (Atrogin-1) encoding a muscle-specific E3 ligase. Direct interactions were identified between these miRNAs and the 3' UTR of Atrogin-1, leading to repression of Atrogin-1 and thereby induction of eIF3f protein content, in both human and mouse skeletal muscle cells. Intramuscular delivery of AAV9 expressing miR-376c-3p, one of the most effective miRNAs in myotube thickening, dramatically ameliorated skeletal muscle atrophy and improved muscle function, including isometric force, twitch force, and fatigue resistance in old mice. Consistent with our findings in mice, the expression of miRNAs in the cluster was significantly down-regulated in human muscle from individuals > 50 years old. CONCLUSIONS Our study suggests that genetic intervention using a muscle-directed miRNA delivery system has therapeutic efficacy in preventing Atrogin-1-mediated muscle atrophy in sarcopenia.
Collapse
Affiliation(s)
- Yeo Jin Shin
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Eun-Soo Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Seung-Min Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Seon-Kyu Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Kyung-Won Min
- Department of Biology, College of Natural Sciences, Gangneung-Wonju National University, Gangneung, Korea
| | - Jae-Young Lim
- Department of Rehabilitation Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea
| | - Bora Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea
| | - Jae Sook Kang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea
| | - Ju Yeon Kwak
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Young Hoon Son
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Jeong Yi Choi
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Yong Ryul Yang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Seokho Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, Korea
| | - Yeon-Soo Kim
- Department of New Drug Discovery and Development, Chungnam National University, Daejeon, Korea
| | - Hak C Jang
- Internal Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, NY, USA
| | - Je-Hyun Yoon
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Korea
| |
Collapse
|
41
|
Rufenach B, Christy D, Flucher BE, Bui JM, Gsponer J, Campiglio M, Van Petegem F. Multiple Sequence Variants in STAC3 Affect Interactions with CaV1.1 and Excitation-Contraction Coupling. Structure 2020; 28:922-932.e5. [DOI: 10.1016/j.str.2020.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/03/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
|
42
|
Zhao Y, Shen F, Gong M, Jin L, Ren X, Liu K, Lu J. Lifelong treadmill training improves muscle function detected by a modified grip strength test during aging in BALB/c mice. Life Sci 2020; 251:117603. [DOI: 10.1016/j.lfs.2020.117603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/15/2020] [Accepted: 03/24/2020] [Indexed: 12/31/2022]
|
43
|
Flucher BE. Skeletal muscle Ca V1.1 channelopathies. Pflugers Arch 2020; 472:739-754. [PMID: 32222817 PMCID: PMC7351834 DOI: 10.1007/s00424-020-02368-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/06/2020] [Accepted: 03/17/2020] [Indexed: 12/15/2022]
Abstract
CaV1.1 is specifically expressed in skeletal muscle where it functions as voltage sensor of skeletal muscle excitation-contraction (EC) coupling independently of its functions as L-type calcium channel. Consequently, all known CaV1.1-related diseases are muscle diseases and the molecular and cellular disease mechanisms relate to the dual functions of CaV1.1 in this tissue. To date, four types of muscle diseases are known that can be linked to mutations in the CACNA1S gene or to splicing defects. These are hypo- and normokalemic periodic paralysis, malignant hyperthermia susceptibility, CaV1.1-related myopathies, and myotonic dystrophy type 1. In addition, the CaV1.1 function in EC coupling is perturbed in Native American myopathy, arising from mutations in the CaV1.1-associated protein STAC3. Here, we first address general considerations concerning the possible roles of CaV1.1 in disease and then discuss the state of the art regarding the pathophysiology of the CaV1.1-related skeletal muscle diseases with an emphasis on molecular disease mechanisms.
Collapse
Affiliation(s)
- Bernhard E Flucher
- Department of Physiology and Medical Biophysics, Medical University Innsbruck, Schöpfstraße 41, A6020, Innsbruck, Austria.
| |
Collapse
|
44
|
Shishmarev D. Excitation-contraction coupling in skeletal muscle: recent progress and unanswered questions. Biophys Rev 2020; 12:143-153. [PMID: 31950344 DOI: 10.1007/s12551-020-00610-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Excitation-contraction coupling (ECC) is a physiological process that links excitation of muscles by the nervous system to their mechanical contraction. In skeletal muscle, ECC is initiated with an action potential, generated by the somatic nervous system, which causes a depolarisation of the muscle fibre membrane (sarcolemma). This leads to a rapid change in the transmembrane potential, which is detected by the voltage-gated Ca2+ channel dihydropyridine receptor (DHPR) embedded in the sarcolemma. DHPR transmits the contractile signal to another Ca2+ channel, ryanodine receptor (RyR1), embedded in the membrane of the sarcoplasmic reticulum (SR), which releases a large amount of Ca2+ ions from the SR that initiate muscle contraction. Despite the fundamental role of ECC in skeletal muscle function of all vertebrate species, the molecular mechanism underpinning the communication between the two key proteins involved in the process (DHPR and RyR1) is still largely unknown. The goal of this work is to review the recent progress in our understanding of ECC in skeletal muscle from the point of view of the structure and interactions of proteins involved in the process, and to highlight the unanswered questions in the field.
Collapse
Affiliation(s)
- Dmitry Shishmarev
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
45
|
Mackrill JJ, Shiels HA. Evolution of Excitation-Contraction Coupling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:281-320. [DOI: 10.1007/978-3-030-12457-1_12] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
46
|
Ca 2+ Channels Mediate Bidirectional Signaling between Sarcolemma and Sarcoplasmic Reticulum in Muscle Cells. Cells 2019; 9:cells9010055. [PMID: 31878335 PMCID: PMC7016941 DOI: 10.3390/cells9010055] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
The skeletal muscle and myocardial cells present highly specialized structures; for example, the close interaction between the sarcoplasmic reticulum (SR) and mitochondria—responsible for excitation-metabolism coupling—and the junction that connects the SR with T-tubules, critical for excitation-contraction (EC) coupling. The mechanisms that underlie EC coupling in these two cell types, however, are fundamentally distinct. They involve the differential expression of Ca2+ channel subtypes: CaV1.1 and RyR1 (skeletal), vs. CaV1.2 and RyR2 (cardiac). The CaV channels transform action potentials into elevations of cytosolic Ca2+, by activating RyRs and thus promoting SR Ca2+ release. The high levels of Ca2+, in turn, stimulate not only the contractile machinery but also the generation of mitochondrial reactive oxygen species (ROS). This forward signaling is reciprocally regulated by the following feedback mechanisms: Ca2+-dependent inactivation (of Ca2+ channels), the recruitment of Na+/Ca2+ exchanger activity, and oxidative changes in ion channels and transporters. Here, we summarize both well-established concepts and recent advances that have contributed to a better understanding of the molecular mechanisms involved in this bidirectional signaling.
Collapse
|
47
|
Kaplan MM, Sultana N, Benedetti A, Obermair GJ, Linde NF, Papadopoulos S, Dayal A, Grabner M, Flucher BE. Calcium Influx and Release Cooperatively Regulate AChR Patterning and Motor Axon Outgrowth during Neuromuscular Junction Formation. Cell Rep 2019; 23:3891-3904. [PMID: 29949772 DOI: 10.1016/j.celrep.2018.05.085] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/07/2018] [Accepted: 05/25/2018] [Indexed: 11/29/2022] Open
Abstract
Formation of synapses between motor neurons and muscles is initiated by clustering of acetylcholine receptors (AChRs) in the center of muscle fibers prior to nerve arrival. This AChR patterning is considered to be critically dependent on calcium influx through L-type channels (CaV1.1). Using a genetic approach in mice, we demonstrate here that either the L-type calcium currents (LTCCs) or sarcoplasmic reticulum (SR) calcium release is necessary and sufficient to regulate AChR clustering at the onset of neuromuscular junction (NMJ) development. The combined lack of both calcium signals results in loss of AChR patterning and excessive nerve branching. In the absence of SR calcium release, the severity of synapse formation defects inversely correlates with the magnitude of LTCCs. These findings highlight the importance of activity-dependent calcium signaling in early neuromuscular junction formation and indicate that both LTCC and SR calcium release individually support proper innervation of muscle by regulating AChR patterning and motor axon outgrowth.
Collapse
Affiliation(s)
- Mehmet Mahsum Kaplan
- Department of Physiology and Medical Physics, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Nasreen Sultana
- Department of Physiology and Medical Physics, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Ariane Benedetti
- Department of Physiology and Medical Physics, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Gerald J Obermair
- Department of Physiology and Medical Physics, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Nina F Linde
- Center of Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Symeon Papadopoulos
- Center of Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Anamika Dayal
- Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Manfred Grabner
- Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Bernhard E Flucher
- Department of Physiology and Medical Physics, Medical University Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
48
|
Kaplan MM, Flucher BE. Postsynaptic Ca V1.1-driven calcium signaling coordinates presynaptic differentiation at the developing neuromuscular junction. Sci Rep 2019; 9:18450. [PMID: 31804576 PMCID: PMC6895222 DOI: 10.1038/s41598-019-54900-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/21/2019] [Indexed: 11/08/2022] Open
Abstract
Proper formation of neuromuscular synapses requires the reciprocal communication between motor neurons and muscle cells. Several anterograde and retrograde signals involved in neuromuscular junction formation are known. However the postsynaptic mechanisms regulating presynaptic differentiation are still incompletely understood. Here we report that the skeletal muscle calcium channel (CaV1.1) is required for motor nerve differentiation and that the mechanism by which CaV1.1 controls presynaptic differentiation utilizes activity-dependent calcium signaling in muscle. In mice lacking CaV1.1 or CaV1.1-driven calcium signaling motor nerves are ectopically located and aberrantly defasciculated. Axons fail to recognize their postsynaptic target structures and synaptic vesicles and active zones fail to correctly accumulate at the nerve terminals opposite AChR clusters. These presynaptic defects are independent of aberrant AChR patterning and more sensitive to deficient calcium signals. Thus, our results identify CaV1.1-driven calcium signaling in muscle as a major regulator coordinating multiple aspects of presynaptic differentiation at the neuromuscular synapse.
Collapse
Affiliation(s)
- Mehmet Mahsum Kaplan
- Department of Physiology and Medical Physics, Medical University Innsbruck, 6020, Innsbruck, Austria
| | - Bernhard E Flucher
- Department of Physiology and Medical Physics, Medical University Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
49
|
El Ghaleb Y, Campiglio M, Flucher BE. Correcting the R165K substitution in the first voltage-sensor of Ca V1.1 right-shifts the voltage-dependence of skeletal muscle calcium channel activation. Channels (Austin) 2019; 13:62-71. [PMID: 30638110 PMCID: PMC6380215 DOI: 10.1080/19336950.2019.1568825] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 11/18/2022] Open
Abstract
The voltage-gated calcium channel CaV1.1a primarily functions as voltage-sensor in skeletal muscle excitation-contraction (EC) coupling. In embryonic muscle the splice variant CaV1.1e, which lacks exon 29, additionally function as a genuine L-type calcium channel. Because previous work in most laboratories used a CaV1.1 expression plasmid containing a single amino acid substitution (R165K) of a critical gating charge in the first voltage-sensing domain (VSD), we corrected this substitution and analyzed its effects on the gating properties of the L-type calcium currents in dysgenic myotubes. Reverting K165 to R right-shifted the voltage-dependence of activation by ~12 mV in both CaV1.1 splice variants without changing their current amplitudes or kinetics. This demonstrates the exquisite sensitivity of the voltage-sensor function to changes in the specific amino acid side chains independent of their charge. Our results further indicate the cooperativity of VSDs I and IV in determining the voltage-sensitivity of CaV1.1 channel gating.
Collapse
Affiliation(s)
- Yousra El Ghaleb
- Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | - Marta Campiglio
- Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| | - Bernhard E. Flucher
- Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
50
|
Traoré M, Gentil C, Benedetto C, Hogrel JY, De la Grange P, Cadot B, Benkhelifa-Ziyyat S, Julien L, Lemaitre M, Ferry A, Piétri-Rouxel F, Falcone S. An embryonic CaVβ1 isoform promotes muscle mass maintenance via GDF5 signaling in adult mouse. Sci Transl Med 2019; 11:11/517/eaaw1131. [DOI: 10.1126/scitranslmed.aaw1131] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 07/17/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022]
Abstract
Deciphering the mechanisms that govern skeletal muscle plasticity is essential to understand its pathophysiological processes, including age-related sarcopenia. The voltage-gated calcium channel CaV1.1 has a central role in excitation-contraction coupling (ECC), raising the possibility that it may also initiate the adaptive response to changes during muscle activity. Here, we revealed the existence of a gene transcription switch of the CaV1.1 β subunit (CaVβ1) that is dependent on the innervation state of the muscle in mice. In a mouse model of sciatic denervation, we showed increased expression of an embryonic isoform of the subunit that we called CaVβ1E. CaVβ1E boosts downstream growth differentiation factor 5 (GDF5) signaling to counteract muscle loss after denervation in mice. We further reported that aged mouse muscle expressed lower quantity of CaVβ1E compared with young muscle, displaying an altered GDF5-dependent response to denervation. Conversely, CaVβ1E overexpression improved mass wasting in aging muscle in mice by increasing GDF5 expression. We also identified the human CaVβ1E analogous and show a correlation between CaVβ1E expression in human muscles and age-related muscle mass decline. These results suggest that strategies targeting CaVβ1E or GDF5 might be effective in reducing muscle mass loss in aging.
Collapse
Affiliation(s)
| | - Christel Gentil
- Sorbonne Université, Centre de Recherche en Myologie, UM76, INSERM U974, Institut de Myologie, F-75013, Paris, France
| | - Chiara Benedetto
- Sorbonne Université, Centre de Recherche en Myologie, UM76, INSERM U974, Institut de Myologie, F-75013, Paris, France
| | - Jean-Yves Hogrel
- Institut de Myologie, GH Pitié-Salpêtrière, F-75013 Paris, France
| | | | - Bruno Cadot
- Sorbonne Université, Centre de Recherche en Myologie, UM76, INSERM U974, Institut de Myologie, F-75013, Paris, France
| | - Sofia Benkhelifa-Ziyyat
- Sorbonne Université, Centre de Recherche en Myologie, UM76, INSERM U974, Institut de Myologie, F-75013, Paris, France
| | - Laura Julien
- Sorbonne Université, Centre de Recherche en Myologie, UM76, INSERM U974, Institut de Myologie, F-75013, Paris, France
| | | | - Arnaud Ferry
- Sorbonne Université, Centre de Recherche en Myologie, UM76, INSERM U974, Institut de Myologie, F-75013, Paris, France
| | - France Piétri-Rouxel
- Sorbonne Université, Centre de Recherche en Myologie, UM76, INSERM U974, Institut de Myologie, F-75013, Paris, France
| | - Sestina Falcone
- Sorbonne Université, Centre de Recherche en Myologie, UM76, INSERM U974, Institut de Myologie, F-75013, Paris, France
| |
Collapse
|