1
|
Linsley JW, Reisine T, Finkbeiner S. Three dimensional and four dimensional live imaging to study mechanisms of progressive neurodegeneration. J Biol Chem 2024; 300:107433. [PMID: 38825007 PMCID: PMC11261153 DOI: 10.1016/j.jbc.2024.107433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Neurodegenerative diseases are complex and progressive, posing challenges to their study and understanding. Recent advances in microscopy imaging technologies have enabled the exploration of neurons in three spatial dimensions (3D) over time (4D). When applied to 3D cultures, tissues, or animals, these technologies can provide valuable insights into the dynamic and spatial nature of neurodegenerative diseases. This review focuses on the use of imaging techniques and neurodegenerative disease models to study neurodegeneration in 4D. Imaging techniques such as confocal microscopy, two-photon microscopy, miniscope imaging, light sheet microscopy, and robotic microscopy offer powerful tools to visualize and analyze neuronal changes over time in 3D tissue. Application of these technologies to in vitro models of neurodegeneration such as mouse organotypic culture systems and human organoid models provide versatile platforms to study neurodegeneration in a physiologically relevant context. Additionally, use of 4D imaging in vivo, including in mouse and zebrafish models of neurodegenerative diseases, allows for the investigation of early dysfunction and behavioral changes associated with neurodegeneration. We propose that these studies have the power to overcome the limitations of two-dimensional monolayer neuronal cultures and pave the way for improved understanding of the dynamics of neurodegenerative diseases and the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Jeremy W Linsley
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, California, USA; Operant Biopharma, San Francisco, California, USA
| | - Terry Reisine
- Independent Scientific Consultant, Santa Cruz, California, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, California, USA; Operant Biopharma, San Francisco, California, USA; Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, California, USA; Departments of Neurology and Physiology, University of California, San Francisco, California, USA; Neuroscience Graduate Program, University of California, San Francisco, California, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, California, USA.
| |
Collapse
|
2
|
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
|
3
|
Campbell L, Fredericks J, Mathivha K, Moshesh P, Coovadia A, Chirwa P, Dillon B, Ghoor A, Lawrence D, Nair L, Mabaso N, Mokwele D, Novellie M, Krause A, Carstens N. The implementation and utility of clinical exome sequencing in a South African infant cohort. Front Genet 2023; 14:1277948. [PMID: 38028619 PMCID: PMC10665497 DOI: 10.3389/fgene.2023.1277948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Genetic disorders are significant contributors to infant hospitalization and mortality globally. The early diagnosis of these conditions in infants remains a considerable challenge. Clinical exome sequencing (CES) has shown to be a successful tool for the early diagnosis of genetic conditions, however, its utility in African infant populations has not been investigated. The impact of the under-representation of African genomic data, the cost of testing, and genomic workforce shortages, need to be investigated and evidence-based implementation strategies accounting for locally available genetics expertise and diagnostic infrastructure need to be developed. We evaluated the diagnostic utility of singleton CES in a cohort of 32 ill, South African infants from two State hospitals in Johannesburg, South Africa. We analysed the data using a series of filtering approaches, including a curated virtual gene panel consisting of genes implicated in neonatal-and early childhood-onset conditions and genes with known founder and common variants in African populations. We reported a diagnostic yield of 22% and identified seven pathogenic variants in the NPHS1, COL2A1, OCRL, SHOC2, TPRV4, MTM1 and STAC3 genes. This study demonstrates the utility value of CES in the South African State healthcare setting, providing a diagnosis to patients who would otherwise not receive one and allowing for directed management. We anticipate an increase in the diagnostic yield of our workflow with further refinement of the study inclusion criteria. This study highlights important considerations for the implementation of genomic medicine in under-resourced settings and in under-represented African populations where variant interpretation remains a challenge.
Collapse
Affiliation(s)
- L. Campbell
- Division of Human Genetics, National Health Laboratory Service andSchool of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - J. Fredericks
- Department of Paediatrics and Child Health, School of Clinical Medicine, Rahima Moosa Mother and Child Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - K. Mathivha
- Department of Paediatrics and Child Health, School of Clinical Medicine, Nelson Mandela Children’s Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - P. Moshesh
- Department of Paediatrics and Child Health, School of Clinical Medicine, Nelson Mandela Children’s Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - A. Coovadia
- Department of Paediatrics and Child Health, School of Clinical Medicine, Rahima Moosa Mother and Child Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - P. Chirwa
- Nelson Mandela Children’s Hospital, Johannesburg, South Africa
| | - B. Dillon
- Division of Human Genetics, National Health Laboratory Service andSchool of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - A. Ghoor
- Department of Paediatrics and Child Health, School of Clinical Medicine, Rahima Moosa Mother and Child Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - D. Lawrence
- Department of Paediatrics and Child Health, School of Clinical Medicine, Rahima Moosa Mother and Child Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - L. Nair
- Department of Paediatrics and Child Health, School of Clinical Medicine, Rahima Moosa Mother and Child Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - N. Mabaso
- Division of Human Genetics, National Health Laboratory Service andSchool of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - D. Mokwele
- Division of Human Genetics, National Health Laboratory Service andSchool of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - M. Novellie
- Division of Human Genetics, National Health Laboratory Service andSchool of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - A. Krause
- Division of Human Genetics, National Health Laboratory Service andSchool of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - N. Carstens
- Division of Human Genetics, National Health Laboratory Service andSchool of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Genomics Platform, South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
4
|
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
|
5
|
García-Castañeda M, Michelucci A, Zhao N, Malik S, Dirksen RT. Postdevelopmental knockout of Orai1 improves muscle pathology in a mouse model of Duchenne muscular dystrophy. J Gen Physiol 2022; 154:213383. [PMID: 35939054 PMCID: PMC9365874 DOI: 10.1085/jgp.202213081] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), an X-linked disorder caused by loss-of-function mutations in the dystrophin gene, is characterized by progressive muscle degeneration and weakness. Enhanced store-operated Ca2+ entry (SOCE), a Ca2+ influx mechanism coordinated by STIM1 sensors of luminal Ca2+ within the sarcoplasmic reticulum (SR) and Ca2+-permeable Orai1 channels in the sarcolemma, is proposed to contribute to Ca2+-mediated muscle damage in DMD. To directly determine the impact of Orai1-dependent SOCE on the dystrophic phenotype, we crossed mdx mice with tamoxifen-inducible, muscle-specific Orai1 knockout mice (mdx-Orai1 KO mice). Both constitutive and SOCE were significantly increased in flexor digitorum brevis fibers from mdx mice, while SOCE was absent in fibers from both Orai1 KO and mdx-Orai1 KO mice. Compared with WT mice, fibers from mdx mice exhibited (1) increased resting myoplasmic Ca2+ levels, (2) reduced total releasable Ca2+ store content, and (3) a prolonged rate of electrically evoked Ca2+ transient decay. These effects were partially normalized in fibers from mdx-Orai1 KO mice. Intact extensor digitorum longus muscles from mdx mice exhibited a significant reduction of maximal specific force, which was rescued in muscles from mdx-Orai1 KO mice. Finally, during exposure to consecutive eccentric contractions, muscles from mdx mice displayed a more pronounced decline in specific force compared with that of WT mice, which was also significantly attenuated by Orai1 ablation. Together, these results indicate that enhanced Orai1-dependent SOCE exacerbates the dystrophic phenotype and that Orai1 deficiency improves muscle pathology by both normalizing Ca2+ homeostasis and promoting sarcolemmal integrity/stability.
Collapse
Affiliation(s)
- Maricela García-Castañeda
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Antonio Michelucci
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY,Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Nan Zhao
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Robert T. Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
6
|
Idoux R, Bretaud S, Berthier C, Ruggiero F, Jacquemond V, Allard B. Superfast excitation-contraction coupling in adult zebrafish skeletal muscle fibers. J Gen Physiol 2022; 154:213310. [PMID: 35767225 PMCID: PMC9247716 DOI: 10.1085/jgp.202213158] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/15/2022] [Indexed: 12/16/2022] Open
Abstract
The zebrafish has emerged as a very relevant animal model for probing the pathophysiology of human skeletal muscle disorders. This vertebrate animal model displays a startle response characterized by high-frequency swimming activity powered by contraction of fast skeletal muscle fibers excited at extremely high frequencies, critical for escaping predators and capturing prey. Such intense muscle performance requires extremely fast properties of the contractile machinery but also of excitation-contraction coupling, the process by which an action potential spreading along the sarcolemma induces a change in configuration of the dihydropyridine receptors, resulting in intramembrane charge movements, which in turn triggers the release of Ca2+ from the sarcoplasmic reticulum. However, thus far, the fastest Ca2+ transients evoked by vertebrate muscle fibers has been described in muscles used to produce sounds, such as those in the toadfish swim bladder, but not in muscles used for locomotion. By performing intracellular Ca2+ measurements under voltage control in isolated fast skeletal muscle fibers from adult zebrafish and mouse, we demonstrate that fish fast muscle fibers display superfast kinetics of action potentials, intramembrane charge movements, and action potential-evoked Ca2+ transient, allowing fusion and fused sustained Ca2+ transients at frequencies of excitation much higher than in mouse fast skeletal muscle fibers and comparable to those recorded in muscles producing sounds. The present study is the first demonstration of superfast kinetics of excitation-contraction coupling in skeletal muscle allowing superfast locomotor behaviors in a vertebrate.
Collapse
Affiliation(s)
- Romane Idoux
- Institut de Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Université de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR 5261, INSERM U1315, Faculté de Médecine Rockefeller, Lyon, France
| | - Sandrine Bretaud
- Institut de Génomique Fonctionnelle de Lyon (IGFL), École normale supérieure de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR 5242, Lyon, France
| | - Christine Berthier
- Institut de Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Université de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR 5261, INSERM U1315, Faculté de Médecine Rockefeller, Lyon, France
| | - Florence Ruggiero
- Institut de Génomique Fonctionnelle de Lyon (IGFL), École normale supérieure de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR 5242, Lyon, France
| | - Vincent Jacquemond
- Institut de Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Université de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR 5261, INSERM U1315, Faculté de Médecine Rockefeller, Lyon, France
| | - Bruno Allard
- Institut de Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Université de Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR 5261, INSERM U1315, Faculté de Médecine Rockefeller, Lyon, France,Correspondence to Bruno Allard:
| |
Collapse
|
7
|
Ashida Y, Himori K, Tamai K, Kimura I, Yamada T. Preconditioning contractions prevent prolonged force depression and Ca 2+-dependent proteolysis of STAC3 after damaging eccentric contractions. J Appl Physiol (1985) 2021; 131:1399-1407. [PMID: 34590910 DOI: 10.1152/japplphysiol.00463.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Preconditioning contractions (PCs) have been shown to markedly improve recovery from eccentric contractions (ECCs)-induced force depression. We here examined the mechanism behind the effects of PCs with focusing on the SH3 and cysteine-rich domain 3 (STAC3) that is essential for coupling membrane depolarization to Ca2+ release from the sarcoplasmic reticulum. Rat medial gastrocnemius (MG) muscles were excised immediately (REC0), 1 day (REC1), and 4 days (REC4) after exposure to 100 repeated damaging ECCs in vivo. PCs with 10 repeated nondamaging ECCs were applied 2 days before the damaging ECCs. Damaging ECCs induced in vivo isometric torque depression at 50 and 100 Hz stimulation frequencies, which was accompanied by a significant decrease in the amount of full-length STAC3, an activation of calpain 1, and an increased number of Evans Blue dye-positive fibers in MG muscles at REC1 and REC4. Interestingly, PCs attenuated all these deleterious alterations induced by damaging ECCs. Moreover, mechanistic experiments performed on normal muscle samples exposed to various concentration of Ca2+ showed a Ca2+-dependent proteolysis of STAC3, which was prevented by calpain inhibitor MDL-28170. In conclusion, PCs may improve recovery from force depression after damaging ECCs, in part by inhibiting the loss of STAC3 due to the increased permeability of cell membrane and subsequent activation of calpain 1.NEW & NOTEWORTHY The SH3 and cysteine-rich domain 3 (STAC3) is a skeletal muscle-specific protein that couples membrane depolarization to sarcoplasmic reticulum Ca2+ release. No studies, however, examined the role of STAC3 in protective effects of preconditioning contractions (PCs) against damaging eccentric contractions (ECCs). Here, we demonstrate that PCs may improve recovery from damaging ECCs-induced force depression, in part by an inhibition of Ca2+-dependent proteolysis of STAC3 due to increased membrane permeability and subsequent calpain 1 activation.
Collapse
Affiliation(s)
- Yuki Ashida
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan.,The Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Koichi Himori
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan.,The Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Katsuyuki Tamai
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Iori Kimura
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
8
|
Genetically encoded cell-death indicators (GEDI) to detect an early irreversible commitment to neurodegeneration. Nat Commun 2021; 12:5284. [PMID: 34489414 PMCID: PMC8421388 DOI: 10.1038/s41467-021-25549-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 08/16/2021] [Indexed: 01/07/2023] Open
Abstract
Cell death is a critical process that occurs normally in health and disease. However, its study is limited due to available technologies that only detect very late stages in the process or specific death mechanisms. Here, we report the development of a family of fluorescent biosensors called genetically encoded death indicators (GEDIs). GEDIs specifically detect an intracellular Ca2+ level that cells achieve early in the cell death process and that marks a stage at which cells are irreversibly committed to die. The time-resolved nature of a GEDI delineates a binary demarcation of cell life and death in real time, reformulating the definition of cell death. We demonstrate that GEDIs acutely and accurately report death of rodent and human neurons in vitro, and show that GEDIs enable an automated imaging platform for single cell detection of neuronal death in vivo in zebrafish larvae. With a quantitative pseudo-ratiometric signal, GEDIs facilitate high-throughput analysis of cell death in time-lapse imaging analysis, providing the necessary resolution and scale to identify early factors leading to cell death in studies of neurodegeneration. Cell death is a critical process in health and disease, yet available markers record later stages of cell death once a cell has already begun to decompose. Here the authors show the use of a genetically encoded calcium indicator that demarcates an irreversible stage of cell death earlier than previously possible.
Collapse
|
9
|
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: 87] [Impact Index Per Article: 29.0] [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
|
10
|
Rufenach B, Van Petegem F. Structure and function of STAC proteins: Calcium channel modulators and critical components of muscle excitation-contraction coupling. J Biol Chem 2021; 297:100874. [PMID: 34129875 PMCID: PMC8258685 DOI: 10.1016/j.jbc.2021.100874] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 12/26/2022] Open
Abstract
In skeletal muscle tissue, an intriguing mechanical coupling exists between two ion channels from different membranes: the L-type voltage-gated calcium channel (CaV1.1), located in the plasma membrane, and ryanodine receptor 1 (RyR1) located in the sarcoplasmic reticulum membrane. Excitable cells rely on Cavs to initiate Ca2+ entry in response to action potentials. RyRs can amplify this signal by releasing Ca2+ from internal stores. Although this process can be mediated through Ca2+ as a messenger, an overwhelming amount of evidence suggests that RyR1 has recruited CaV1.1 directly as its voltage sensor. The exact mechanisms that underlie this coupling have been enigmatic, but a recent wave of reports have illuminated the coupling protein STAC3 as a critical player. Without STAC3, the mechanical coupling between Cav1.1 and RyR1 is lost, and muscles fail to contract. Various sequence variants of this protein have been linked to congenital myopathy. Other STAC isoforms are expressed in the brain and may serve as regulators of L-type CaVs. Despite the short length of STACs, several points of contacts have been proposed between them and CaVs. However, it is currently unclear whether STAC3 also forms direct interactions with RyR1, and whether this modulates RyR1 function. In this review, we discuss the 3D architecture of STAC proteins, the biochemical evidence for their interactions, the relevance of these connections for functional modulation, and their involvement in myopathy.
Collapse
Affiliation(s)
- Britany Rufenach
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
11
|
Desaphy JF, Altamura C, Vicart S, Fontaine B. Targeted Therapies for Skeletal Muscle Ion Channelopathies: Systematic Review and Steps Towards Precision Medicine. J Neuromuscul Dis 2021; 8:357-381. [PMID: 33325393 PMCID: PMC8203248 DOI: 10.3233/jnd-200582] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Skeletal muscle ion channelopathies include non-dystrophic myotonias (NDM), periodic paralyses (PP), congenital myasthenic syndrome, and recently identified congenital myopathies. The treatment of these diseases is mainly symptomatic, aimed at reducing muscle excitability in NDM or modifying triggers of attacks in PP. OBJECTIVE This systematic review collected the evidences regarding effects of pharmacological treatment on muscle ion channelopathies, focusing on the possible link between treatments and genetic background. METHODS We searched databases for randomized clinical trials (RCT) and other human studies reporting pharmacological treatments. Preclinical studies were considered to gain further information regarding mutation-dependent drug effects. All steps were performed by two independent investigators, while two others critically reviewed the entire process. RESULTS For NMD, RCT showed therapeutic benefits of mexiletine and lamotrigine, while other human studies suggest some efficacy of various sodium channel blockers and of the carbonic anhydrase inhibitor (CAI) acetazolamide. Preclinical studies suggest that mutations may alter sensitivity of the channel to sodium channel blockers in vitro, which has been translated to humans in some cases. For hyperkalemic and hypokalemic PP, RCT showed efficacy of the CAI dichlorphenamide in preventing paralysis. However, hypokalemic PP patients carrying sodium channel mutations may have fewer benefits from CAI compared to those carrying calcium channel mutations. Few data are available for treatment of congenital myopathies. CONCLUSIONS These studies provided limited information about the response to treatments of individual mutations or groups of mutations. A major effort is needed to perform human studies for designing a mutation-driven precision medicine in muscle ion channelopathies.
Collapse
Affiliation(s)
- Jean-François Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Savine Vicart
- Sorbonne Université, INSERM, Assistance Publique Hôpitaux de Paris, Centre de Recherche en Myologie-UMR 974, Reference center in neuro-muscular channelopathies, Institute of Myology, Hôpital Universitaire Pitié-Salpêtrière, Paris, France
| | - Bertrand Fontaine
- Sorbonne Université, INSERM, Assistance Publique Hôpitaux de Paris, Centre de Recherche en Myologie-UMR 974, Reference center in neuro-muscular channelopathies, Institute of Myology, Hôpital Universitaire Pitié-Salpêtrière, Paris, France
| |
Collapse
|
12
|
Abstract
Neuropeptides are important for regulating numerous neural functions and behaviors. Release of neuropeptides requires long-lasting, high levels of cytosolic Ca2+ However, the molecular regulation of neuropeptide release remains to be clarified. Recently, Stac3 was identified as a key regulator of L-type Ca2+ channels (CaChs) and excitation-contraction coupling in vertebrate skeletal muscles. There is a small family of stac genes in vertebrates with other members expressed by subsets of neurons in the central nervous system. The function of neural Stac proteins, however, is poorly understood. Drosophila melanogaster contain a single stac gene, Dstac, which is expressed by muscles and a subset of neurons, including neuropeptide-expressing motor neurons. Here, genetic manipulations, coupled with immunolabeling, Ca2+ imaging, electrophysiology, and behavioral analysis, revealed that Dstac regulates L-type CaChs (Dmca1D) in Drosophila motor neurons and this, in turn, controls the release of neuropeptides.
Collapse
|
13
|
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: 0.8] [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
|
14
|
Shen C, Zhou Y, Tang C, He C, Zuo Z. Developmental exposure to mepanipyrim induces locomotor hyperactivity in zebrafish (Danio rerio) larvae. CHEMOSPHERE 2020; 256:127106. [PMID: 32447115 DOI: 10.1016/j.chemosphere.2020.127106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 06/11/2023]
Abstract
Mepanipyrim is a widely used fungicide, and residues of mepanipyrim are frequently detected in commodities. However, the neurotoxicity and underlying mechanisms of mepanipyrim are still insufficiently understood. In this study, zebrafish embryos at 0.5-1.0 post-fertilization hours (hpf) were exposed to 0.1, 1, 10 and 100 μg/L mepanipyrim for 7 days. Our results showed that mepanipyrim could cause the locomotor hyperactivity and increase the concentration of γ-amino butyric acid (GABA) and the Na+/K+- and Ca2+-ATPase activities in zebrafish larvae. We have conducted the RNA-sequence and RT-qPCR to analyze the gene expressions. The mRNA expression levels of calcium/sodium ion conduction associated genes were observably up-regulated, demonstrating that mepanipyrim could enhance the cell energy metabolism, the synaptic transmission and skeletal muscle contraction, which were consistent with the locomotor hyperactivity. Meanwhile, exposure to mepanipyrim could significantly change the gene expression levels of gad1, bdnf, nlgn1, and type A and B GABA receptors in zebrafish larvae. This is the first study focusing on the underlying mechanisms of the neurotoxic effects that are induced by mepanipyrim.
Collapse
Affiliation(s)
- Chao Shen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yixi Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chen Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian, 361005, China.
| |
Collapse
|
15
|
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: 2.3] [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]
|
16
|
Cote DR, Fuentes E, Elsayes AH, Ross JJ, Quraishi SA. A "crush" course on rhabdomyolysis: risk stratification and clinical management update for the perioperative clinician. J Anesth 2020; 34:585-598. [PMID: 32424487 DOI: 10.1007/s00540-020-02792-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 05/09/2020] [Indexed: 12/14/2022]
Abstract
Rhabdomyolysis, the release of myoglobin and other cellular breakdown products from necrotic muscle tissue, is seen in patients with crush injuries, drug overdose, malignant hyperthermia, muscular dystrophy, and with increasing frequency in obese patients undergoing routine procedures. For the perioperative clinician, managing the resultant shock, hyperkalemia, acidosis, and myoglobinuric acute kidney injury can present a significant challenge. Prompt recognition, hydration, and correction of metabolic disturbances may reduce or eliminate the need for long-term renal replacement therapy. This article reviews the pathophysiology and discusses key issues in the perioperative diagnosis, risk stratification, and management of rhabdomyolysis.
Collapse
Affiliation(s)
- Devan R Cote
- Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Eva Fuentes
- Department of Surgery, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Ali H Elsayes
- Department of Anesthesiology and Perioperative Medicine, Tufts Medical Center, Tufts University School of Medicine, 800 Washington Street, Boston, MA, 02111, USA
| | - Jonathan J Ross
- Department of Anesthesiology, Baystate Medical Center, Tufts University School of Medicine, Springfield, MA, USA
| | - Sadeq A Quraishi
- Department of Anesthesiology and Perioperative Medicine, Tufts Medical Center, Tufts University School of Medicine, 800 Washington Street, Boston, MA, 02111, USA.
| |
Collapse
|
17
|
Abstract
Sarcopenia - the accelerated age-related loss of muscle mass and function - is an under-diagnosed condition, and is central to deteriorating mobility, disability and frailty in older age. There is a lack of treatment options for older adults at risk of sarcopenia. Although sarcopenia's pathogenesis is multifactorial, its major phenotypes - muscle mass and muscle strength - are highly heritable. Several genome-wide association studies of muscle-related traits were published recently, providing dozens of candidate genes, many with unknown function. Therefore, animal models are required not only to identify causal mechanisms, but also to clarify the underlying biology and translate this knowledge into new interventions. Over the past several decades, small teleost fishes had emerged as powerful systems for modeling the genetics of human diseases. Owing to their amenability to rapid genetic intervention and the large number of conserved genetic and physiological features, small teleosts - such as zebrafish, medaka and killifish - have become indispensable for skeletal muscle genomic studies. The goal of this Review is to summarize evidence supporting the utility of small fish models for accelerating our understanding of human skeletal muscle in health and disease. We do this by providing a basic foundation of the (zebra)fish skeletal muscle morphology and physiology, and evidence of muscle-related gene homology. We also outline challenges in interpreting zebrafish mutant phenotypes and in translating them to human disease. Finally, we conclude with recommendations on future directions to leverage the large body of tools developed in small fish for the needs of genomic exploration in sarcopenia.
Collapse
Affiliation(s)
- Alon Daya
- The Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel
| | - Rajashekar Donaka
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 130010, Israel
| | - David Karasik
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 130010, Israel
- Hebrew SeniorLife, Hinda and Arthur Marcus Institute for Aging Research, Boston, MA 02131, USA
| |
Collapse
|
18
|
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: 26] [Impact Index Per Article: 6.5] [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
|
19
|
Abstract
The zebrafish (Danio rerio) has emerged as a widely used model system during the last four decades. The fact that the zebrafish larva is transparent enables sophisticated in vivo imaging, including calcium imaging of intracellular transients in many different tissues. While being a vertebrate, the reduced complexity of its nervous system and small size make it possible to follow large-scale activity in the whole brain. Its genome is sequenced and many genetic and molecular tools have been developed that simplify the study of gene function in health and disease. Since the mid 90's, the development and neuronal function of the embryonic, larval, and later, adult zebrafish have been studied using calcium imaging methods. This updated chapter is reviewing the advances in methods and research findings of zebrafish calcium imaging during the last decade. The choice of calcium indicator depends on the desired number of cells to study and cell accessibility. Synthetic calcium indicators, conjugated to dextrans and acetoxymethyl (AM) esters, are still used to label specific neuronal cell types in the hindbrain and the olfactory system. However, genetically encoded calcium indicators, such as aequorin and the GCaMP family of indicators, expressed in various tissues by the use of cell-specific promoters, are now the choice for most applications, including brain-wide imaging. Calcium imaging in the zebrafish has contributed greatly to our understanding of basic biological principles during development and adulthood, and the function of disease-related genes in a vertebrate system.
Collapse
|
20
|
Flucher BE, Campiglio M. STAC proteins: The missing link in skeletal muscle EC coupling and new regulators of calcium channel function. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2019; 1866:1101-1110. [PMID: 30543836 DOI: 10.1016/j.bbamcr.2018.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 01/19/2023]
Abstract
Excitation-contraction coupling is the signaling process by which action potentials control calcium release and consequently the force of muscle contraction. Until recently, three triad proteins were known to be essential for skeletal muscle EC coupling: the voltage-gated calcium channel CaV1.1 acting as voltage sensor, the SR calcium release channel RyR1 representing the only relevant calcium source, and the auxiliary CaV β1a subunit. Whether CaV1.1 and RyR1 are directly coupled or whether their interaction is mediated by another triad protein is still unknown. The recent identification of the adaptor protein STAC3 as fourth essential component of skeletal muscle EC coupling prompted vigorous research to reveal its role in this signaling process. Accumulating evidence supports its possible involvement in linking CaV1.1 and RyR1 in skeletal muscle EC coupling, but also indicates a second, much broader role of STAC proteins in the regulation of calcium/calmodulin-dependent feedback regulation of L-type calcium channels.
Collapse
Affiliation(s)
- Bernhard E Flucher
- Department of Physiology and Medical Physics, Medical University Innsbruck, Schöpfstraße 41, A6020 Innsbruck, Austria.
| | - Marta Campiglio
- Department of Physiology and Medical Physics, Medical University Innsbruck, Schöpfstraße 41, A6020 Innsbruck, Austria
| |
Collapse
|
21
|
Ríos E, Gillespie D, Franzini-Armstrong C. The binding interactions that maintain excitation-contraction coupling junctions in skeletal muscle. J Gen Physiol 2019; 151:593-605. [PMID: 30728215 PMCID: PMC6445584 DOI: 10.1085/jgp.201812268] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/02/2019] [Indexed: 12/03/2022] Open
Abstract
Calcium for contraction of skeletal muscles is released via tetrameric ryanodine receptor (RYR1) channels of the sarcoplasmic reticulum (SR), which are assembled in ordered arrays called couplons at junctions where the SR abuts T tubules or plasmalemma. Voltage-gated Ca2+ (CaV1.1) channels, found in tubules or plasmalemma, form symmetric complexes called CaV tetrads that associate with and activate underlying RYR tetramers during membrane depolarization by conveying a conformational change. Intriguingly, CaV tetrads regularly skip every other RYR tetramer within the array; therefore, the RYRs underlying tetrads (named V), but not the voltage sensor-lacking (C) RYRs, should be activated by depolarization. Here we hypothesize that the checkerboard association is maintained solely by reversible binary interactions between CaVs and RYRs and test this hypothesis using a quantitative model of the energies that govern CaV1.1-RYR1 binding, which are assumed to depend on number and location of bound CaVs. A Monte Carlo simulation generates large statistical samples and distributions of state variables that can be compared with quantitative features in freeze-fracture images of couplons from various sources. This analysis reveals two necessary model features: (1) the energy of a tetramer must have wells at low and high occupation by CaVs, so that CaVs positively cooperate in binding RYR (an allosteric effect), and (2) a large energy penalty results when two CaVs bind simultaneously to adjacent RYR protomers in adjacent tetramers (a steric clash). Under the hypothesis, V and C channels will eventually reverse roles. Role reversal justifies the presence of sensor-lacking C channels, as a structural and functional reserve for control of muscle contraction.
Collapse
Affiliation(s)
- Eduardo Ríos
- Section of Cellular Signaling, Department of Physiology and Biophysics, Rush University, Chicago, IL
| | - Dirk Gillespie
- Section of Cellular Signaling, Department of Physiology and Biophysics, Rush University, Chicago, IL
| | | |
Collapse
|
22
|
Abstract
Congenital myopathies (CM) are a genetically heterogeneous group of neuromuscular disorders most commonly presenting with neonatal/childhood-onset hypotonia and muscle weakness, a relatively static or slowly progressive disease course, and originally classified into subcategories based on characteristic histopathologic findings in muscle biopsies. This enduring concept of disease definition and classification based on the clinicopathologic phenotype was pioneered in the premolecular era. Advances in molecular genetics have brought into focus the increased blurring of the original seemingly "watertight" categories through broadening of the clinical phenotypes in existing genes, and continuous identification of novel genetic backgrounds. This review summarizes the histopathologic landscape of the 4 "classical" subtypes of CM-nemaline myopathies, core myopathies, centronuclear myopathies, and congenital fiber type disproportion and some of the emerging and novel genetic diseases with a CM presentation.
Collapse
Affiliation(s)
- Rahul Phadke
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children and Division of Neuropathology, National Hospital for Neurology and Neurosurgery, London, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
23
|
Zaharieva IT, Sarkozy A, Munot P, Manzur A, O'Grady G, Rendu J, Malfatti E, Amthor H, Servais L, Urtizberea JA, Neto OA, Zanoteli E, Donkervoort S, Taylor J, Dixon J, Poke G, Foley AR, Holmes C, Williams G, Holder M, Yum S, Medne L, Quijano-Roy S, Romero NB, Fauré J, Feng L, Bastaki L, Davis MR, Phadke R, Sewry CA, Bönnemann CG, Jungbluth H, Bachmann C, Treves S, Muntoni F. STAC3 variants cause a congenital myopathy with distinctive dysmorphic features and malignant hyperthermia susceptibility. Hum Mutat 2018; 39:1980-1994. [PMID: 30168660 DOI: 10.1002/humu.23635] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/27/2018] [Accepted: 08/28/2018] [Indexed: 12/14/2022]
Abstract
SH3 and cysteine-rich domain-containing protein 3 (STAC3) is an essential component of the skeletal muscle excitation-contraction coupling (ECC) machinery, though its role and function are not yet completely understood. Here, we report 18 patients carrying a homozygous p.(Trp284Ser) STAC3 variant in addition to a patient compound heterozygous for the p.(Trp284Ser) and a novel splice site change (c.997-1G > T). Clinical severity ranged from prenatal onset with severe features at birth, to a milder and slowly progressive congenital myopathy phenotype. A malignant hyperthermia (MH)-like reaction had occurred in several patients. The functional analysis demonstrated impaired ECC. In particular, KCl-induced membrane depolarization resulted in significantly reduced sarcoplasmic reticulum Ca2+ release. Co-immunoprecipitation of STAC3 with CaV 1.1 in patients and control muscle samples showed that the protein interaction between STAC3 and CaV 1.1 was not significantly affected by the STAC3 variants. This study demonstrates that STAC3 gene analysis should be included in the diagnostic work up of patients of any ethnicity presenting with congenital myopathy, in particular if a history of MH-like episodes is reported. While the precise pathomechanism remains to be elucidated, our functional characterization of STAC3 variants revealed that defective ECC is not a result of CaV 1.1 sarcolemma mislocalization or impaired STAC3-CaV 1.1 interaction.
Collapse
Affiliation(s)
- Irina T Zaharieva
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,Great Ormond Street Hospital, London, UK
| | - Pinki Munot
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,Great Ormond Street Hospital, London, UK
| | - Adnan Manzur
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,Great Ormond Street Hospital, London, UK
| | - Gina O'Grady
- Institute of Neuroscience and Muscle Research, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Paediatrics and Child Health Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - John Rendu
- UFR de Médecine, Centre Hospitalier Universitaire Grenoble Alpes, Université Grenoble Alpes, Grenoble, France
| | - Eduardo Malfatti
- Neuromuscular Morphology Unit and Neuromuscular Pathology Reference Center Paris-Est, Center for Research in Myology, Groupe Hospitalier Universitaire La Pitié-Salpêtrière, Paris, France
| | - Helge Amthor
- UFR des sciences de la santé, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France.,Service de Pédiatrie, Centre Hospitalier Universitaire Raymond Poincaré, Garches, France
| | | | - J Andoni Urtizberea
- Centre de Compétence Neuromusculaire, FILNEMUS, Hôpital Marin, Hendaye, France
| | - Osorio Abath Neto
- Departamento de Neurologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil.,Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Edmar Zanoteli
- Departamento de Neurologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Juliet Taylor
- Genetic Health Service New Zealand, Auckland, New Zealand
| | - Joanne Dixon
- Genetic Health Service New Zealand, Christchurch, New Zealand
| | - Gemma Poke
- Genetic Health Service New Zealand, Wellington, New Zealand
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Muriel Holder
- Department of Clinical Genetics, Guy's Hospital, London, UK
| | - Sabrina Yum
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Livija Medne
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Susana Quijano-Roy
- Service de Pédiatrie, Centre Hospitalier Universitaire Raymond Poincaré, Garches, France.,Centre de Référence Neuromusculaire GNMH, FILNEMUS, Université de Versailles, Versailles, France
| | - Norma B Romero
- Neuromuscular Morphology Unit and Neuromuscular Pathology Reference Center Paris-Est, Center for Research in Myology, Groupe Hospitalier Universitaire La Pitié-Salpêtrière, Paris, France
| | - Julien Fauré
- UFR de Médecine, Centre Hospitalier Universitaire Grenoble Alpes, Université Grenoble Alpes, Grenoble, France
| | - Lucy Feng
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Laila Bastaki
- Kuwait Medical Genetics Centre, Maternity Hospital, Kuwait City, Kuwait
| | - Mark R Davis
- Department of Diagnostic Genomics, Pathwest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Rahul Phadke
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,Great Ormond Street Hospital, London, UK
| | - Caroline A Sewry
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,Wolfson Centre for Inherited Neuromuscular Diseases, RJAH Orthopaedic Hospital, Oswestry, UK
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Heinz Jungbluth
- Randall Division for Cell and Molecular Biophysics, Muscle Signaling Section, King's College London, London, UK.,Department of Basic and Clinical Neuroscience, IoPPN, King's College London, London, UK.,Department of Anesthesia and Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Christoph Bachmann
- Department of Anesthesia and Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Susan Treves
- Department of Anesthesia and Biomedicine, University Hospital Basel, Basel, Switzerland.,Department of Life Sciences, University of Ferrara, Ferrara, Italy
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,Great Ormond Street Hospital, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| |
Collapse
|
24
|
Niu J, Dick IE, Yang W, Bamgboye MA, Yue DT, Tomaselli G, Inoue T, Ben-Johny M. Allosteric regulators selectively prevent Ca 2+-feedback of Ca V and Na V channels. eLife 2018; 7:35222. [PMID: 30198845 PMCID: PMC6156082 DOI: 10.7554/elife.35222] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 09/09/2018] [Indexed: 12/31/2022] Open
Abstract
Calmodulin (CaM) serves as a pervasive regulatory subunit of CaV1, CaV2, and NaV1 channels, exploiting a functionally conserved carboxy-tail element to afford dynamic Ca2+-feedback of cellular excitability in neurons and cardiomyocytes. Yet this modularity counters functional adaptability, as global changes in ambient CaM indiscriminately alter its targets. Here, we demonstrate that two structurally unrelated proteins, SH3 and cysteine-rich domain (stac) and fibroblast growth factor homologous factors (fhf) selectively diminish Ca2+/CaM-regulation of CaV1 and NaV1 families, respectively. The two proteins operate on allosteric sites within upstream portions of respective channel carboxy-tails, distinct from the CaM-binding interface. Generalizing this mechanism, insertion of a short RxxK binding motif into CaV1.3 carboxy-tail confers synthetic switching of CaM regulation by Mona SH3 domain. Overall, our findings identify a general class of auxiliary proteins that modify Ca2+/CaM signaling to individual targets allowing spatial and temporal orchestration of feedback, and outline strategies for engineering Ca2+/CaM signaling to individual targets.
Collapse
Affiliation(s)
- Jacqueline Niu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, United States
| | - Ivy E Dick
- Department of Physiology, University of Maryland, Baltimore, United States
| | - Wanjun Yang
- Department of Cardiology, Johns Hopkins University, Baltimore, United States
| | | | - David T Yue
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, United States
| | - Gordon Tomaselli
- Department of Cardiology, Johns Hopkins University, Baltimore, United States
| | - Takanari Inoue
- Department of Cell Biology, Johns Hopkins University, Baltimore, United States.,Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, United States
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, United States
| |
Collapse
|
25
|
Stac Proteins Suppress Ca 2+-Dependent Inactivation of Neuronal l-type Ca 2+ Channels. J Neurosci 2018; 38:9215-9227. [PMID: 30201773 DOI: 10.1523/jneurosci.0695-18.2018] [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: 03/15/2018] [Revised: 08/30/2018] [Accepted: 09/01/2018] [Indexed: 01/28/2023] Open
Abstract
Stac protein (named for its SH3- and cysteine-rich domains) was first identified in brain 20 years ago and is currently known to have three isoforms. Stac2, Stac1, and Stac3 transcripts are found at high, modest, and very low levels, respectively, in the cerebellum and forebrain, but their neuronal functions have been little investigated. Here, we tested the effects of Stac proteins on neuronal, high-voltage-activated Ca2+ channels. Overexpression of the three Stac isoforms eliminated Ca2+-dependent inactivation (CDI) of l-type current in rat neonatal hippocampal neurons (sex unknown), but not CDI of non-l-type current. Using heterologous expression in tsA201 cells (together with β and α2-δ1 auxiliary subunits), we found that CDI for CaV1.2 and CaV1.3 (the predominant, neuronal l-type Ca2+ channels) was suppressed by all three Stac isoforms, whereas CDI for the P/Q channel, CaV2.1, was not. For CaV1.2, the inhibition of CDI by the Stac proteins appeared to involve their direct interaction with the channel's C terminus. Within the Stac proteins, a weakly conserved segment containing ∼100 residues and linking the structurally conserved PKC C1 and SH3_1 domains was sufficient to fully suppress CDI. The presence of CDI for l-type current in control neonatal neurons raised the possibility that endogenous Stac levels are low in these neurons and Western blotting indicated that the expression of Stac2 was substantially increased in adult forebrain and cerebellum compared with neonate. Together, our results indicate that one likely function of neuronal Stac proteins is to tune Ca2+ entry via neuronal l-type channels.SIGNIFICANCE STATEMENT Stac protein, first identified 20 years ago in brain, has recently been found to be essential for proper trafficking and function of the skeletal muscle l-type Ca2+ channel and is the site of mutations causing a severe, inherited human myopathy. In neurons, however, functions for Stac protein have remained unexplored. Here, we report that one likely function of neuronal Stac proteins is tuning Ca2+ entry via l-type, but not that via non-l-type, Ca2+ channels. Moreover, there is a large postnatal increase in protein levels of the major neuronal isoform (Stac2) in forebrain and cerebellum, which could provide developmental regulation of l-type channel Ca2+ signaling in these brain regions.
Collapse
|
26
|
Niu J, Yang W, Yue DT, Inoue T, Ben-Johny M. Duplex signaling by CaM and Stac3 enhances Ca V1.1 function and provides insights into congenital myopathy. J Gen Physiol 2018; 150:1145-1161. [PMID: 29950399 PMCID: PMC6080896 DOI: 10.1085/jgp.201812005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/23/2018] [Accepted: 05/11/2018] [Indexed: 01/24/2023] Open
Abstract
CaV1.1 is essential for skeletal muscle excitation-contraction coupling. Its functional expression is tuned by numerous regulatory proteins, yet underlying modulatory mechanisms remain ambiguous as CaV1.1 fails to function in heterologous systems. In this study, by dissecting channel trafficking versus gating, we evaluated the requirements for functional CaV1.1 in heterologous systems. Although coexpression of the auxiliary β subunit is sufficient for surface-membrane localization, this baseline trafficking is weak, and channels elicit a diminished open probability. The regulatory proteins calmodulin and stac3 independently enhance channel trafficking and gating via their interaction with the CaV1.1 carboxy terminus. Myopathic stac3 mutations weaken channel binding and diminish trafficking. Our findings demonstrate that multiple regulatory proteins orchestrate CaV1.1 function via duplex mechanisms. Our work also furnishes insights into the pathophysiology of stac3-associated congenital myopathy and reveals novel avenues for pharmacological intervention.
Collapse
Affiliation(s)
- Jacqueline Niu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD
| | - Wanjun Yang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD
| | | | - Takanari Inoue
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD
- Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, MD
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY
| |
Collapse
|
27
|
Campiglio M, Kaplan MM, Flucher BE. STAC3 incorporation into skeletal muscle triads occurs independent of the dihydropyridine receptor. J Cell Physiol 2018; 233:9045-9051. [PMID: 30071129 PMCID: PMC6334165 DOI: 10.1002/jcp.26767] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/27/2018] [Indexed: 01/02/2023]
Abstract
Excitation‐contraction (EC) coupling in skeletal muscles operates through a physical interaction between the dihydropyridine receptor (DHPR), acting as a voltage sensor, and the ryanodine receptor (RyR1), acting as a calcium release channel. Recently, the adaptor protein SH3 and cysteine‐rich containing protein 3 (STAC3) has been identified as a myopathy disease gene and as an additional essential EC coupling component. STAC3 interacts with DHPR sequences including the critical EC coupling domain and has been proposed to function in linking the DHPR and RyR1. However, we and others demonstrated that incorporation of recombinant STAC3 into skeletal muscle triads critically depends only on the DHPR but not the RyR1. On the contrary, here, we provide evidence that endogenous STAC3 incorporates into triads in the absence of the DHPR in myotubes and muscle fibers of dysgenic mice. This finding demonstrates that STAC3 interacts with additional triad proteins and is consistent with its proposed role in directly or indirectly linking the DHPR with the RyR1.
Collapse
Affiliation(s)
- Marta Campiglio
- Department of Physiology, Medical University, Innsbruck, Innsbruck, Austria
| | - Mehmet M Kaplan
- Department of Physiology, Medical University, Innsbruck, Innsbruck, Austria
| | - Bernhard E Flucher
- Department of Physiology, Medical University, Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Linsley JW, Hsu IU, Wang W, Kuwada JY. Transport of the alpha subunit of the voltage gated L-type calcium channel through the sarcoplasmic reticulum occurs prior to localization to triads and requires the beta subunit but not Stac3 in skeletal muscles. Traffic 2018; 18:622-632. [PMID: 28697281 DOI: 10.1111/tra.12502] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 12/20/2022]
Abstract
Contraction of skeletal muscle is initiated by excitation-contraction (EC) coupling during which membrane voltage is transduced to intracellular Ca2+ release. EC coupling requires L-type voltage gated Ca2+ channels (the dihydropyridine receptor or DHPR) located at triads, which are junctions between the transverse (T) tubule and sarcoplasmic reticulum (SR) membranes, that sense membrane depolarization in the T tubule membrane. Reduced EC coupling is associated with ageing, and disruptions of EC coupling result in congenital myopathies for which there are few therapies. The precise localization of DHPRs to triads is critical for EC coupling, yet trafficking of the DHPR to triads is not well understood. Using dynamic imaging of zebrafish muscle fibers, we find that DHPR is transported along the longitudinal SR in a microtubule-independent mechanism. Furthermore, transport of DHPR in the SR membrane is differentially affected in null mutants of Stac3 or DHPRβ, two essential components of EC coupling. These findings reveal previously unappreciated features of DHPR motility within the SR prior to assembly at triads.
Collapse
Affiliation(s)
- Jeremy W Linsley
- Cell and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan.,Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - I-Uen Hsu
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Wenjia Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - John Y Kuwada
- Cell and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan.,Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
29
|
Hsu IU, Linsley JW, Varineau JE, Shafer OT, Kuwada JY. Dstac is required for normal circadian activity rhythms in Drosophila. Chronobiol Int 2018; 35:1016-1026. [PMID: 29621409 DOI: 10.1080/07420528.2018.1454937] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The genetic, molecular and neuronal mechanism underlying circadian activity rhythms is well characterized in the brain of Drosophila. The small ventrolateral neurons (s-LNVs) and pigment dispersing factor (PDF) expressed by them are especially important for regulating circadian locomotion. Here we describe a novel gene, Dstac, which is similar to the stac genes found in vertebrates that encode adaptor proteins, which bind and regulate L-type voltage-gated Ca2+ channels (CaChs). We show that Dstac is coexpressed with PDF by the s-LNVs and regulates circadian activity. Furthermore, the L-type CaCh, Dmca1D, appears to be expressed by the s-LNVs. Since vertebrate Stac3 regulates an L-type CaCh we hypothesize that Dstac regulates Dmca1D in s-LNVs and circadian activity.
Collapse
Affiliation(s)
- I-Uen Hsu
- a Department of Molecular, Cellular and Developmental Biology , University of Michigan , Ann Arbor , MI , USA
| | - Jeremy W Linsley
- b Cell and Molecular Biology Program , University of Michigan , Ann Arbor , MI , USA
| | - Jade E Varineau
- a Department of Molecular, Cellular and Developmental Biology , University of Michigan , Ann Arbor , MI , USA
| | - Orie T Shafer
- a Department of Molecular, Cellular and Developmental Biology , University of Michigan , Ann Arbor , MI , USA
| | - John Y Kuwada
- a Department of Molecular, Cellular and Developmental Biology , University of Michigan , Ann Arbor , MI , USA.,b Cell and Molecular Biology Program , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
30
|
STAC proteins associate to the IQ domain of Ca V1.2 and inhibit calcium-dependent inactivation. Proc Natl Acad Sci U S A 2018; 115:1376-1381. [PMID: 29363593 PMCID: PMC5819422 DOI: 10.1073/pnas.1715997115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Association of the adaptor protein STAC3 to the calcium channel was recently identified as essential for skeletal muscle contraction. While STAC3 is expressed exclusively in skeletal muscle, the other two isoforms, STAC1 and STAC2, are found in the brain, but their function has not yet been investigated. Recently we identified the C1 domain of STAC as crucial for its association with calcium channels. However, the corresponding binding domain remained unknown. Here, we demonstrate that STAC proteins associate with the C terminus of CaV1.2 at the IQ domain, and that this interaction results in the inhibition of calcium-dependent inactivation. Taken together, our data identify a functionally important STAC interaction domain and suggest that STAC proteins modulate calcium entry through CaV1 channels. The adaptor proteins STAC1, STAC2, and STAC3 represent a newly identified family of regulators of voltage-gated calcium channel (CaV) trafficking and function. The skeletal muscle isoform STAC3 is essential for excitation–contraction coupling and its mutation causes severe muscle disease. Recently, two distinct molecular domains in STAC3 were identified, necessary for its functional interaction with CaV1.1: the C1 domain, which recruits STAC proteins to the calcium channel complex in skeletal muscle triads, and the SH3-1 domain, involved in excitation–contraction coupling. These interaction sites are conserved in the three STAC proteins. However, the molecular domain in CaV1 channels interacting with the STAC C1 domain and the possible role of this interaction in neuronal CaV1 channels remained unknown. Using CaV1.2/2.1 chimeras expressed in dysgenic (CaV1.1−/−) myotubes, we identified the amino acids 1,641–1,668 in the C terminus of CaV1.2 as necessary for association of STAC proteins. This sequence contains the IQ domain and alanine mutagenesis revealed that the amino acids important for STAC association overlap with those making contacts with the C-lobe of calcium-calmodulin (Ca/CaM) and mediating calcium-dependent inactivation of CaV1.2. Indeed, patch-clamp analysis demonstrated that coexpression of either one of the three STAC proteins with CaV1.2 opposed calcium-dependent inactivation, although to different degrees, and that substitution of the CaV1.2 IQ domain with that of CaV2.1, which does not interact with STAC, abolished this effect. These results suggest that STAC proteins associate with the CaV1.2 C terminus at the IQ domain and thus inhibit calcium-dependent feedback regulation of CaV1.2 currents.
Collapse
|
31
|
Franzini-Armstrong C. The relationship between form and function throughout the history of excitation-contraction coupling. J Gen Physiol 2018; 150:189-210. [PMID: 29317466 PMCID: PMC5806676 DOI: 10.1085/jgp.201711889] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Franzini-Armstrong reviews the development of the excitation–contraction coupling field over time. The concept of excitation–contraction coupling is almost as old as Journal of General Physiology. It was understood as early as the 1940s that a series of stereotyped events is responsible for the rapid contraction response of muscle fibers to an initial electrical event at the surface. These early developments, now lost in what seems to be the far past for most young investigators, have provided an endless source of experimental approaches. In this Milestone in Physiology, I describe in detail the experiments and concepts that introduced and established the field of excitation–contraction coupling in skeletal muscle. More recent advances are presented in an abbreviated form, as readers are likely to be familiar with recent work in the field.
Collapse
Affiliation(s)
- Clara Franzini-Armstrong
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
32
|
Avey SR, Ojehomon M, Dawson JF, Gillis TE. How the expression of green fluorescent protein and human cardiac actin in the heart influences cardiac function and aerobic performance in zebrafish Danio rerio. JOURNAL OF FISH BIOLOGY 2018; 92:177-189. [PMID: 29194605 DOI: 10.1111/jfb.13507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 10/26/2017] [Indexed: 06/07/2023]
Abstract
The present study examined how the expression of enhanced green fluorescent protein (eGFP) and human cardiac actin (ACTC) in zebrafish Danio rerio influences embryonic heart rate (RH ) and the swim performance and metabolic rate of adult fish. Experiments with the adults involved determining the critical swimming speed (Ucrit , the highest speed sustainable and measure of aerobic capacity) while measuring oxygen consumption. Two different transgenic D. rerio lines were examined: one expressed eGFP in the heart (tg(cmlc:egfp)), while the second expressed ACTC in the heart and eGFP throughout the body (tg(cmlc:actc,ba:egfp)). It was found that RH was significantly lower in the tg(cmlc:actc,ba:egfp) embryos 4 days post-fertilization compared to wild-type (WT) and tg(cmlc:egfp). The swim experiments demonstrated that there was no significant difference in Ucrit between the transgenic lines and the wild-type fish, but metabolic rate and cost of transport (oxygen used to travel a set distance) was nearly two-fold higher in the tg(cmlc:actc,ba:egfp) fish compared to WT at their respective Ucrit . These results suggest that the expression of ACTC in the D. rerio heart and the expression of eGFP throughout the animal, alters cardiac function in the embryo and reduces the aerobic efficiency of the animal at high levels of activity.
Collapse
Affiliation(s)
- S R Avey
- Department of Integrative Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - M Ojehomon
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - J F Dawson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - T E Gillis
- Department of Integrative Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| |
Collapse
|
33
|
|
34
|
De novo reconstitution reveals the proteins required for skeletal muscle voltage-induced Ca 2+ release. Proc Natl Acad Sci U S A 2017; 114:13822-13827. [PMID: 29229815 DOI: 10.1073/pnas.1716461115] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Skeletal muscle contraction is triggered by Ca2+ release from the sarcoplasmic reticulum (SR) in response to plasma membrane (PM) excitation. In vertebrates, this depends on activation of the RyR1 Ca2+ pore in the SR, under control of conformational changes of CaV1.1, located ∼12 nm away in the PM. Over the last ∼30 y, gene knockouts have revealed that CaV1.1/RyR1 coupling requires additional proteins, but leave open the possibility that currently untested proteins are also necessary. Here, we demonstrate the reconstitution of conformational coupling in tsA201 cells by expression of CaV1.1, β1a, Stac3, RyR1, and junctophilin2. As in muscle, depolarization evokes Ca2+ transients independent of external Ca2+ entry and having amplitude with a saturating dependence on voltage. Moreover, freeze-fracture electron microscopy indicates that the five identified proteins are sufficient to establish physical links between CaV1.1 and RyR1. Thus, these proteins constitute the key elements essential for excitation-contraction coupling in skeletal muscle.
Collapse
|
35
|
Cassandrini D, Trovato R, Rubegni A, Lenzi S, Fiorillo C, Baldacci J, Minetti C, Astrea G, Bruno C, Santorelli FM. Congenital myopathies: clinical phenotypes and new diagnostic tools. Ital J Pediatr 2017; 43:101. [PMID: 29141652 PMCID: PMC5688763 DOI: 10.1186/s13052-017-0419-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/02/2017] [Indexed: 12/26/2022] Open
Abstract
Congenital myopathies are a group of genetic muscle disorders characterized clinically by hypotonia and weakness, usually from birth, and a static or slowly progressive clinical course. Historically, congenital myopathies have been classified on the basis of major morphological features seen on muscle biopsy. However, different genes have now been identified as associated with the various phenotypic and histological expressions of these disorders, and in recent years, because of their unexpectedly wide genetic and clinical heterogeneity, next-generation sequencing has increasingly been used for their diagnosis. We reviewed clinical and genetic forms of congenital myopathy and defined possible strategies to improve cost-effectiveness in histological and imaging diagnosis.
Collapse
Affiliation(s)
| | - Rosanna Trovato
- Molecular Medicine, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Anna Rubegni
- Molecular Medicine, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Sara Lenzi
- Neurology, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Chiara Fiorillo
- Molecular Medicine, IRCCS Fondazione Stella Maris, Pisa, Italy.,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genoa, Italy
| | - Jacopo Baldacci
- Molecular Medicine, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Carlo Minetti
- Unit of Pediatric Neurology and Muscular Disorders, Istituto G. Gaslini, Genoa, Italy.,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genoa, Italy
| | - Guja Astrea
- Neurology, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Claudio Bruno
- Department of Neuroscience, Center of Myology and Neurodegenerative Disorders, Istituto G. Gaslini, Genoa, Italy
| | | | | |
Collapse
|
36
|
Structural insights into binding of STAC proteins to voltage-gated calcium channels. Proc Natl Acad Sci U S A 2017; 114:E9520-E9528. [PMID: 29078335 PMCID: PMC5692558 DOI: 10.1073/pnas.1708852114] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Excitation-contraction (EC) coupling in skeletal muscle requires functional and mechanical coupling between L-type voltage-gated calcium channels (CaV1.1) and the ryanodine receptor (RyR1). Recently, STAC3 was identified as an essential protein for EC coupling and is part of a group of three proteins that can bind and modulate L-type voltage-gated calcium channels. Here, we report crystal structures of tandem-SH3 domains of different STAC isoforms up to 1.2-Å resolution. These form a rigid interaction through a conserved interdomain interface. We identify the linker connecting transmembrane repeats II and III in two different CaV isoforms as a binding site for the SH3 domains and report a crystal structure of the complex with the STAC2 isoform. The interaction site includes the location for a disease variant in STAC3 that has been linked to Native American myopathy (NAM). Introducing the mutation does not cause misfolding of the SH3 domains, but abolishes the interaction. Disruption of the interaction via mutations in the II-III loop perturbs skeletal muscle EC coupling, but preserves the ability of STAC3 to slow down inactivation of CaV1.2.
Collapse
|