1
|
Serano M, Perni S, Pierantozzi E, Laurino A, Sorrentino V, Rossi D. Intracellular Membrane Contact Sites in Skeletal Muscle Cells. MEMBRANES 2025; 15:29. [PMID: 39852269 PMCID: PMC11767089 DOI: 10.3390/membranes15010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025]
Abstract
Intracellular organelles are common to eukaryotic cells and provide physical support for the assembly of specialized compartments. In skeletal muscle fibers, the largest intracellular organelle is the sarcoplasmic reticulum, a specialized form of the endoplasmic reticulum primarily devoted to Ca2+ storage and release for muscle contraction. Occupying about 10% of the total cell volume, the sarcoplasmic reticulum forms multiple membrane contact sites, some of which are unique to skeletal muscle. These contact sites primarily involve the plasma membrane; among these, specialized membrane contact sites between the transverse tubules and the terminal cisternae of the sarcoplasmic reticulum form triads. Triads are skeletal muscle-specific contact sites where Ca2+ channels and regulatory proteins assemble to form the so-called calcium release complex. Additionally, the sarcoplasmic reticulum contacts mitochondria to enable a more precise regulation of Ca2+ homeostasis and energy metabolism. The sarcoplasmic reticulum and the plasma membrane also undergo dynamic remodeling to allow Ca2+ entry from the extracellular space and replenish the stores. This process involves the formation of dynamic membrane contact sites called Ca2+ Entry Units. This review explores the key processes in biogenesis and assembly of intracellular membrane contact sites as well as the membrane remodeling that occurs in response to muscle fatigue.
Collapse
Affiliation(s)
- Matteo Serano
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Stefano Perni
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Annunziatina Laurino
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
- Program of Molecular Diagnosis of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
- Program of Molecular Diagnosis of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| |
Collapse
|
2
|
Gomaa AAE, Zeid AMA, Nagy IM, Zahran AM. The effect of genetic polymorphisms in STIM1 and ORAI1 on erythropoietin resistance in Egyptian patients with end-stage renal disease. Clin Chim Acta 2025; 564:119948. [PMID: 39214396 DOI: 10.1016/j.cca.2024.119948] [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: 08/02/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Chronic renal failure (CRF) is an incurable disease with unique challenges. Anemia is a frequent complication affecting dialysis patients. Erythropoietin (EPO) is used to treat anemia, but a poor response may result. We investigated genetic polymorphisms of store-operated calcium channel (SOC) signaling, an important erythropoietin-activated pathway that may induce EPO resistance in patients with renal failure. A total of 108 end stage renal disease (ESRD) patients were selected for this study. Patients were divided into two groups according to their erythropoietin resistance index (ERI): 39 patients with an ERI>10 and 69 patients with an ERI<10. We selected four tagging single nucleotide polymorphisms (tSNPs) in STIM1 and five in ORAI1 in our study. A polymerase chain reaction was performed, and genotyping against EPO resistance was correlated. Patients with the AG genotype of rs1561876 in STIM1, the TC genotype of rs6486795 in ORAI1, and the TG or GG genotypes of rs12320939 in ORAI1 were associated with an increased risk of erythropoietin resistance. Overall, we reported a moderately significant relationship between genetic polymorphisms of STIM1 and EPO resistance. We also reported a highly significant relationship between genetic polymorphisms of ORAI1 and EPO resistance. The (A-A-G) haplotype of STIM1 and the (G-T-G-T-A, G-C-G-C-G, or G-T-T-C-G) haplotypes of ORAI1 were significantly associated with EPO resistance.
Collapse
Affiliation(s)
- Azza A E Gomaa
- Internal Medicine Department, Menofia University, Menofia, Egypt.
| | - Amany M A Zeid
- Clinical Pathology Department, Menofia University, Menofia, Egypt
| | - Ibrahim M Nagy
- Medicinal Chemistry Department, Menofia University, Menofia, Egypt.
| | - Ahmed M Zahran
- Internal Medicine Department, Menofia University, Menofia, Egypt
| |
Collapse
|
3
|
Carreras-Sureda A, Zhang X, Laubry L, Brunetti J, Koenig S, Wang X, Castelbou C, Hetz C, Liu Y, Frieden M, Demaurex N. The ER stress sensor IRE1 interacts with STIM1 to promote store-operated calcium entry, T cell activation, and muscular differentiation. Cell Rep 2023; 42:113540. [PMID: 38060449 DOI: 10.1016/j.celrep.2023.113540] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/29/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
Store-operated Ca2+ entry (SOCE) mediated by stromal interacting molecule (STIM)-gated ORAI channels at endoplasmic reticulum (ER) and plasma membrane (PM) contact sites maintains adequate levels of Ca2+ within the ER lumen during Ca2+ signaling. Disruption of ER Ca2+ homeostasis activates the unfolded protein response (UPR) to restore proteostasis. Here, we report that the UPR transducer inositol-requiring enzyme 1 (IRE1) interacts with STIM1, promotes ER-PM contact sites, and enhances SOCE. IRE1 deficiency reduces T cell activation and human myoblast differentiation. In turn, STIM1 deficiency reduces IRE1 signaling after store depletion. Using a CaMPARI2-based Ca2+ genome-wide screen, we identify CAMKG2 and slc105a as SOCE enhancers during ER stress. Our findings unveil a direct crosstalk between SOCE and UPR via IRE1, acting as key regulator of ER Ca2+ and proteostasis in T cells and muscles. Under ER stress, this IRE1-STIM1 axis boosts SOCE to preserve immune cell functions, a pathway that could be targeted for cancer immunotherapy.
Collapse
Affiliation(s)
- Amado Carreras-Sureda
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
| | - Xin Zhang
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Loann Laubry
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Jessica Brunetti
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Stéphane Koenig
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Xiaoxia Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cyril Castelbou
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience (GERO), Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
4
|
Carrion SA, Michal JJ, Jiang Z. Alternative Transcripts Diversify Genome Function for Phenome Relevance to Health and Diseases. Genes (Basel) 2023; 14:2051. [PMID: 38002994 PMCID: PMC10671453 DOI: 10.3390/genes14112051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Manipulation using alternative exon splicing (AES), alternative transcription start (ATS), and alternative polyadenylation (APA) sites are key to transcript diversity underlying health and disease. All three are pervasive in organisms, present in at least 50% of human protein-coding genes. In fact, ATS and APA site use has the highest impact on protein identity, with their ability to alter which first and last exons are utilized as well as impacting stability and translation efficiency. These RNA variants have been shown to be highly specific, both in tissue type and stage, with demonstrated importance to cell proliferation, differentiation and the transition from fetal to adult cells. While alternative exon splicing has a limited effect on protein identity, its ubiquity highlights the importance of these minor alterations, which can alter other features such as localization. The three processes are also highly interwoven, with overlapping, complementary, and competing factors, RNA polymerase II and its CTD (C-terminal domain) chief among them. Their role in development means dysregulation leads to a wide variety of disorders and cancers, with some forms of disease disproportionately affected by specific mechanisms (AES, ATS, or APA). Challenges associated with the genome-wide profiling of RNA variants and their potential solutions are also discussed in this review.
Collapse
Affiliation(s)
| | | | - Zhihua Jiang
- Department of Animal Sciences and Center for Reproductive Biology, Washington State University, Pullman, WA 99164-7620, USA; (S.A.C.); (J.J.M.)
| |
Collapse
|
5
|
Gorza L, Germinario E, Vitadello M, Guerra I, De Majo F, Gasparella F, Caliceti P, Vitiello L, Danieli-Betto D. Curcumin Administration Improves Force of mdx Dystrophic Diaphragm by Acting on Fiber-Type Composition, Myosin Nitrotyrosination and SERCA1 Protein Levels. Antioxidants (Basel) 2023; 12:1181. [PMID: 37371910 DOI: 10.3390/antiox12061181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
The vegetal polyphenol curcumin displays beneficial effects against skeletal muscle derangement induced by oxidative stress, disuse or aging. Since oxidative stress and inflammation are involved in the progression of muscle dystrophy, the effects of curcumin administration were investigated in the diaphragm of mdx mice injected intraperitoneally or subcutaneously with curcumin for 4-12-24 weeks. Curcumin treatment independently of the way and duration of administration (i) ameliorated myofiber maturation index without affecting myofiber necrosis, inflammation and degree of fibrosis; (ii) counteracted the decrease in type 2X and 2B fiber percentage; (iii) increased about 30% both twitch and tetanic tensions of diaphragm strips; (iv) reduced myosin nitrotyrosination and tropomyosin oxidation; (v) acted on two opposite nNOS regulators by decreasing active AMP-Kinase and increasing SERCA1 protein levels, the latter effect being detectable also in myotube cultures from mdx satellite cells. Interestingly, increased contractility, decreased myosin nitrotyrosination and SERCA1 upregulation were also detectable in the mdx diaphragm after a 4-week administration of the NOS inhibitor 7-Nitroindazole, and were not improved further by a combined treatment. In conclusion, curcumin has beneficial effects on the dystrophic muscle, mechanistically acting for the containment of a deregulated nNOS activity.
Collapse
Affiliation(s)
- Luisa Gorza
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Elena Germinario
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Maurizio Vitadello
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Irene Guerra
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Federica De Majo
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | | | - Paolo Caliceti
- Department of Pharmaceutical Sciences, University of Padova, 35131 Padova, Italy
| | - Libero Vitiello
- Department of Biology, University of Padova, 35131 Padova, Italy
| | | |
Collapse
|
6
|
Protasi F, Girolami B, Roccabianca S, Rossi D. Store-operated calcium entry: From physiology to tubular aggregate myopathy. Curr Opin Pharmacol 2023; 68:102347. [PMID: 36608411 DOI: 10.1016/j.coph.2022.102347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 01/06/2023]
Abstract
Store-Operated Ca2+ entry (SOCE) is recognized as a key mechanism in muscle physiology necessary to refill intracellular Ca2+ stores during sustained muscle activity. For many years the cell structures expected to mediate SOCE in skeletal muscle fibres remained unknown. Recently, the identification of Ca2+ Entry Units (CEUs) in exercised muscle fibres opened new insights into the role of extracellular Ca2+ in muscle contraction and, more generally, in intracellular Ca2+ homeostasis. Accordingly, intracellular Ca2+ unbalance due to alterations in SOCE strictly correlates with muscle disfunction and disease. Mutations in proteins involved in SOCE (STIM1, ORAI1, and CASQ1) have been linked to tubular aggregate myopathy (TAM), a disease that causes muscle weakness and myalgia and is characterized by a typical accumulation of highly ordered and packed membrane tubules originated from the sarcoplasmic reticulum (SR). Achieving a full understanding of the molecular pathways activated by alterations in Ca2+ entry mechanisms is a necessary step to design effective therapies for human SOCE-related disorders.
Collapse
Affiliation(s)
- Feliciano Protasi
- CAST, Center for Advanced Studies and Technology; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy; DMSI, Department of Medicine and Aging Sciences; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy
| | - Barbara Girolami
- CAST, Center for Advanced Studies and Technology; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy; DMSI, Department of Medicine and Aging Sciences; University G. d'Annunzio of Chieti-Pescara, I-66100, Italy
| | - Sara Roccabianca
- DMMS, Department of Molecular and Developmental Medicine; University of Siena, I-53100, Siena Italy
| | - Daniela Rossi
- DMMS, Department of Molecular and Developmental Medicine; University of Siena, I-53100, Siena Italy.
| |
Collapse
|
7
|
Dowling P, Gargan S, Swandulla D, Ohlendieck K. Proteomic profiling of impaired excitation-contraction coupling and abnormal calcium handling in muscular dystrophy. Proteomics 2022; 22:e2200003. [PMID: 35902360 PMCID: PMC10078611 DOI: 10.1002/pmic.202200003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/06/2022]
Abstract
The X-linked inherited neuromuscular disorder Duchenne muscular dystrophy is characterised by primary abnormalities in the membrane cytoskeletal component dystrophin. The almost complete absence of the Dp427-M isoform of dystrophin in skeletal muscles renders contractile fibres more susceptible to progressive degeneration and a leaky sarcolemma membrane. This in turn results in abnormal calcium homeostasis, enhanced proteolysis and impaired excitation-contraction coupling. Biochemical and mass spectrometry-based proteomic studies of both patient biopsy specimens and genetic animal models of dystrophinopathy have demonstrated significant changes in the concentration and/or physiological function of essential calcium-regulatory proteins in dystrophin-lacking voluntary muscles. Abnormalities include dystrophinopathy-associated changes in voltage sensing receptors, calcium release channels, calcium pumps and calcium binding proteins. This review article provides an overview of the importance of the sarcolemmal dystrophin-glycoprotein complex and the wider dystrophin complexome in skeletal muscle and its linkage to depolarisation-induced calcium-release mechanisms and the excitation-contraction-relaxation cycle. Besides chronic inflammation, fat substitution and reactive myofibrosis, a major pathobiochemical hallmark of X-linked muscular dystrophy is represented by the chronic influx of calcium ions through the damaged plasmalemma in conjunction with abnormal intracellular calcium fluxes and buffering. Impaired calcium handling proteins should therefore be included in an improved biomarker signature of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | | | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
8
|
Márquez-Nogueras KM, Vuchkovska V, DiNello E, Osorio-Valencia S, Kuo IY. Polycystin-2 (PC2) is a key determinant of in vitro myogenesis. Am J Physiol Cell Physiol 2022; 323:C333-C346. [PMID: 35675637 PMCID: PMC9291421 DOI: 10.1152/ajpcell.00159.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The development of skeletal muscle (myogenesis) is a well-orchestrated process where myoblasts withdraw from the cell cycle and differentiate into myotubes. Signaling by fluxes in intracellular calcium (Ca2+) is known to contribute to myogenesis, and increased mitochondrial biogenesis is required to meet the metabolic demand of mature myotubes. However, gaps remain in the understanding of how intracellular Ca2+ signals can govern myogenesis. Polycystin-2 (PC2 or TRPP1) is a nonselective cation channel permeable to Ca2+. It can interact with intracellular calcium channels to control Ca2+ release and concurrently modulates mitochondrial function and remodeling. Due to these features, we hypothesized that PC2 is a central protein in mediating both the intracellular Ca2+ responses and mitochondrial changes seen in myogenesis. To test this hypothesis, we created CRISPR/Cas9 knockout (KO) C2C12 murine myoblast cell lines. PC2 KO cells were unable to differentiate into myotubes, had impaired spontaneous Ca2+ oscillations, and did not develop depolarization-evoked Ca2+ transients. The autophagic-associated pathway beclin-1 was downregulated in PC2 KO cells, and direct activation of the autophagic pathway resulted in decreased mitochondrial remodeling. Re-expression of full-length PC2, but not a calcium channel dead pathologic mutant, restored the differentiation phenotype and increased the expression of mitochondrial proteins. Our results establish that PC2 is a novel regulator of in vitro myogenesis by integrating PC2-dependent Ca2+ signals and metabolic pathways.
Collapse
Affiliation(s)
| | | | - Elisabeth DiNello
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois
| | - Sara Osorio-Valencia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois
| | - Ivana Y Kuo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois
| |
Collapse
|
9
|
Servello D, Abdinghoff J, Grissmer A, Tschernig T. Transient receptor potential channel 6 in human skeletal muscle fibers: Investigation in fresh and conserved tissue samples. Biomed Rep 2022; 17:60. [PMID: 35719837 PMCID: PMC9198990 DOI: 10.3892/br.2022.1543] [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: 01/28/2022] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
Transient receptor potential channel 6 (TRPC6) channels constitute non-selective cation channels that are localized in the plasmalemma or sarcolemma, and have a leading permeability for the bivalent calcium ion. Animal models indicate an involvement of TRPC6 in malignant hyperthermia. The expression of TRPC6 in the sarcolemma has been demonstrated in the skeletal muscle fibers of mice. The importance of TRPC6 channels for the influx of calcium into the muscle cell has also been established. The presence of TRPC6 in tissues of human skeletal muscle is surmised. In order to confirm the presence of TRPC6 in human skeletal muscle, tissue samples of various skeletal muscles (Musculus deltoideus, pectoralis major, trizeps brachii and rectus femoris) from eight different human donors (n=8; six preserved cadavers and two non-preserved cadavers) were examined using immunohistochemistry. TRPC6 was found in all muscle fibers of all investigated bodies. Appropriate controls yielded the expected results. As demonstrated in animal studies and in studies of human cells, the presented results confirmed the presence of TRPC6 in human skeletal muscle tissue. Thus, TRPC6 is most likely important for calcium homeostasis and the proper function of human muscle fibers and may be a target for treatment in various muscular diseases.
Collapse
Affiliation(s)
- Davide Servello
- Institute of Anatomy and Cell Biology, Saarland University, Faculty of Medicine, D‑66424 Homburg, Saar, Germany
| | - Jan Abdinghoff
- Institute of Anatomy and Cell Biology, Saarland University, Faculty of Medicine, D‑66424 Homburg, Saar, Germany
| | - Alexander Grissmer
- Institute of Anatomy and Cell Biology, Saarland University, Faculty of Medicine, D‑66424 Homburg, Saar, Germany
| | - Thomas Tschernig
- Institute of Anatomy and Cell Biology, Saarland University, Faculty of Medicine, D‑66424 Homburg, Saar, Germany
| |
Collapse
|
10
|
Henry C, Carreras-Sureda A, Demaurex N. Enforced tethering elongates the cortical endoplasmic reticulum and limits store-operated calcium entry. J Cell Sci 2022; 135:274483. [PMID: 35191477 PMCID: PMC8995094 DOI: 10.1242/jcs.259313] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/14/2022] [Indexed: 12/03/2022] Open
Abstract
Recruitment of STIM proteins to cortical endoplasmic reticulum (cER) domains forming membrane contact sites (MCSs) mediate the store-operated Ca2+ entry (SOCE) pathway essential for human immunity. The cER is dynamically regulated by STIM and tethering proteins during SOCE, but the ultrastructural rearrangement and functional consequences of cER remodeling are unknown. Here, we express natural (E-Syt1 and E-Syt2) and artificial (MAPPER-S and MAPPER-L) protein tethers in HEK-293T cells and correlate the changes in cER length and gap distance, as measured by electron microscopy, with ionic fluxes. We found that native cER cisternae extended during store depletion and remained elongated at a constant ER-plasma membrane (PM) gap distance during subsequent Ca2+ elevations. Tethering proteins enhanced store-dependent cER expansion, anchoring the enlarged cER at tether-specific gap distances of 12-15 nm (E-Syts) and 5-9 nm (MAPPERs). Cells with artificially extended cER had reduced SOCE and reduced agonist-induced Ca2+ release. SOCE remained modulated by calmodulin and exhibited enhanced Ca2+-dependent inhibition. We propose that cER expansion mediated by ER-PM tethering at a close distance negatively regulates SOCE by confining STIM-ORAI complexes to the periphery of enlarged cER sheets, a process that might participate in the termination of store-operated Ca2+ entry. Summary: ER-PM tethering at close distance limits Ca2+ entry by confining STIM-ORAI complexes to the periphery of contact sites.
Collapse
Affiliation(s)
- Christopher Henry
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| | - Amado Carreras-Sureda
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| |
Collapse
|
11
|
Acute RyR1 Ca 2+ leak enhances NADH-linked mitochondrial respiratory capacity. Nat Commun 2021; 12:7219. [PMID: 34893614 PMCID: PMC8664928 DOI: 10.1038/s41467-021-27422-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/18/2021] [Indexed: 12/25/2022] Open
Abstract
Sustained ryanodine receptor (RyR) Ca2+ leak is associated with pathological conditions such as heart failure or skeletal muscle weakness. We report that a single session of sprint interval training (SIT), but not of moderate intensity continuous training (MICT), triggers RyR1 protein oxidation and nitrosylation leading to calstabin1 dissociation in healthy human muscle and in in vitro SIT models (simulated SIT or S-SIT). This is accompanied by decreased sarcoplasmic reticulum Ca2+ content, increased levels of mitochondrial oxidative phosphorylation proteins, supercomplex formation and enhanced NADH-linked mitochondrial respiratory capacity. Mechanistically, (S-)SIT increases mitochondrial Ca2+ uptake in mouse myotubes and muscle fibres, and decreases pyruvate dehydrogenase phosphorylation in human muscle and mouse myotubes. Countering Ca2+ leak or preventing mitochondrial Ca2+ uptake blunts S-SIT-induced adaptations, a result supported by proteomic analyses. Here we show that triggering acute transient Ca2+ leak through RyR1 in healthy muscle may contribute to the multiple health promoting benefits of exercise.
Collapse
|
12
|
Uchimura T, Sakurai H. Orai1-STIM1 Regulates Increased Ca 2+ Mobilization, Leading to Contractile Duchenne Muscular Dystrophy Phenotypes in Patient-Derived Induced Pluripotent Stem Cells. Biomedicines 2021; 9:biomedicines9111589. [PMID: 34829817 PMCID: PMC8615222 DOI: 10.3390/biomedicines9111589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022] Open
Abstract
Ca2+ overload is one of the factors leading to Duchenne muscular dystrophy (DMD) pathogenesis. However, the molecular targets of dystrophin deficiency-dependent Ca2+ overload and the correlation between Ca2+ overload and contractile DMD phenotypes in in vitro human models remain largely elusive. In this study, we utilized DMD patient-derived induced pluripotent stem cells (iPSCs) to differentiate myotubes using doxycycline-inducible MyoD overexpression, and searched for a target molecule that mediates dystrophin deficiency-dependent Ca2+ overload using commercially available chemicals and siRNAs. We found that several store-operated Ca2+ channel (SOC) inhibitors effectively prevented Ca2+ overload and identified that STIM1–Orai1 is a molecular target of SOCs. These findings were further confirmed by demonstrating that STIM1–Orai1 inhibitors, CM4620, AnCoA4, and GSK797A, prevented Ca2+ overload in dystrophic myotubes. Finally, we evaluated CM4620, AnCoA4, and GSK7975A activities using a previously reported model recapitulating a muscle fatigue-like decline in contractile performance in DMD. All three chemicals ameliorated the decline in contractile performance, indicating that modulating STIM1–Orai1-mediated Ca2+ overload is effective in rescuing contractile phenotypes. In conclusion, SOCs are major contributors to dystrophin deficiency-dependent Ca2+ overload through STIM1–Orai1 as molecular mediators. Modulating STIM1–Orai1 activity was effective in ameliorating the decline in contractile performance in DMD.
Collapse
Affiliation(s)
- Tomoya Uchimura
- Center for iPSC Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Takeda-CiRA Joint Program, Fujisawa 251-8555, Japan
- Correspondence: (T.U.); (H.S.)
| | - Hidetoshi Sakurai
- Center for iPSC Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
- Takeda-CiRA Joint Program, Fujisawa 251-8555, Japan
- Correspondence: (T.U.); (H.S.)
| |
Collapse
|
13
|
Conte E, Imbrici P, Mantuano P, Coppola MA, Camerino GM, De Luca A, Liantonio A. Alteration of STIM1/Orai1-Mediated SOCE in Skeletal Muscle: Impact in Genetic Muscle Diseases and Beyond. Cells 2021; 10:2722. [PMID: 34685702 PMCID: PMC8534495 DOI: 10.3390/cells10102722] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 02/08/2023] Open
Abstract
Intracellular Ca2+ ions represent a signaling mediator that plays a critical role in regulating different muscular cellular processes. Ca2+ homeostasis preservation is essential for maintaining skeletal muscle structure and function. Store-operated Ca2+ entry (SOCE), a Ca2+-entry process activated by depletion of intracellular stores contributing to the regulation of various function in many cell types, is pivotal to ensure a proper Ca2+ homeostasis in muscle fibers. It is coordinated by STIM1, the main Ca2+ sensor located in the sarcoplasmic reticulum, and ORAI1 protein, a Ca2+-permeable channel located on transverse tubules. It is commonly accepted that Ca2+ entry via SOCE has the crucial role in short- and long-term muscle function, regulating and adapting many cellular processes including muscle contractility, postnatal development, myofiber phenotype and plasticity. Lack or mutations of STIM1 and/or Orai1 and the consequent SOCE alteration have been associated with serious consequences for muscle function. Importantly, evidence suggests that SOCE alteration can trigger a change of intracellular Ca2+ signaling in skeletal muscle, participating in the pathogenesis of different progressive muscle diseases such as tubular aggregate myopathy, muscular dystrophy, cachexia, and sarcopenia. This review provides a brief overview of the molecular mechanisms underlying STIM1/Orai1-dependent SOCE in skeletal muscle, focusing on how SOCE alteration could contribute to skeletal muscle wasting disorders and on how SOCE components could represent pharmacological targets with high therapeutic potential.
Collapse
Affiliation(s)
- Elena Conte
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (P.I.); (P.M.); (M.A.C.); (G.M.C.); (A.D.L.)
| | | | | | | | | | | | - Antonella Liantonio
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (P.I.); (P.M.); (M.A.C.); (G.M.C.); (A.D.L.)
| |
Collapse
|
14
|
Yuan R, Zhang J, Wang Y, Zhu X, Hu S, Zeng J, Liang F, Tang Q, Chen Y, Chen L, Zhu W, Li M, Mo D. Reorganization of chromatin architecture during prenatal development of porcine skeletal muscle. DNA Res 2021; 28:6261936. [PMID: 34009337 PMCID: PMC8154859 DOI: 10.1093/dnares/dsab003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/26/2021] [Indexed: 11/18/2022] Open
Abstract
Myofibres (primary and secondary myofibre) are the basic structure of muscle and the determinant of muscle mass. To explore the skeletal muscle developmental processes from primary myofibres to secondary myofibres in pigs, we conducted an integrative three-dimensional structure of genome and transcriptomic characterization of longissimus dorsi muscle of pig from primary myofibre formation stage [embryonic Day 35 (E35)] to secondary myofibre formation stage (E80). In the hierarchical genomic structure, we found that 11.43% of genome switched compartment A/B status, 14.53% of topologically associating domains are changed intradomain interactions (D-scores) and 2,730 genes with differential promoter–enhancer interactions and (or) enhancer activity from E35 to E80. The alterations of genome architecture were found to correlate with expression of genes that play significant roles in neuromuscular junction, embryonic morphogenesis, skeletal muscle development or metabolism, typically, NEFL, MuSK, SLN, Mef2D and GCK. Significantly, Sox6 and MATN2 play important roles in the process of primary to secondary myofibres formation and increase the regulatory potential score and genes expression in it. In brief, we reveal the genomic reorganization from E35 to E80 and construct genome-wide high-resolution interaction maps that provide a resource for studying long-range control of gene expression from E35 to E80.
Collapse
Affiliation(s)
- Renqiang Yuan
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.,Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaman Zhang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yujie Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xingxing Zhu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Silu Hu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jianhua Zeng
- Guangdong YIHAO Food Co., Ltd, Guangzhou 510620, China
| | - Feng Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Luxi Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.,Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wei Zhu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
15
|
Michelucci A, Liang C, Protasi F, Dirksen RT. Altered Ca 2+ Handling and Oxidative Stress Underlie Mitochondrial Damage and Skeletal Muscle Dysfunction in Aging and Disease. Metabolites 2021; 11:metabo11070424. [PMID: 34203260 PMCID: PMC8304741 DOI: 10.3390/metabo11070424] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/26/2022] Open
Abstract
Skeletal muscle contraction relies on both high-fidelity calcium (Ca2+) signals and robust capacity for adenosine triphosphate (ATP) generation. Ca2+ release units (CRUs) are highly organized junctions between the terminal cisternae of the sarcoplasmic reticulum (SR) and the transverse tubule (T-tubule). CRUs provide the structural framework for rapid elevations in myoplasmic Ca2+ during excitation-contraction (EC) coupling, the process whereby depolarization of the T-tubule membrane triggers SR Ca2+ release through ryanodine receptor-1 (RyR1) channels. Under conditions of local or global depletion of SR Ca2+ stores, store-operated Ca2+ entry (SOCE) provides an additional source of Ca2+ that originates from the extracellular space. In addition to Ca2+, skeletal muscle also requires ATP to both produce force and to replenish SR Ca2+ stores. Mitochondria are the principal intracellular organelles responsible for ATP production via aerobic respiration. This review provides a broad overview of the literature supporting a role for impaired Ca2+ handling, dysfunctional Ca2+-dependent production of reactive oxygen/nitrogen species (ROS/RNS), and structural/functional alterations in CRUs and mitochondria in the loss of muscle mass, reduction in muscle contractility, and increase in muscle damage in sarcopenia and a wide range of muscle disorders including muscular dystrophy, rhabdomyolysis, central core disease, and disuse atrophy. Understanding the impact of these processes on normal muscle function will provide important insights into potential therapeutic targets designed to prevent or reverse muscle dysfunction during aging and disease.
Collapse
Affiliation(s)
- Antonio Michelucci
- DNICS, Department of Neuroscience, Imaging, and Clinical Sciences, University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
- Correspondence:
| | - Chen Liang
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA; (C.L.); (R.T.D.)
| | - 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, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA; (C.L.); (R.T.D.)
| |
Collapse
|
16
|
Conte E, Pannunzio A, Imbrici P, Camerino GM, Maggi L, Mora M, Gibertini S, Cappellari O, De Luca A, Coluccia M, Liantonio A. Gain-of-Function STIM1 L96V Mutation Causes Myogenesis Alteration in Muscle Cells From a Patient Affected by Tubular Aggregate Myopathy. Front Cell Dev Biol 2021; 9:635063. [PMID: 33718371 PMCID: PMC7952532 DOI: 10.3389/fcell.2021.635063] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Tubular Aggregate Myopathy (TAM) is a hereditary ultra-rare muscle disorder characterized by muscle weakness and cramps or myasthenic features. Biopsies from TAM patients show the presence of tubular aggregates originated from sarcoplasmic reticulum due to altered Ca2+ homeostasis. TAM is caused by gain-of-function mutations in STIM1 or ORAI1, proteins responsible for Store-Operated-Calcium-Entry (SOCE), a pivotal mechanism in Ca2+ signaling. So far there is no cure for TAM and the mechanisms through which STIM1 or ORAI1 gene mutation lead to muscle dysfunction remain to be clarified. It has been established that post-natal myogenesis critically relies on Ca2+ influx through SOCE. To explore how Ca2+ homeostasis dysregulation associated with TAM impacts on muscle differentiation cascade, we here performed a functional characterization of myoblasts and myotubes deriving from patients carrying STIM1 L96V mutation by using fura-2 cytofluorimetry, high content imaging and real-time PCR. We demonstrated a higher resting Ca2+ concentration and an increased SOCE in STIM1 mutant compared with control, together with a compensatory down-regulation of genes involved in Ca2+ handling (RyR1, Atp2a1, Trpc1). Differentiating STIM1 L96V myoblasts persisted in a mononuclear state and the fewer multinucleated myotubes had distinct morphology and geometry of mitochondrial network compared to controls, indicating a defect in the late differentiation phase. The alteration in myogenic pathway was confirmed by gene expression analysis regarding early (Myf5, Mef2D) and late (DMD, Tnnt3) differentiation markers together with mitochondrial markers (IDH3A, OGDH). We provided evidences of mechanisms responsible for a defective myogenesis associated to TAM mutant and validated a reliable cellular model usefull for TAM preclinical studies.
Collapse
Affiliation(s)
- Elena Conte
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | | | - Paola Imbrici
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | | | - Lorenzo Maggi
- Neuromuscular Diseases and Neuroimmunology Unit, Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Marina Mora
- Neuromuscular Diseases and Neuroimmunology Unit, Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Sara Gibertini
- Neuromuscular Diseases and Neuroimmunology Unit, Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | | | - Annamaria De Luca
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Mauro Coluccia
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | | |
Collapse
|
17
|
Zhang I, Hu H. Store-Operated Calcium Channels in Physiological and Pathological States of the Nervous System. Front Cell Neurosci 2020; 14:600758. [PMID: 33328896 PMCID: PMC7732603 DOI: 10.3389/fncel.2020.600758] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Store-operated calcium channels (SOCs) are widely expressed in excitatory and non-excitatory cells where they mediate significant store-operated calcium entry (SOCE), an important pathway for calcium signaling throughout the body. While the activity of SOCs has been well studied in non-excitable cells, attention has turned to their role in neurons and glia in recent years. In particular, the role of SOCs in the nervous system has been extensively investigated, with links to their dysregulation found in a wide variety of neurological diseases from Alzheimer’s disease (AD) to pain. In this review, we provide an overview of their molecular components, expression, and physiological role in the nervous system and describe how the dysregulation of those roles could potentially lead to various neurological disorders. Although further studies are still needed to understand how SOCs are activated under physiological conditions and how they are linked to pathological states, growing evidence indicates that SOCs are important players in neurological disorders and could be potential new targets for therapies. While the role of SOCE in the nervous system continues to be multifaceted and controversial, the study of SOCs provides a potentially fruitful avenue into better understanding the nervous system and its pathologies.
Collapse
Affiliation(s)
- Isis Zhang
- Department of Anesthesiology, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Huijuan Hu
- Department of Anesthesiology, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
18
|
Fine Tuning of Calcium Constitutive Entry by Optogenetically-Controlled Membrane Polarization: Impact on Cell Migration. Cells 2020; 9:cells9071684. [PMID: 32668787 PMCID: PMC7408270 DOI: 10.3390/cells9071684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 11/16/2022] Open
Abstract
Anomalies in constitutive calcium entry (CCE) have been commonly attributed to cell dysfunction in pathological conditions such as cancer. Calcium influxes of this type rely on channels, such as transient receptor potential (TRP) channels, to be constitutively opened and strongly depend on membrane potential and a calcium driving force. We developed an optogenetic approach based on the expression of the halorhodopsin chloride pump to study CCE in non-excitable cells. Using C2C12 cells, we found that halorhodopsin can be used to achieve a finely tuned control of membrane polarization. Escalating the membrane polarization by incremental changes in light led to a concomitant increase in CCE through transient receptor potential vanilloid 2 (TRPV2) channels. Moreover, light-induced calcium entry through TRPV2 channels promoted cell migration. Our study shows for the first time that by modulating CCE and related physiological responses, such as cell motility, halorhodopsin serves as a potentially powerful tool that could open new avenues for the study of CCE and associated cellular behaviors.
Collapse
|
19
|
Choi JH, Jeong SY, Oh MR, Allen PD, Lee EH. TRPCs: Influential Mediators in Skeletal Muscle. Cells 2020; 9:cells9040850. [PMID: 32244622 PMCID: PMC7226745 DOI: 10.3390/cells9040850] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023] Open
Abstract
Ca2+ itself or Ca2+-dependent signaling pathways play fundamental roles in various cellular processes from cell growth to death. The most representative example can be found in skeletal muscle cells where a well-timed and adequate supply of Ca2+ is required for coordinated Ca2+-dependent skeletal muscle functions, such as the interactions of contractile proteins during contraction. Intracellular Ca2+ movements between the cytosol and sarcoplasmic reticulum (SR) are strictly regulated to maintain the appropriate Ca2+ supply in skeletal muscle cells. Added to intracellular Ca2+ movements, the contribution of extracellular Ca2+ entry to skeletal muscle functions and its significance have been continuously studied since the early 1990s. Here, studies on the roles of channel proteins that mediate extracellular Ca2+ entry into skeletal muscle cells using skeletal myoblasts, myotubes, fibers, tissue, or skeletal muscle-originated cell lines are reviewed with special attention to the proposed functions of transient receptor potential canonical proteins (TRPCs) as store-operated Ca2+ entry (SOCE) channels under normal conditions and the potential abnormal properties of TRPCs in muscle diseases such as Duchenne muscular dystrophy (DMD).
Collapse
Affiliation(s)
- Jun Hee Choi
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Seung Yeon Jeong
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Mi Ri Oh
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Paul D. Allen
- Leeds Institute of Biomedical & Clinical Sciences, St. James’s University Hospital, University of Leeds, Leeds LS97TF, UK
| | - Eun Hui Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-7279
| |
Collapse
|
20
|
Avila-Medina J, Mayoral-González I, Galeano-Otero I, Redondo PC, Rosado JA, Smani T. Pathophysiological Significance of Store-Operated Calcium Entry in Cardiovascular and Skeletal Muscle Disorders and Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:489-504. [PMID: 31646522 DOI: 10.1007/978-3-030-12457-1_19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Store-Operated Ca2+ Entry (SOCE) is an important Ca2+ influx pathway expressed by several excitable and non-excitable cell types. SOCE is recognized as relevant signaling pathway not only for physiological process, but also for its involvement in different pathologies. In fact, independent studies demonstrated the implication of essential protein regulating SOCE, such as STIM, Orai and TRPCs, in different pathogenesis and cell disorders, including cardiovascular disease, muscular dystrophies and angiogenesis. Compelling evidence showed that dysregulation in the function and/or expression of isoforms of STIM, Orai or TRPC play pivotal roles in cardiac hypertrophy and heart failure, vascular remodeling and hypertension, skeletal myopathies, and angiogenesis. In this chapter, we summarized the current knowledge concerning the mechanisms underlying abnormal SOCE and its involvement in some diseases, as well as, we discussed the significance of STIM, Orai and TRPC isoforms as possible therapeutic targets for the treatment of angiogenesis, cardiovascular and skeletal muscle diseases.
Collapse
Affiliation(s)
- Javier Avila-Medina
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain
| | - Isabel Mayoral-González
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain
- Department of Surgery, University of Seville, Sevilla, Spain
| | - Isabel Galeano-Otero
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain
| | - Pedro C Redondo
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Juan A Rosado
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain.
- Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, Spain.
- CIBERCV, Madrid, Spain.
| |
Collapse
|
21
|
Cacheux M, Strauss B, Raad N, Ilkan Z, Hu J, Benard L, Feske S, Hulot JS, Akar FG. Cardiomyocyte-Specific STIM1 (Stromal Interaction Molecule 1) Depletion in the Adult Heart Promotes the Development of Arrhythmogenic Discordant Alternans. Circ Arrhythm Electrophysiol 2019; 12:e007382. [PMID: 31726860 PMCID: PMC6867678 DOI: 10.1161/circep.119.007382] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND STIM1 (stromal interaction molecule 1) is a calcium (Ca2+) sensor that regulates cardiac hypertrophy by triggering store-operated Ca2+ entry. Because STIM1 binding to phospholamban increases sarcoplasmic reticulum Ca2+ load independent of store-operated Ca2+ entry, we hypothesized that it controls electrophysiological function and arrhythmias in the adult heart. METHODS Inducible myocyte-restricted STIM1-KD (STIM1 knockdown) was achieved in adult mice using an αMHC (α-myosin heavy chain)-MerCreMer system. Mechanical and electrophysiological properties were examined using echocardiography in vivo and optical action potential (AP) mapping ex vivo in tamoxifen-induced STIM1flox/flox-Cretg/- (STIM1-KD) and littermate controls for STIM1flox/flox (referred to as STIM1-Ctl) and for Cretg/- without STIM deletion (referred to as Cre-Ctl). RESULTS STIM1-KD mice (N=23) exhibited poor survival compared with STIM1-Ctl (N=22) and Cre-Ctl (N=11) with >50% mortality after only 8-days of cardiomyocyte-restricted STIM1-KD. STIM1-KD but not STIM1-Ctl or Cre-Ctl hearts exhibited a proclivity for arrhythmic behavior, ranging from frequent ectopy to pacing-induced ventricular tachycardia/ventricular fibrillation (VT/VF). Examination of the electrophysiological substrate revealed decreased conduction velocity and increased AP duration (APD) heterogeneity in STIM1-KD. These features, however, were comparable in VT/VF(+) and VT/VF(-) hearts. We also uncovered a marked increase in the magnitude of APD alternans during rapid pacing, and the emergence of a spatially discordant alternans profile in STIM1-KD hearts. Unlike conduction velocity slowing and APD heterogeneity, the magnitude of APD alternans was greater (by 80%, P<0.05) in VT/VF(+) versus VT/VF(-) STIM1-KD hearts. Detailed phase mapping during the initial beats of VT/VF identified one or more rotors that were localized along the nodal line separating out-of-phase alternans regions. CONCLUSIONS In an adult murine model with inducible and myocyte-specific STIM1 depletion, we demonstrate for the first time the regulation of spatially discordant alternans by STIM1. Early mortality in STIM1-KD mice is likely related to enhanced susceptibility to VT/VF secondary to discordant APD alternans.
Collapse
Affiliation(s)
- Marine Cacheux
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Benjamin Strauss
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Nour Raad
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Zeki Ilkan
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Jun Hu
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Ludovic Benard
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine (S.F.)
| | - Jean-Sebastien Hulot
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| | - Fadi G Akar
- Cardiovascular Research Center, Division of Cardiology, Icahn School of Medicine at Mount Sinai (M.C., B.S., N.R., Z.I., J.H., L.B., J.-S.H., F.G.A.)
| |
Collapse
|
22
|
Glucocorticoid stimulation increases cardiac contractility by SGK1-dependent SOCE-activation in rat cardiac myocytes. PLoS One 2019; 14:e0222341. [PMID: 31498847 PMCID: PMC6733454 DOI: 10.1371/journal.pone.0222341] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/27/2019] [Indexed: 01/28/2023] Open
Abstract
Aims Glucocorticoid (GC) stimulation has been shown to increase cardiac contractility by elevated intracellular [Ca] but the sources for Ca entry are unclear. This study aims to determine the role of store-operated Ca entry (SOCE) for GC-mediated inotropy. Methods and results Dexamethasone (Dex) pretreatment significantly increased cardiac contractile force ex vivo in Langendorff-perfused Sprague-Dawley rat hearts (2 mg/kg BW i.p. Dex 24 h prior to experiment). Moreover, Ca transient amplitude as well as fractional shortening were significantly enhanced in Fura-2-loaded isolated rat ventricular myocytes exposed to Dex (1 mg/mL Dex, 24 h). Interestingly, these Dex-dependent effects could be abolished in the presence of SOCE-inhibitors SKF-96356 (SKF, 2 μM) and BTP2 (5 μM). Ca transient kinetics (time to peak, decay time) were not affected by SOCE stimulation. Direct SOCE measurements revealed a negligible magnitude in untreated myocytes but a dramatic increase in SOCE upon Dex-pretreatment. Importantly, the Dex-dependent stimulation of SOCE could be blocked by inhibition of serum and glucocorticoid-regulated kinase 1 (SGK1) using EMD638683 (EMD, 50 μM). Dex preincubation also resulted in increased mRNA expression of proteins involved in SOCE (stromal interaction molecule 2, STIM2, and transient receptor potential cation channels 3/6, TRPC 3/6), which were also prevented in the presence of EMD. Conclusion Short-term GC-stimulation with Dex improves cardiac contractility by a SOCE-dependent mechanism, which appears to involve increased SGK1-dependent expression of the SOCE-related proteins. Since Ca transient kinetics were unaffected, SOCE appears to influence Ca cycling more by an integrated response across multiple cardiac cycles but not on a beat-to-beat basis.
Collapse
|
23
|
Beringer A, Gouriou Y, Lavocat F, Ovize M, Miossec P. Blockade of Store-Operated Calcium Entry Reduces IL-17/TNF Cytokine-Induced Inflammatory Response in Human Myoblasts. Front Immunol 2019; 9:3170. [PMID: 30693003 PMCID: PMC6339936 DOI: 10.3389/fimmu.2018.03170] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/24/2018] [Indexed: 12/20/2022] Open
Abstract
Muscle inflammation as in idiopathic inflammatory myopathies (IIM) leads to muscle weakness, mononuclear cell infiltration, and myofiber dysfunction affecting calcium channels. The effects of interleukin-17A (IL-17) and tumor necrosis factor-α (TNFα) on inflammation and calcium changes were investigated in human myoblasts. Human myoblasts were exposed to IL-17 and/or TNFα with/without store-operated Ca2+ entry (SOCE) inhibitors (2-ABP or BTP2). For co-cultures, peripheral blood mononuclear cells (PBMC) from healthy donors activated or not with phytohemagglutinin (PHA) were added to myoblasts at a 5:1 ratio. IL-17 and TNFα induced in synergy CCL20 and IL-6 production by myoblasts (>14-fold). PBMC-myoblast co-cultures enhanced CCL20 and IL-6 production in the presence or not of PHA compared to PBMC or myoblast monocultures. Anti-IL-17 and/or anti-TNFα decreased the production of IL-6 in co-cultures (p < 0.05). Transwell system that prevents direct cell-cell contact reduced CCL20 (p < 0.01) but not IL-6 secretion. IL-17 and/or TNFα increased the level of the ER stress marker Grp78, mitochondrial ROS and promoted SOCE activation by 2-fold (p < 0.01) in isolated myoblasts. SOCE inhibitors reduced the IL-6 production induced by IL-17/TNFα. Therefore, muscle inflammation induced by IL-17 and/or TNFα may increase muscle cell dysfunction, which, in turn, increased inflammation. Such close interplay between immune and non-immune mechanisms may drive and increase muscle inflammation and weakness.
Collapse
Affiliation(s)
- Audrey Beringer
- Immunogenomics and Inflammation Research Unit EA4130, Department of Clinical Immunology and Rheumatology, Edouard Herriot Hospital, University of Lyon, Hospices Civils de Lyon, Lyon, France
| | - Yves Gouriou
- CarMeN Laboratory, University of Lyon, INSERM 1060, INRA 1397, Université Claude Bernard Lyon1, INSA Lyon, Groupement Hospitalier Est, Bron, France.,Service d'Explorations Fonctionnelles Cardiovasculaires and CIC de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | - Fabien Lavocat
- Immunogenomics and Inflammation Research Unit EA4130, Department of Clinical Immunology and Rheumatology, Edouard Herriot Hospital, University of Lyon, Hospices Civils de Lyon, Lyon, France
| | - Michel Ovize
- CarMeN Laboratory, University of Lyon, INSERM 1060, INRA 1397, Université Claude Bernard Lyon1, INSA Lyon, Groupement Hospitalier Est, Bron, France.,Service d'Explorations Fonctionnelles Cardiovasculaires and CIC de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit EA4130, Department of Clinical Immunology and Rheumatology, Edouard Herriot Hospital, University of Lyon, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
24
|
Role of STIM1/ORAI1-mediated store-operated Ca 2+ entry in skeletal muscle physiology and disease. Cell Calcium 2018; 76:101-115. [PMID: 30414508 DOI: 10.1016/j.ceca.2018.10.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 11/23/2022]
Abstract
Store-operated Ca2+ entry (SOCE) is a Ca2+ entry mechanism activated by depletion of intracellular Ca2+ stores. In skeletal muscle, SOCE is mediated by an interaction between stromal-interacting molecule-1 (STIM1), the Ca2+ sensor of the sarcoplasmic reticulum, and ORAI1, the Ca2+-release-activated-Ca2+ (CRAC) channel located in the transverse tubule membrane. This review focuses on the molecular mechanisms and physiological role of SOCE in skeletal muscle, as well as how alterations in STIM1/ORAI1-mediated SOCE contribute to muscle disease. Recent evidence indicates that SOCE plays an important role in both muscle development/growth and fatigue. The importance of SOCE in muscle is further underscored by the discovery that loss- and gain-of-function mutations in STIM1 and ORAI1 result in an eclectic array of disorders with clinical myopathy as central defining component. Despite differences in clinical phenotype, all STIM1/ORAI1 gain-of-function mutations-linked myopathies are characterized by the abnormal accumulation of intracellular membranes, known as tubular aggregates. Finally, dysfunctional STIM1/ORAI1-mediated SOCE also contributes to the pathogenesis of muscular dystrophy, malignant hyperthermia, and sarcopenia. The picture to emerge is that tight regulation of STIM1/ORAI1-dependent Ca2+ signaling is critical for optimal skeletal muscle development/function such that either aberrant increases or decreases in SOCE activity result in muscle dysfunction.
Collapse
|
25
|
Agrawal A, Suryakumar G, Rathor R. Role of defective Ca 2+ signaling in skeletal muscle weakness: Pharmacological implications. J Cell Commun Signal 2018; 12:645-659. [PMID: 29982883 DOI: 10.1007/s12079-018-0477-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/27/2018] [Indexed: 01/19/2023] Open
Abstract
The misbehaving attitude of Ca2+ signaling pathways could be the probable reason in many muscular disorders such as myopathies, systemic disorders like hypoxia, sepsis, cachexia, sarcopenia, heart failure, and dystrophy. The present review throws light upon the calcium flux regulating signaling channels like ryanodine receptor complex (RyR1), SERCA (Sarco-endoplasmic Reticulum Calcium ATPase), DHPR (Dihydropyridine Receptor) or Cav1.1 and Na+/Ca2+ exchange pump in detail and how remodelling of these channels contribute towards disturbed calcium homeostasis. Understanding these pathways will further provide an insight for establishing new therapeutic approaches for the prevention and treatment of muscle atrophy under stress conditions, targeting calcium ion channels and associated regulatory proteins.
Collapse
Affiliation(s)
- Akanksha Agrawal
- DRDO, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Geetha Suryakumar
- DRDO, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Richa Rathor
- DRDO, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
26
|
Nguyen NT, Han W, Cao W, Wang Y, Wen S, Huang Y, Li M, Du L, Zhou Y. Store‐Operated Calcium Entry Mediated by ORAI and STIM. Compr Physiol 2018; 8:981-1002. [DOI: 10.1002/cphy.c170031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
27
|
Ma G, Zheng S, Ke Y, Zhou L, He L, Huang Y, Wang Y, Zhou Y. Molecular Determinants for STIM1 Activation During Store- Operated Ca2+ Entry. Curr Mol Med 2018; 17:60-69. [PMID: 28231751 DOI: 10.2174/1566524017666170220103731] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 01/31/2017] [Accepted: 02/01/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND STIM/ORAI-mediated store-operated Ca2+ entry (SOCE) mediates a myriad of Ca2+-dependent cellular activities in mammals. Genetic defects in STIM1/ORAI1 lead to devastating severe combined immunodeficiency; whereas gain-offunction mutations in STIM1/ORAI1 are intimately associated with tubular aggregate myopathy. At molecular level, a decrease in the Ca2+ concentrations within the lumen of endoplasmic reticulum (ER) initiates multimerization of the STIM1 luminal domain to switch on the STIM1 cytoplasmic domain to engage and gate ORAI channels, thereby leading to the ultimate Ca2+ influx from the extracellular space into the cytosol. Despite tremendous progress made in dissecting functional STIM1-ORAI1 coupling, the activation mechanism of SOCE remains to be fully characterized. OBJECTIVE AND METHODS Building upon a robust fluorescence resonance energy transfer assay designed to monitor STIM1 intramolecular autoinhibition, we aimed to systematically dissect the molecular determinants required for the activation and oligomerization of STIM1. RESULTS Here we showed that truncation of the STIM1 luminal domain predisposes STIM1 to adopt a more active conformation. Replacement of the single transmembrane (TM) domain of STIM1 by a more rigid dimerized TM domain of glycophorin A abolished STIM1 activation. But this adverse effect could be partially reversed by disrupting the TM dimerization interface. Moreover, our study revealed regions that are important for the optimal assembly of hetero-oligomers composed of full-length STIM1 with its minimal STIM1-ORAI activating region, SOAR. CONCLUSIONS Our study clarifies the roles of major STIM1 functional domains in maintaining a quiescent configuration of STIM1 to prevent preactivation of SOCE.
Collapse
Affiliation(s)
- G Ma
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030. United States
| | - S Zheng
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875. China
| | - Y Ke
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030. United States
| | - L Zhou
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875. China
| | - L He
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030. United States
| | - Y Huang
- Center for Epigenetic and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030. United States
| | - Y Wang
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University System Health Science Center, 2121 W. Holcombe Blvd., Houston, TX 77030. United States
| | - Y Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University System Health Science Center, 2121 W. Holcombe Blvd., Houston, TX 77030. United States
| |
Collapse
|
28
|
Avila-Medina J, Mayoral-Gonzalez I, Dominguez-Rodriguez A, Gallardo-Castillo I, Ribas J, Ordoñez A, Rosado JA, Smani T. The Complex Role of Store Operated Calcium Entry Pathways and Related Proteins in the Function of Cardiac, Skeletal and Vascular Smooth Muscle Cells. Front Physiol 2018; 9:257. [PMID: 29618985 PMCID: PMC5872157 DOI: 10.3389/fphys.2018.00257] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/06/2018] [Indexed: 12/11/2022] Open
Abstract
Cardiac, skeletal, and smooth muscle cells shared the common feature of contraction in response to different stimuli. Agonist-induced muscle's contraction is triggered by a cytosolic free Ca2+ concentration increase due to a rapid Ca2+ release from intracellular stores and a transmembrane Ca2+ influx, mainly through L-type Ca2+ channels. Compelling evidences have demonstrated that Ca2+ might also enter through other cationic channels such as Store-Operated Ca2+ Channels (SOCCs), involved in several physiological functions and pathological conditions. The opening of SOCCs is regulated by the filling state of the intracellular Ca2+ store, the sarcoplasmic reticulum, which communicates to the plasma membrane channels through the Stromal Interaction Molecule 1/2 (STIM1/2) protein. In muscle cells, SOCCs can be mainly non-selective cation channels formed by Orai1 and other members of the Transient Receptor Potential-Canonical (TRPC) channels family, as well as highly selective Ca2+ Release-Activated Ca2+ (CRAC) channels, formed exclusively by subunits of Orai proteins likely organized in macromolecular complexes. This review summarizes the current knowledge of the complex role of Store Operated Calcium Entry (SOCE) pathways and related proteins in the function of cardiac, skeletal, and vascular smooth muscle cells.
Collapse
Affiliation(s)
- Javier Avila-Medina
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain.,Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, CSIC, University of Seville, Sevilla, Spain.,CIBERCV, Madrid, Spain
| | | | - Alejandro Dominguez-Rodriguez
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain.,Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, CSIC, University of Seville, Sevilla, Spain.,CIBERCV, Madrid, Spain
| | | | - Juan Ribas
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain
| | - Antonio Ordoñez
- CIBERCV, Madrid, Spain.,Department of Surgery, University of Seville, Sevilla, Spain
| | - Juan A Rosado
- Cell Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, University of Seville, Sevilla, Spain.,Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío, CSIC, University of Seville, Sevilla, Spain.,CIBERCV, Madrid, Spain
| |
Collapse
|
29
|
Abstract
During the complex series of events leading to muscle contraction, the initial electric signal coming from motor neurons is transformed into an increase in calcium concentration that triggers sliding of myofibrils. This process, referred to as excitation-contraction coupling, is reliant upon the calcium-release complex, which is restricted spatially to a sub-compartment of muscle cells ("the triad") and regulated precisely. Any dysfunction in the calcium-release complex leads to muscle impairment and myopathy. Various causes can lead to alterations in excitation-contraction coupling and to muscle diseases. The latter are reviewed and classified into four categories: (i) mutation in a protein of the calcium-release complex; (ii) alteration in triad structure; (iii) modification of regulation of channels; (iv) modification in calcium stores within the muscle. Current knowledge of the pathophysiologic mechanisms in each category is described and discussed.
Collapse
Affiliation(s)
- Isabelle Marty
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000 Grenoble, France.,INSERM, U1216, F-38000 Grenoble, France
| | - Julien Fauré
- University Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000 Grenoble, France.,INSERM, U1216, F-38000 Grenoble, France.,CHU de Grenoble, F-38000 Grenoble, France
| |
Collapse
|
30
|
Domenichini F, Terrié E, Arnault P, Harnois T, Magaud C, Bois P, Constantin B, Coronas V. Store-Operated Calcium Entries Control Neural Stem Cell Self-Renewal in the Adult Brain Subventricular Zone. Stem Cells 2018; 36:761-774. [PMID: 29359518 DOI: 10.1002/stem.2786] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/19/2017] [Accepted: 01/10/2018] [Indexed: 12/13/2022]
Abstract
The subventricular zone (SVZ) is the major stem cell niche in the brain of adult mammals. Within this region, neural stem cells (NSC) proliferate, self-renew and give birth to neurons and glial cells. Previous studies underlined enrichment in calcium signaling-related transcripts in adult NSC. Because of their ability to mobilize sustained calcium influxes in response to a wide range of extracellular factors, store-operated channels (SOC) appear to be, among calcium channels, relevant candidates to induce calcium signaling in NSC whose cellular activities are continuously adapted to physiological signals from the microenvironment. By Reverse Transcription Polymerase Chain Reaction (RT-PCR), Western blotting and immunocytochemistry experiments, we demonstrate that SVZ cells express molecular actors known to build up SOC, namely transient receptor potential canonical 1 (TRPC1) and Orai1, as well as their activator stromal interaction molecule 1 (STIM1). Calcium imaging reveals that SVZ cells display store-operated calcium entries. Pharmacological blockade of SOC with SKF-96365 or YM-58483 (also called BTP2) decreases proliferation, impairs self-renewal by shifting the type of SVZ stem cell division from symmetric proliferative to asymmetric, thereby reducing the stem cell population. Brain section immunostainings show that TRPC1, Orai1, and STIM1 are expressed in vivo, in SOX2-positive SVZ NSC. Injection of SKF-96365 in brain lateral ventricle diminishes SVZ cell proliferation and reduces the ability of SVZ cells to form neurospheres in vitro. The present study combining in vitro and in vivo approaches uncovers a major role for SOC in the control of SVZ NSC population and opens new fields of investigation for stem cell biology in health and disease. Stem Cells 2018;36:761-774.
Collapse
Affiliation(s)
- Florence Domenichini
- Signalisation et Transports Ioniques Membranaires, University of Poitiers, CNRS ERL 7003, Poitiers Cedex 09, France
| | - Elodie Terrié
- Signalisation et Transports Ioniques Membranaires, University of Poitiers, CNRS ERL 7003, Poitiers Cedex 09, France
| | - Patricia Arnault
- Signalisation et Transports Ioniques Membranaires, University of Poitiers, CNRS ERL 7003, Poitiers Cedex 09, France
| | - Thomas Harnois
- Signalisation et Transports Ioniques Membranaires, University of Poitiers, CNRS ERL 7003, Poitiers Cedex 09, France
| | - Christophe Magaud
- Signalisation et Transports Ioniques Membranaires, University of Poitiers, CNRS ERL 7003, Poitiers Cedex 09, France
| | - Patrick Bois
- Signalisation et Transports Ioniques Membranaires, University of Poitiers, CNRS ERL 7003, Poitiers Cedex 09, France
| | - Bruno Constantin
- Signalisation et Transports Ioniques Membranaires, University of Poitiers, CNRS ERL 7003, Poitiers Cedex 09, France
| | - Valérie Coronas
- Signalisation et Transports Ioniques Membranaires, University of Poitiers, CNRS ERL 7003, Poitiers Cedex 09, France
| |
Collapse
|
31
|
Nelson HA, Roe MW. Molecular physiology and pathophysiology of stromal interaction molecules. Exp Biol Med (Maywood) 2018; 243:451-472. [PMID: 29363328 DOI: 10.1177/1535370218754524] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Ca2+ release from the endoplasmic reticulum is an important component of Ca2+ signal transduction that controls numerous physiological processes in eukaryotic cells. Release of Ca2+ from the endoplasmic reticulum is coupled to the activation of store-operated Ca2+ entry into cells. Store-operated Ca2+ entry provides Ca2+ for replenishing depleted endoplasmic reticulum Ca2+ stores and a Ca2+ signal that regulates Ca2+-dependent intracellular biochemical events. Central to connecting discharge of endoplasmic reticulum Ca2+ stores following G protein-coupled receptor activation with the induction of store-operated Ca2+ entry are stromal interaction molecules (STIM1 and STIM2). These highly homologous endoplasmic reticulum transmembrane proteins function as sensors of the Ca2+ concentration within the endoplasmic reticulum lumen and activators of Ca2+ release-activated Ca2+ channels. Emerging evidence indicates that in addition to their role in Ca2+ release-activated Ca2+ channel gating and store-operated Ca2+ entry, STIM1 and STIM2 regulate other cellular signaling events. Recent studies have shown that disruption of STIM expression and function is associated with the pathogenesis of several diseases including autoimmune disorders, cancer, cardiovascular disease, and myopathies. Here, we provide an overview of the latest developments in the molecular physiology and pathophysiology of STIM1 and STIM2. Impact statement Intracellular Ca2+ signaling is a fundamentally important regulator of cell physiology. Recent studies have revealed that Ca2+-binding stromal interaction molecules (Stim1 and Stim2) expressed in the membrane of the endoplasmic reticulum (ER) are essential components of eukaryote Ca2+ signal transduction that control the activity of ion channels and other signaling effectors present in the plasma membrane. This review summarizes the most recent information on the molecular physiology and pathophysiology of stromal interaction molecules. We anticipate that the work presented in our review will provide new insights into molecular interactions that participate in interorganelle signaling crosstalk, cell function, and the pathogenesis of human diseases.
Collapse
Affiliation(s)
- Heather A Nelson
- 1 Department of Cell and Developmental Biology, 12302 SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Michael W Roe
- 1 Department of Cell and Developmental Biology, 12302 SUNY Upstate Medical University, Syracuse, NY 13210, USA.,2 Department of Medicine, 12302 SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
32
|
SOCE Is Important for Maintaining Sarcoplasmic Calcium Content and Release in Skeletal Muscle Fibers. Biophys J 2018; 113:2496-2507. [PMID: 29212003 DOI: 10.1016/j.bpj.2017.09.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/01/2017] [Accepted: 09/21/2017] [Indexed: 11/20/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) is a Ca2+-entry process activated by the depletion of intracellular stores and has an important role in many cell types. In skeletal muscle, however, its role during physiological muscle activation has been controversial. To address this question, sarcoplasmic reticulum (SR) calcium release in a mouse strain with a naturally occurring mutation in the myostatin gene (Compact (Cmpt)) leading to a hypermuscular yet reduced muscle-force phenotype was compared to that in wild-type mice. To elicit Ca2+ release from the SR of flexor digitorum brevis (FDB) fibers, either a ryanodine receptor agonist (4-chloro-meta-cresol) or depolarizing pulses were used. In muscles from Cmpt mice, endogenous protein levels of STIM1 and Orai1 were reduced, and consequently, SOCE after 4-chloro-meta-cresol-induced store depletion was suppressed. Although the voltage dependence of SR calcium release was not statistically different between wild-type and Cmpt fibers, the amount of releasable calcium was significantly reduced in the latter, indicating a smaller SR content. To assess the immediate role of SOCE in replenishing the SR calcium store, the evolution of intracellular calcium concentration during a train of long-lasting depolarizations to a maximally activating voltage was monitored. Cmpt mice exhibited a faster decline in calcium release, suggesting a compromised ability to refill the SR. A simple model that incorporates a reduced SOCE as an important partner in regulating immediate calcium influx through the surface membrane readily accounts for the steady-state reduction in SR calcium content and its more pronounced decline after calcium release.
Collapse
|
33
|
Oh MR, Lee KJ, Huang M, Kim JO, Kim DH, Cho CH, Lee EH. STIM2 regulates both intracellular Ca 2+ distribution and Ca 2+ movement in skeletal myotubes. Sci Rep 2017; 7:17936. [PMID: 29263348 PMCID: PMC5738411 DOI: 10.1038/s41598-017-18256-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/08/2017] [Indexed: 01/09/2023] Open
Abstract
Stromal interaction molecule 1 (STIM1) along with Orai1 mediates extracellular Ca2+ entry into the cytosol through a store-operated Ca2+ entry (SOCE) mechanism in various tissues including skeletal muscle. However, the role(s) of STIM2, a homolog of STIM1, in skeletal muscle has not been well addressed. The present study, first, was focused on searching for STIM2-binding proteins from among proteins mediating skeletal muscle functions. This study used a binding assay, quadrupole time-of-flight mass spectrometry, and co-immunoprecipitation assay with bona-fide STIM2- and SERCA1a-expressing rabbit skeletal muscle. The region for amino acids from 453 to 729 of STIM2 binds to sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 1a (SERCA1a). Next, oxalate-supported 45Ca2+-uptake experiments and various single-myotube Ca2+ imaging experiments using STIM2-knockdown mouse primary skeletal myotubes have suggested that STIM2 attenuates SERCA1a activity during skeletal muscle contraction, which contributes to the intracellular Ca2+ distribution between the cytosol and the SR at rest. In addition, STIM2 regulates Ca2+ movement through RyR1 during skeletal muscle contraction as well as SOCE. Therefore, via regulation of SERCA1a activity, STIM2 regulates both intracellular Ca2+ distribution and Ca2+ movement in skeletal muscle, which makes it both similar to, yet different from, STIM1.
Collapse
Affiliation(s)
- Mi Ri Oh
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Keon Jin Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Mei Huang
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Jin Ock Kim
- School of Life Sciences, GIST, Gwangju, 61005, Republic of Korea
| | - Do Han Kim
- School of Life Sciences, GIST, Gwangju, 61005, Republic of Korea
| | - Chung-Hyun Cho
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eun Hui Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
34
|
Boghossian NS, Sicko RJ, Giannakou A, Dimopoulos A, Caggana M, Tsai MY, Yeung EH, Pankratz N, Cole BR, Romitti PA, Browne ML, Fan R, Liu A, Kay DM, Mills JL. Rare copy number variants identified in prune belly syndrome. Eur J Med Genet 2017; 61:145-151. [PMID: 29174092 DOI: 10.1016/j.ejmg.2017.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/31/2017] [Accepted: 11/21/2017] [Indexed: 11/26/2022]
Abstract
Prune belly syndrome (PBS), also known as Eagle-Barrett syndrome, is a rare congenital disorder characterized by absence or hypoplasia of the abdominal wall musculature, urinary tract anomalies, and cryptorchidism in males. The etiology of PBS is largely unresolved, but genetic factors are implicated given its recurrence in families. We examined cases of PBS to identify novel pathogenic copy number variants (CNVs). A total of 34 cases (30 males and 4 females) with PBS identified from all live births in New York State (1998-2005) were genotyped using Illumina HumanOmni2.5 microarrays. CNVs were prioritized if they were absent from in-house controls, encompassed ≥10 consecutive probes, were ≥20 Kb in size, had ≤20% overlap with common variants in population reference controls, and had ≤20% overlap with any variant previously detected in other birth defect phenotypes screened in our laboratory. We identified 17 candidate autosomal CNVs; 10 cases each had one CNV and four cases each had two CNVs. The CNVs included a 158 Kb duplication at 4q22 that overlaps the BMPR1B gene; duplications of different sizes carried by two cases in the intron of STIM1 gene; a 67 Kb duplication 202 Kb downstream of the NOG gene, and a 1.34 Mb deletion including the MYOCD gene. The identified rare CNVs spanned genes involved in mesodermal, muscle, and urinary tract development and differentiation, which might help in elucidating the genetic contribution to PBS. We did not have parental DNA and cannot identify whether these CNVs were de novo or inherited. Further research on these CNVs, particularly BMP signaling is warranted to elucidate the pathogenesis of PBS.
Collapse
Affiliation(s)
- Nansi S Boghossian
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States; Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States.
| | - Robert J Sicko
- Division of Genetics, Wadsworth Center, Department of Health, Albany, NY, United States
| | - Andreas Giannakou
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Aggeliki Dimopoulos
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Michele Caggana
- Division of Genetics, Wadsworth Center, Department of Health, Albany, NY, United States
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Edwina H Yeung
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Benjamin R Cole
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Paul A Romitti
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, IA, United States
| | - Marilyn L Browne
- New York State Department of Health, Congenital Malformations Registry, Albany, NY, United States; University at Albany School of Public Health, Rensselaer, NY, United States
| | - Ruzong Fan
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University Medical Center (GUMC), Washington, DC, United States
| | - Aiyi Liu
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Denise M Kay
- Division of Genetics, Wadsworth Center, Department of Health, Albany, NY, United States
| | - James L Mills
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
35
|
A focus on extracellular Ca 2+ entry into skeletal muscle. Exp Mol Med 2017; 49:e378. [PMID: 28912570 PMCID: PMC5628281 DOI: 10.1038/emm.2017.208] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/16/2017] [Accepted: 06/28/2017] [Indexed: 01/06/2023] Open
Abstract
The main task of skeletal muscle is contraction and relaxation for body movement and posture maintenance. During contraction and relaxation, Ca2+ in the cytosol has a critical role in activating and deactivating a series of contractile proteins. In skeletal muscle, the cytosolic Ca2+ level is mainly determined by Ca2+ movements between the cytosol and the sarcoplasmic reticulum. The importance of Ca2+ entry from extracellular spaces to the cytosol has gained significant attention over the past decade. Store-operated Ca2+ entry with a low amplitude and relatively slow kinetics is a main extracellular Ca2+ entryway into skeletal muscle. Herein, recent studies on extracellular Ca2+ entry into skeletal muscle are reviewed along with descriptions of the proteins that are related to extracellular Ca2+ entry and their influences on skeletal muscle function and disease.
Collapse
|
36
|
Lee JM, Noguchi S. Calcium Dyshomeostasis in Tubular Aggregate Myopathy. Int J Mol Sci 2016; 17:ijms17111952. [PMID: 27879676 PMCID: PMC5133946 DOI: 10.3390/ijms17111952] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/15/2016] [Accepted: 11/15/2016] [Indexed: 11/16/2022] Open
Abstract
Calcium is a crucial mediator of cell signaling in skeletal muscles for basic cellular functions and specific functions, including contraction, fiber-type differentiation and energy production. The sarcoplasmic reticulum (SR) is an organelle that provides a large supply of intracellular Ca2+ in myofibers. Upon excitation, it releases Ca2+ into the cytosol, inducing contraction of myofibrils. During relaxation, it takes up cytosolic Ca2+ to terminate the contraction. During exercise, Ca2+ is cycled between the cytosol and the SR through a system by which the Ca2+ pool in the SR is restored by uptake of extracellular Ca2+ via a specific channel on the plasma membrane. This channel is called the store-operated Ca2+ channel or the Ca2+ release-activated Ca2+ channel. It is activated by depletion of the Ca2+ store in the SR by coordination of two main molecules: stromal interaction molecule 1 (STIM1) and calcium release-activated calcium channel protein 1 (ORAI1). Recently, myopathies with a dominant mutation in these genes have been reported and the pathogenic mechanism of such diseases have been proposed. This review overviews the calcium signaling in skeletal muscles and role of store-operated Ca2+ entry in calcium homeostasis. Finally, we discuss the phenotypes and the pathomechanism of myopathies caused by mutations in the STIM1 and ORAI1 genes.
Collapse
Affiliation(s)
- Jong-Mok Lee
- Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Neuropsychiatry, Kodaira, Tokyo 187-8551, Japan.
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Neuropsychiatry, Kodaira, Tokyo 187-8502, Japan.
| | - Satoru Noguchi
- Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Neuropsychiatry, Kodaira, Tokyo 187-8551, Japan.
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Neuropsychiatry, Kodaira, Tokyo 187-8502, Japan.
| |
Collapse
|
37
|
Dufresne SS, Boulanger-Piette A, Bossé S, Frenette J. Physiological role of receptor activator nuclear factor-kB (RANK) in denervation-induced muscle atrophy and dysfunction. ACTA ACUST UNITED AC 2016; 3:e13231-e13236. [PMID: 27547781 PMCID: PMC4991940 DOI: 10.14800/rci.1323] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The bone remodeling and homeostasis are mainly controlled by the receptor-activator of nuclear factor kB (RANK), its ligand RANKL, and the soluble decoy receptor osteoprotegerin (OPG) pathway. While there is a strong association between osteoporosis and skeletal muscle dysfunction, the functional relevance of a particular biological pathway that synchronously regulates bone and skeletal muscle physiopathology remains elusive. Our recent article published in the American Journal of Physiology (Cell Physiology) showed that RANK is also expressed in fully differentiated C2C12 myotubes and skeletal muscles. We used the Cre-Lox approach to inactivate muscle RANK (RANKmko) and showed that RANK deletion preserves the force of denervated fast-twitch EDL muscles. However, RANK deletion had no positive impact on slow-twitch Sol muscles. In addition, denervating RANKmko EDL muscles induced an increase in the total calcium concentration ([CaT]), which was associated with a surprising decrease in SERCA activity. Interestingly, the levels of STIM-1, which mediates Ca2+ influx following the depletion of SR Ca2+ stores, were markedly higher in denervated RANKmko EDL muscles. We speculated that extracellular Ca2+ influx mediated by STIM-1 may be important for the increase in [CaT] and the gain of force in denervated RANKmko EDL muscles. Overall, these findings showed for the first time that the RANKL/RANK interaction plays a role in denervation-induced muscle atrophy and dysfunction.
Collapse
Affiliation(s)
- Sébastien S Dufresne
- Centre Hospitalier Universitaire de Québec-Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CRCHUL), Université Laval, Quebec City, Quebec, G1V 4G2, Canada
| | - Antoine Boulanger-Piette
- Centre Hospitalier Universitaire de Québec-Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CRCHUL), Université Laval, Quebec City, Quebec, G1V 4G2, Canada
| | - Sabrina Bossé
- Centre Hospitalier Universitaire de Québec-Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CRCHUL), Université Laval, Quebec City, Quebec, G1V 4G2, Canada
| | - Jérôme Frenette
- Centre Hospitalier Universitaire de Québec-Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CRCHUL), Université Laval, Quebec City, Quebec, G1V 4G2, Canada; Département de Réadaptation, Faculté de Médecine, Université Laval, Quebec City, Quebec, G1V 4G2, Canada
| |
Collapse
|
38
|
Xia L, Cheung KK, Yeung SS, Yeung EW. The involvement of transient receptor potential canonical type 1 in skeletal muscle regrowth after unloading-induced atrophy. J Physiol 2016; 594:3111-26. [PMID: 26752511 DOI: 10.1113/jp271705] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/06/2016] [Indexed: 02/05/2023] Open
Abstract
KEY POINTS Decreased mechanical loading results in skeletal muscle atrophy. The transient receptor potential canonical type 1 (TRPC1) protein is implicated in this process. Investigation of the regulation of TRPC1 in vivo has rarely been reported. In the present study, we employ the mouse hindlimb unloading and reloading model to examine the involvement of TRPC1 in the regulation of muscle atrophy and regrowth, respectively. We establish the physiological relevance of the concept that manipulation of TRPC1 could interfere with muscle regrowth processes following an atrophy-inducing event. Specifically, we show that suppressing TRPC1 expression during reloading impairs the recovery of the muscle mass and slow myosin heavy chain profile. Calcineurin appears to be part of the signalling pathway involved in the regulation of TRPC1 expression during muscle regrowth. These results provide new insights concerning the function of TRPC1. Interventions targeting TRPC1 or its downstream or upstream pathways could be useful for promoting muscle regeneration. ABSTRACT Decreased mechanical loading, such as bed rest, results in skeletal muscle atrophy. The functional consequences of decreased mechanical loading include a loss of muscle mass and decreased muscle strength, particularly in anti-gravity muscles. The purpose of this investigation was to clarify the regulatory role of the transient receptor potential canonical type 1 (TRPC1) protein during muscle atrophy and regrowth. Mice were subjected to 14 days of hindlimb unloading followed by 3, 7, 14 and 28 days of reloading. Weight-bearing mice were used as controls. TRPC1 expression in the soleus muscle decreased significantly and persisted at 7 days of reloading. Small interfering RNA (siRNA)-mediated downregulation of TRPC1 in weight-bearing soleus muscles resulted in a reduced muscle mass and a reduced myofibre cross-sectional area (CSA). Microinjecting siRNA into soleus muscles in vivo after 7 days of reloading provided further evidence for the role of TRPC1 in regulating muscle regrowth. Myofibre CSA, as well as the percentage of slow myosin heavy chain-positive myofibres, was significantly lower in TRPC1-siRNA-expressing muscles than in control muscles after 14 days of reloading. Additionally, inhibition of calcineurin (CaN) activity downregulated TRPC1 expression in both weight-bearing and reloaded muscles, suggesting a possible association between CaN and TRPC1 during skeletal muscle regrowth.
Collapse
Affiliation(s)
- Lu Xia
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.,Rehabilitation Key Laboratory of Sichuan Province, Department of Rehabilitation, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kwok-Kuen Cheung
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Simon S Yeung
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Ella W Yeung
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| |
Collapse
|
39
|
Abstract
Store-operated calcium channels (SOCs) are a major pathway for calcium signaling in virtually all metozoan cells and serve a wide variety of functions ranging from gene expression, motility, and secretion to tissue and organ development and the immune response. SOCs are activated by the depletion of Ca(2+) from the endoplasmic reticulum (ER), triggered physiologically through stimulation of a diverse set of surface receptors. Over 15 years after the first characterization of SOCs through electrophysiology, the identification of the STIM proteins as ER Ca(2+) sensors and the Orai proteins as store-operated channels has enabled rapid progress in understanding the unique mechanism of store-operate calcium entry (SOCE). Depletion of Ca(2+) from the ER causes STIM to accumulate at ER-plasma membrane (PM) junctions where it traps and activates Orai channels diffusing in the closely apposed PM. Mutagenesis studies combined with recent structural insights about STIM and Orai proteins are now beginning to reveal the molecular underpinnings of these choreographic events. This review describes the major experimental advances underlying our current understanding of how ER Ca(2+) depletion is coupled to the activation of SOCs. Particular emphasis is placed on the molecular mechanisms of STIM and Orai activation, Orai channel properties, modulation of STIM and Orai function, pharmacological inhibitors of SOCE, and the functions of STIM and Orai in physiology and disease.
Collapse
Affiliation(s)
- Murali Prakriya
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| | - Richard S Lewis
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
40
|
Wang YH, Zhang CL, Plath M, Fang XT, Lan XY, Zhou Y, Chen H. Global transcriptional profiling of longissimus thoracis muscle tissue in fetal and juvenile domestic goat using RNA sequencing. Anim Genet 2015; 46:655-65. [PMID: 26364974 DOI: 10.1111/age.12338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2015] [Indexed: 01/05/2023]
Abstract
Domestic goats are important meat production animals; however, data from transcriptional profiling of skeletal muscle tissue in goat have thus far been scarce. We used comparative transcriptional profiling based on RNA sequencing of longissimus thoracis muscle tissue obtained from fetal goat muscle tissue (27 512 850 clean cDNA reads) and 6-month-old goat muscle tissue (27 582 908 reads) to identify genes that are differentially expressed, novel transcript units and alternative splicing events. Gene annotation revealed that 15 960 and 14 981 genes were expressed in the fetal and juvenile libraries respectively. We detected 6432 differentially expressed genes and, when considering GO terms, found 34, 27 and 55 terms to be significantly enriched in molecular function, cellular component and biological process categories respectively. Pathway analysis revealed that larger numbers of differentially expressed genes were enriched in fetal myogenesis or cell proliferation and differentiation-related pathways (such as Wnt), genes involved in the cell cycle and the Notch signaling pathway, and most of the differentially expressed genes involved in these pathways were downregulated in the juvenile goat library. These genes may be involved in various regulation mechanisms during muscle tissue differentiation between the two development stages examined herein. The identified novel transcript units, including both non-coding and coding RNA, as well as alternative splicing events increase the level of complexity of regulation mechanisms during muscle tissue formation and differentiation. Our study provides a comparative transcriptome analysis on goat muscle tissue, which will provide a valuable genomic resource for future studies investigating the molecular basis of skeletal muscle development.
Collapse
Affiliation(s)
- Y H Wang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi, 712100, China.,Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - C L Zhang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - M Plath
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi, 712100, China
| | - X T Fang
- Institute of Cellular and Molecular Biology, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - X Y Lan
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi, 712100, China
| | - Y Zhou
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi, 712100, China
| | - H Chen
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi, 712100, China
| |
Collapse
|
41
|
Pinto MCX, Kihara AH, Goulart VAM, Tonelli FMP, Gomes KN, Ulrich H, Resende RR. Calcium signaling and cell proliferation. Cell Signal 2015; 27:2139-49. [PMID: 26275497 DOI: 10.1016/j.cellsig.2015.08.006] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/04/2015] [Accepted: 08/10/2015] [Indexed: 12/17/2022]
Abstract
Cell proliferation is orchestrated through diverse proteins related to calcium (Ca(2+)) signaling inside the cell. Cellular Ca(2+) influx that occurs first by various mechanisms at the plasma membrane, is then followed by absorption of Ca(2+) ions by mitochondria and endoplasmic reticulum, and, finally, there is a connection of calcium stores to the nucleus. Experimental evidence indicates that the fluctuation of Ca(2+) from the endoplasmic reticulum provides a pivotal and physiological role for cell proliferation. Ca(2+) depletion in the endoplasmatic reticulum triggers Ca(2+) influx across the plasma membrane in an phenomenon called store-operated calcium entries (SOCEs). SOCE is activated through a complex interplay between a Ca(2+) sensor, denominated STIM, localized in the endoplasmic reticulum and a Ca(2+) channel at the cell membrane, denominated Orai. The interplay between STIM and Orai proteins with cell membrane receptors and their role in cell proliferation is discussed in this review.
Collapse
Affiliation(s)
- Mauro Cunha Xavier Pinto
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Presyes 748, 05508-000 São Paulo, SP, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - Alexandre Hiroaki Kihara
- Universidade Federal do ABC, Centro de Matemática, Computação e Cognição, Rua Arcturus (Jd Antares), 09606-070, São Bernardo do Campo, SP, Brazil
| | - Vânia A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - Fernanda M P Tonelli
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - Katia N Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Presyes 748, 05508-000 São Paulo, SP, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil.
| |
Collapse
|
42
|
Sopariwala DH, Pant M, Shaikh SA, Goonasekera SA, Molkentin JD, Weisleder N, Ma J, Pan Z, Periasamy M. Sarcolipin overexpression improves muscle energetics and reduces fatigue. J Appl Physiol (1985) 2015; 118:1050-8. [PMID: 25701006 DOI: 10.1152/japplphysiol.01066.2014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/17/2015] [Indexed: 12/21/2022] Open
Abstract
Sarcolipin (SLN) is a regulator of sarcoendoplasmic reticulum calcium ATPase in skeletal muscle. Recent studies using SLN-null mice have identified SLN as a key player in muscle thermogenesis and metabolism. In this study, we exploited a SLN overexpression (Sln(OE)) mouse model to determine whether increased SLN level affected muscle contractile properties, exercise capacity/fatigue, and metabolic rate in whole animals and isolated muscle. We found that Sln(OE) mice are more resistant to fatigue and can run significantly longer distances than wild-type (WT). Studies with isolated extensor digitorum longus (EDL) muscles showed that Sln(OE) EDL produced higher twitch force than WT. The force-frequency curves were not different between WT and Sln(OE) EDLs, but at lower frequencies the pyruvate-induced potentiation of force was significantly higher in Sln(OE) EDL. SLN overexpression did not alter the twitch and force-frequency curve in isolated soleus muscle. However, during a 10-min fatigue protocol, both EDL and soleus from Sln(OE) mice fatigued significantly less than WT muscles. Interestingly, Sln(OE) muscles showed higher carnitine palmitoyl transferase-1 protein expression, which could enhance fatty acid metabolism. In addition, lactate dehydrogenase expression was higher in Sln(OE) EDL, suggesting increased glycolytic capacity. We also found an increase in store-operated calcium entry (SOCE) in isolated flexor digitorum brevis fibers of Sln(OE) compared with WT mice. These data allow us to conclude that increased SLN expression improves skeletal muscle performance during prolonged muscle activity by increasing SOCE and muscle energetics.
Collapse
Affiliation(s)
- Danesh H Sopariwala
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Meghna Pant
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Sana A Shaikh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | | | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Noah Weisleder
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Jianjie Ma
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Zui Pan
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Muthu Periasamy
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio;
| |
Collapse
|
43
|
Muslikhov ER, Sukhanova IF, Avdonin PV. Arachidonic acid activates release of calcium ions from reticulum via ryanodine receptor channels in C2C12 skeletal myotubes. BIOCHEMISTRY (MOSCOW) 2015; 79:435-9. [PMID: 24954594 DOI: 10.1134/s0006297914050071] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Arachidonic acid causes an increase in free cytoplasmic calcium concentration ([Ca2+]i) in differentiated skeletal multinucleated myotubes C2C12 and does not induce calcium response in C2C12 myoblasts. The same reaction of myotubes to arachidonic acid is observed in Ca2+-free medium. This indicates that arachidonic acid induces release of calcium ions from intracellular stores. The blocker of ryanodine receptor channels of sarcoplasmic reticulum dantrolene (20 µM) inhibits this effect by 68.7 ± 6.3% (p < 0.001). The inhibitor of two-pore calcium channels of endolysosomal vesicles trans-NED19 (10 µM) decreases the response to arachidonic acid by 35.8 ± 5.4% (p < 0.05). The phospholipase C inhibitor U73122 (10 µM) has no effect. These data indicate the involvement of ryanodine receptor calcium channels of sarcoplasmic reticulum in [Ca2+]i elevation in skeletal myotubes caused by arachidonic acid and possible participation of two-pore calcium channels from endolysosomal vesicles in this process.
Collapse
Affiliation(s)
- E R Muslikhov
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia.
| | | | | |
Collapse
|
44
|
Onopiuk M, Brutkowski W, Young C, Krasowska E, Róg J, Ritso M, Wojciechowska S, Arkle S, Zabłocki K, Górecki DC. Store-operated calcium entry contributes to abnormal Ca²⁺ signalling in dystrophic mdx mouse myoblasts. Arch Biochem Biophys 2015; 569:1-9. [PMID: 25659883 DOI: 10.1016/j.abb.2015.01.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 01/20/2015] [Accepted: 01/28/2015] [Indexed: 01/13/2023]
Abstract
Sarcolemma damage and activation of various calcium channels are implicated in altered Ca(2+) homeostasis in muscle fibres of both Duchenne muscular dystrophy (DMD) sufferers and in the mdx mouse model of DMD. Previously we have demonstrated that also in mdx myoblasts extracellular nucleotides trigger elevated cytoplasmic Ca(2+) concentrations due to alterations of both ionotropic and metabotropic purinergic receptors. Here we extend these findings to show that the mdx mutation is associated with enhanced store-operated calcium entry (SOCE). Substantially increased rate of SOCE in mdx myoblasts in comparison to that in control cells correlated with significantly elevated STIM1 protein levels. These results reveal that mutation in the dystrophin-encoding Dmd gene may significantly impact cellular calcium response to metabotropic stimulation involving depletion of the intracellular calcium stores followed by activation of the store-operated calcium entry, as early as in undifferentiated myoblasts. These data are in agreement with the increasing number of reports showing that the dystrophic pathology resulting from dystrophin mutations may be developmentally regulated. Moreover, our results showing that aberrant responses to extracellular stimuli may contribute to DMD pathogenesis suggest that treatments inhibiting such responses might alter progression of this lethal disease.
Collapse
Affiliation(s)
- Marta Onopiuk
- Nencki Institute of Experimental Biology, Warsaw, Poland; Departments of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA(1)
| | - Wojciech Brutkowski
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK; Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Christopher Young
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Elżbieta Krasowska
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK; Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Justyna Róg
- Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Morten Ritso
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Stephen Arkle
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | | | - Dariusz C Górecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
45
|
Ca2+-dependent regulations and signaling in skeletal muscle: from electro-mechanical coupling to adaptation. Int J Mol Sci 2015; 16:1066-95. [PMID: 25569087 PMCID: PMC4307291 DOI: 10.3390/ijms16011066] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/22/2014] [Indexed: 01/07/2023] Open
Abstract
Calcium (Ca2+) plays a pivotal role in almost all cellular processes and ensures the functionality of an organism. In skeletal muscle fibers, Ca(2+) is critically involved in the innervation of skeletal muscle fibers that results in the exertion of an action potential along the muscle fiber membrane, the prerequisite for skeletal muscle contraction. Furthermore and among others, Ca(2+) regulates also intracellular processes, such as myosin-actin cross bridging, protein synthesis, protein degradation and fiber type shifting by the control of Ca(2+)-sensitive proteases and transcription factors, as well as mitochondrial adaptations, plasticity and respiration. These data highlight the overwhelming significance of Ca(2+) ions for the integrity of skeletal muscle tissue. In this review, we address the major functions of Ca(2+) ions in adult muscle but also highlight recent findings of critical Ca(2+)-dependent mechanisms essential for skeletal muscle-regulation and maintenance.
Collapse
|
46
|
Wu Z, Qing J, Xia Y, Wang K, Zhang F. Suppression of stromal interaction molecule 1 inhibits SMMC7721 hepatocellular carcinoma cell proliferation by inducing cell cycle arrest. Biotechnol Appl Biochem 2014; 62:107-11. [PMID: 24842273 DOI: 10.1002/bab.1245] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 05/13/2014] [Indexed: 01/07/2023]
Abstract
Stromal interaction molecule 1 (STIM1), an endoplasmic reticulum luminal Ca(2+) sensor, activates Ca(2+) -release-activated Ca(2+) channels and migrates from the Ca(2+) store to the plasma membrane. Recently, STIM1 was shown be critical for the progression of several cancers, including breast cancer and cervical cancer. However, its role in hepatocellular carcinoma has remained unknown. The current study was aimed to evaluate the effect of STIM1 on the growth of hepatocellular cancer. Lentivirus-mediated short hairpin RNA targeting STIM1 was transduced into SMMC7721 cells to knock down STIM1 expression. Knockdown of STIM1 significantly inhibited cell proliferation and colony-forming ability and arrested the cell cycle at the G0/G1 phase. Moreover, the DNA synthesis progression was also decreased. Furthermore, lentiviral vector-mediated overexpression of STIM1 promoted the proliferation of SMMC7721 cells. Our findings suggest that STIM1 may play an important role in the development of hepatocellular cancer and may be a potential target for therapy.
Collapse
Affiliation(s)
- Zhengshan Wu
- The Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | | | | | | | | |
Collapse
|
47
|
Kósa M, Brinyiczki K, van Damme P, Goemans N, Hancsák K, Mendler L, Zádor E. The neonatal sarcoplasmic reticulum Ca2+-ATPase gives a clue to development and pathology in human muscles. J Muscle Res Cell Motil 2014; 36:195-203. [PMID: 25487304 DOI: 10.1007/s10974-014-9403-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/03/2014] [Indexed: 01/07/2023]
Abstract
The sarcoplasmic/endoplasmic reticulum calcium ATPase 1 (SERCA1) has two muscle specific splice isoforms; SERCA1a in fast-type adult and SERCA1b in neonatal and regenerating skeletal muscles. At the protein level the only difference between these two isoforms is that SERCA1a has C-terminal glycine while SERCA1b has an octapeptide tail instead. This makes the generation of a SERCA1a specific antibody not feasible. The switch between the two isoforms is a hallmark of differentiation so we describe here a method based on the signal ratios of the SERCA1b specific and pan SERCA1 antibodies to estimate the SERCA1b/SERCA1a dominance on immunoblot of human muscles. Using this method we showed that unlike in mouse and rat, SERCA1b was only expressed in pre-matured infant leg and arm muscles; it was replaced by SERCA1a in more matured neonatal muscles and was completely absent in human foetal and neonatal diaphragms. Interestingly, only SERCA1a and no SERCA1b were detected in muscles of 7-12 years old boys with Duchenne, a degenerative-regenerative muscular dystrophy. However, in adult patients with myotonic dystrophy type 2 (DM2), the SERCA1b dominated over SERCA1a. Thus the human SERCA1b has a different expression pattern from that of rodents and it is associated with DM2.
Collapse
Affiliation(s)
- Magdolna Kósa
- Department of Biochemistry, Faculty of General Medicine, University of Szeged, Dóm tér 9, H-6720, Szeged, Hungary
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Ca(2+) release from intracellular stores and influx from extracellular reservoir regulate a wide range of physiological functions including muscle contraction and rhythmic heartbeat. One of the most ubiquitous pathways involved in controlled Ca(2+) influx into cells is store-operated Ca(2+) entry (SOCE), which is activated by the reduction of Ca(2+) concentration in the lumen of endoplasmic or sarcoplasmic reticulum (ER/SR). Although SOCE is pronounced in non-excitable cells, accumulating evidences highlight its presence and important roles in skeletal muscle and heart. Recent discovery of STIM proteins as ER/SR Ca(2+) sensors and Orai proteins as Ca(2+) channel pore forming unit expedited the mechanistic understanding of this pathway. This review focuses on current advances of SOCE components, regulation and physiologic and pathophysiologic roles in muscles. The specific property and the dysfunction of this pathway in muscle diseases, and new directions for future research in this rapidly growing field are discussed.
Collapse
Affiliation(s)
- Zui Pan
- Department of Internal Medicine-Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Marco Brotto
- Muscle Biology Research Group-MUBIG, Schools of Nursing & Medicine, University of Missouri-Kansas City, MO, USA
| | - Jianjie Ma
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
49
|
Endo Y, Noguchi S, Hara Y, Hayashi YK, Motomura K, Miyatake S, Murakami N, Tanaka S, Yamashita S, Kizu R, Bamba M, Goto YI, Matsumoto N, Nonaka I, Nishino I. Dominant mutations in ORAI1 cause tubular aggregate myopathy with hypocalcemia via constitutive activation of store-operated Ca²⁺ channels. Hum Mol Genet 2014; 24:637-48. [PMID: 25227914 DOI: 10.1093/hmg/ddu477] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The store-operated Ca(2+) release-activated Ca(2+) (CRAC) channel is activated by diminished luminal Ca(2+) levels in the endoplasmic reticulum and sarcoplasmic reticulum (SR), and constitutes one of the major Ca(2+) entry pathways in various tissues. Tubular aggregates (TAs) are abnormal structures in the skeletal muscle, and although their mechanism of formation has not been clarified, altered Ca(2+) homeostasis related to a disordered SR is suggested to be one of the main contributing factors. TA myopathy is a hereditary muscle disorder that is pathologically characterized by the presence of TAs. Recently, dominant mutations in the STIM1 gene, encoding a Ca(2+) sensor that controls CRAC channels, have been identified to cause tubular aggregate myopathy (TAM). Here, we identified heterozygous missense mutations in the ORAI1 gene, encoding the CRAC channel itself, in three families affected by dominantly inherited TAM with hypocalcemia. Skeletal myotubes from an affected individual and HEK293 cells expressing mutated ORAI1 proteins displayed spontaneous extracellular Ca(2+) entry into cells without diminishment of luminal Ca(2+) or the association with STIM1. Our results indicate that STIM1-independent activation of CRAC channels induced by dominant mutations in ORAI1 cause altered Ca(2+) homeostasis, resulting in TAM with hypocalcemia.
Collapse
Affiliation(s)
- Yukari Endo
- Department of Clinical Development, Translational Medical Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8556, Japan Department of Neuromuscular Research and Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Satoru Noguchi
- Department of Clinical Development, Translational Medical Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8556, Japan Department of Neuromuscular Research and
| | - Yuji Hara
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan Tokyo Women's Medical University Institute for Integrated Medical Sciences (TIIMS), Shinjuku, Tokyo 162-8666, Japan
| | - Yukiko K Hayashi
- Department of Clinical Development, Translational Medical Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8556, Japan Department of Neuromuscular Research and Department of Neurophysiology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Kazushi Motomura
- Department of Clinical Development, Translational Medical Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8556, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Nobuyuki Murakami
- Department of Pediatrics, Dokkyo Medical University, Koshigaya Hospital, Koshigaya, Saitama 343-8555, Japan
| | - Satsuki Tanaka
- Department of Diabetes and Endocrinology, Osaka Saiseikai Nakatsu Hospital, Osaka 530-0012, Japan
| | - Sumimasa Yamashita
- Division of Pediatric Neurology, Kanagawa Children's Medical Center (KCMC), Yokohama 232-8555, Japan
| | - Rika Kizu
- Division of Pediatrics, Yokosuka Kyosai Hospital, Yokosuka, Kanagawa 238-8558, Japan and
| | - Masahiro Bamba
- Division of Pediatrics, Kawasaki Municipal Hospital, Kawasaki, Kanagawa 210-0013, Japan
| | - Yu-Ichi Goto
- Department of Clinical Development, Translational Medical Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8556, Japan Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | | | - Ichizo Nishino
- Department of Clinical Development, Translational Medical Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8556, Japan Department of Neuromuscular Research and
| |
Collapse
|
50
|
Liantonio A, Camerino GM, Scaramuzzi A, Cannone M, Pierno S, De Bellis M, Conte E, Fraysse B, Tricarico D, Conte Camerino D. Calcium homeostasis is altered in skeletal muscle of spontaneously hypertensive rats: cytofluorimetric and gene expression analysis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2803-15. [PMID: 25084345 DOI: 10.1016/j.ajpath.2014.06.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 06/11/2014] [Accepted: 06/17/2014] [Indexed: 11/25/2022]
Abstract
Hypertension is often associated with skeletal muscle pathological conditions related to function and metabolism. The mechanisms underlying the development of these pathological conditions remain undefined. Because calcium homeostasis is a biomarker of muscle function, we assessed whether it is altered in hypertensive muscles. We measured resting intracellular calcium and store-operated calcium entry (SOCE) in fast- and slow-twitch muscle fibers from normotensive Wistar-Kyoto rats and spontaneously hypertensive rats (SHRs) by cytofluorimetric technique and determined the expression of SOCE gene machinery by real-time PCR. Hypertension caused a phenotype-dependent dysregulation of calcium homeostasis; the resting intracellular calcium of extensor digitorum longus and soleus muscles of SHRs were differently altered with respect to the related muscle of normotensive animals. In addition, soleus muscles of SHR showed reduced activity of the sarcoplasmic reticulum and decreased sarcolemmal calcium permeability at rest and after SOCE activation. Accordingly, we found an alteration of the expression levels of some SOCE components, such as stromal interaction molecule 1, calcium release-activated calcium modulator 1, and transient receptor potential canonical 1. The hypertension-induced alterations of calcium homeostasis in the soleus muscle of SHRs occurred with changes of some functional outcomes as excitability and resting chloride conductance. We provide suitable targets for therapeutic interventions aimed at counterbalancing muscle performance decline in hypertension, and propose the reported calcium-dependent parameters as indexes to predict how the antihypertensive drugs could influence muscle function.
Collapse
Affiliation(s)
- Antonella Liantonio
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy.
| | - Giulia M Camerino
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Antonia Scaramuzzi
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Maria Cannone
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Sabata Pierno
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Michela De Bellis
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Elena Conte
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Bodvael Fraysse
- INRA UMR703, LUNAM Université, Oniris, École Nationale Vétérinaire, Agro-Alimentaire et de l'Alimentation Nantes-Atlantique, Nantes, France
| | - Domenico Tricarico
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Diana Conte Camerino
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| |
Collapse
|