1
|
Kim MY, Shin HY, Cho SC, Yang S, Intisar A, Woo HJ, Choi YS, You CL, Kang JS, Lee YI, Park SC, Yea K, Oh TG, Downes M, Evans RM, Kim MS. Silver electroceutical technology to treat sarcopenia. Proc Natl Acad Sci U S A 2023; 120:e2300036120. [PMID: 37549292 PMCID: PMC10438839 DOI: 10.1073/pnas.2300036120] [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: 01/02/2023] [Accepted: 06/22/2023] [Indexed: 08/09/2023] Open
Abstract
While the world is rapidly transforming into a superaging society, pharmaceutical approaches to treat sarcopenia have hitherto not been successful due to their insufficient efficacy and failure to specifically target skeletal muscle cells (skMCs). Although electrical stimulation (ES) is emerging as an alternative intervention, its efficacy toward treating sarcopenia remains unexplored. In this study, we demonstrate a silver electroceutical technology with the potential to treat sarcopenia. First, we developed a high-throughput ES screening platform that can simultaneously stimulate 15 independent conditions, while utilizing only a small number of human-derived primary aged/young skMCs (hAskMC/hYskMC). The in vitro screening showed that specific ES conditions induced hypertrophy and rejuvenation in hAskMCs, and the optimal ES frequency in hAskMCs was different from that in hYskMCs. When applied to aged mice in vivo, specific ES conditions improved the prevalence and thickness of Type IIA fibers, along with biomechanical attributes, toward a younger skMC phenotype. This study is expected to pave the way toward an electroceutical treatment for sarcopenia with minimal side effects and help realize personalized bioelectronic medicine.
Collapse
Affiliation(s)
- Min Young Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
| | - Hyun Young Shin
- CTCELLS, Inc., Daegu42988, Republic of Korea
- SBCure Corp., Daegu43017, Republic of Korea
| | - Sung Chun Cho
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
| | - Sohae Yang
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
| | - Aseer Intisar
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
| | - Hyeong Jung Woo
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
| | - Youn-Suk Choi
- Samsung Advanced Institute of Technology, Samsung Electronics Co. Ltd., Suwon16678, Republic of Korea
| | - Chang-Lim You
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon16419, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon16419, Republic of Korea
| | - Yun-Il Lee
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
| | - Sang Chul Park
- Chonnam National University Medical School, Gwangju61186, Republic of Korea
| | - Kyungmoo Yea
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
| | - Tae Gyu Oh
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Minseok S. Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
- CTCELLS, Inc., Daegu42988, Republic of Korea
- Translational Responsive Medicine Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Republic of Korea
| |
Collapse
|
2
|
Jaque-Fernandez F, Allard B, Monteiro L, Lafoux A, Huchet C, Jaimovich E, Berthier C, Jacquemond V. Probenecid affects muscle Ca2+ homeostasis and contraction independently from pannexin channel block. J Gen Physiol 2023; 155:e202213203. [PMID: 36820799 PMCID: PMC9998970 DOI: 10.1085/jgp.202213203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 12/21/2022] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Tight control of skeletal muscle contractile activation is secured by the excitation-contraction (EC) coupling protein complex, a molecular machinery allowing the plasma membrane voltage to control the activity of the ryanodine receptor Ca2+ release channel in the sarcoplasmic reticulum (SR) membrane. This machinery has been shown to be intimately linked to the plasma membrane protein pannexin-1 (Panx1). We investigated whether the prescription drug probenecid, a widely used Panx1 blocker, affects Ca2+ signaling, EC coupling, and muscle force. The effect of probenecid was tested on membrane current, resting Ca2+, and SR Ca2+ release in isolated mouse muscle fibers, using a combination of whole-cell voltage-clamp and Ca2+ imaging, and on electrically triggered contraction of isolated muscles. Probenecid (1 mM) induces SR Ca2+ leak at rest and reduces peak voltage-activated SR Ca2+ release and contractile force by 40%. Carbenoxolone, another Panx1 blocker, also reduces Ca2+ release, but neither a Panx1 channel inhibitory peptide nor a purinergic antagonist affected Ca2+ release, suggesting that probenecid and carbenoxolone do not act through inhibition of Panx1-mediated ATP release and consequently altered purinergic signaling. Probenecid may act by altering Panx1 interaction with the EC coupling machinery, yet the implication of another molecular target cannot be excluded. Since probenecid has been used both in the clinic and as a masking agent for doping in sports, these results should encourage evaluation of possible effects on muscle function in treated individuals. In addition, they also raise the question of whether probenecid-induced altered Ca2+ homeostasis may be shared by other tissues.
Collapse
Affiliation(s)
- Francisco Jaque-Fernandez
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| | - Bruno Allard
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| | - Laloé Monteiro
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| | - Aude Lafoux
- Therassay Platform, CAPACITES, Université de Nantes, Nantes, France
| | - Corinne Huchet
- Therassay Platform, CAPACITES, Université de Nantes, Nantes, France
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, Nantes, France
| | - Enrique Jaimovich
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Christine Berthier
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| | - Vincent Jacquemond
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| |
Collapse
|
3
|
Arias-Calderón M, Casas M, Balanta-Melo J, Morales-Jiménez C, Hernández N, Llanos P, Jaimovich E, Buvinic S. Fibroblast growth factor 21 is expressed and secreted from skeletal muscle following electrical stimulation via extracellular ATP activation of the PI3K/Akt/mTOR signaling pathway. Front Endocrinol (Lausanne) 2023; 14:1059020. [PMID: 36909316 PMCID: PMC9997036 DOI: 10.3389/fendo.2023.1059020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 02/08/2023] [Indexed: 02/25/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a hormone involved in the regulation of lipid, glucose, and energy metabolism. Although it is released mainly from the liver, in recent years it has been shown that it is a "myokine", synthesized in skeletal muscles after exercise and stress conditions through an Akt-dependent pathway and secreted for mediating autocrine and endocrine roles. To date, the molecular mechanism for the pathophysiological regulation of FGF21 production in skeletal muscle is not totally understood. We have previously demonstrated that muscle membrane depolarization controls gene expression through extracellular ATP (eATP) signaling, by a mechanism defined as "Excitation-Transcription coupling". eATP signaling regulates the expression and secretion of interleukin 6, a well-defined myokine, and activates the Akt/mTOR signaling pathway. This work aimed to study the effect of electrical stimulation in the regulation of both production and secretion of skeletal muscle FGF21, through eATP signaling and PI3K/Akt pathway. Our results show that electrical stimulation increases both mRNA and protein (intracellular and secreted) levels of FGF21, dependent on an extracellular ATP signaling mechanism in skeletal muscle. Using pharmacological inhibitors, we demonstrated that FGF21 production and secretion from muscle requires the activation of the P2YR/PI3K/Akt/mTOR signaling pathway. These results confirm skeletal muscle as a source of FGF21 in physiological conditions and unveil a new molecular mechanism for regulating FGF21 production in this tissue. Our results will allow to identify new molecular targets to understand the regulation of FGF21 both in physiological and pathological conditions, such as exercise, aging, insulin resistance, and Duchenne muscular dystrophy, all characterized by an alteration in both FGF21 levels and ATP signaling components. These data reinforce that eATP signaling is a relevant mechanism for myokine expression in skeletal muscle.
Collapse
Affiliation(s)
- Manuel Arias-Calderón
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Mariana Casas
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Center for Exercise, Metabolism and Cancer Studies CEMC, Universidad de Chile, Santiago, Chile
| | - Julián Balanta-Melo
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- School of Dentistry, Faculty of Health, Universidad del Valle, Cali, Colombia
| | - Camilo Morales-Jiménez
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Department of Basic Sciences of Health, Faculty of Health Sciences, Pontificia Universidad Javeriana, Cali, Colombia
| | - Nadia Hernández
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Paola Llanos
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Center for Exercise, Metabolism and Cancer Studies CEMC, Universidad de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Center for Exercise, Metabolism and Cancer Studies CEMC, Universidad de Chile, Santiago, Chile
| | - Sonja Buvinic
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Center for Exercise, Metabolism and Cancer Studies CEMC, Universidad de Chile, Santiago, Chile
- *Correspondence: Sonja Buvinic,
| |
Collapse
|
4
|
Senneff S, Lowery MM. Computational Model of the Effect of Mitochondrial Dysfunction on Excitation–Contraction Coupling in Skeletal Muscle. Bull Math Biol 2022; 84:123. [PMID: 36114931 PMCID: PMC9482608 DOI: 10.1007/s11538-022-01079-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 08/31/2022] [Indexed: 11/28/2022]
Abstract
It has become well established that mitochondria not only regulate myoplasmic calcium in skeletal muscle, but also use that calcium to stimulate oxidative phosphorylation (OXPHOS). While experimental approaches have allowed for imaging of mitochondrial calcium and membrane potentials in isolated fibers, capturing the role of mitochondria and the impact of mitochondrial impairments on excitation–contraction coupling (ECC) remains difficult to explore in intact muscle. Computational models have been widely used to examine the structure and function of skeletal muscle contraction; however, models of ECC to date lack communication between the myoplasm and mitochondria for regulating calcium and ATP during sustained contractions. To address this, a mathematical model of mitochondrial calcium handling and OXPHOS was integrated into a physiological model of ECC incorporating action potential propagation, calcium handling between the sarcoplasmic reticulum (SR) and the myoplasm, and crossbridge cycling. The model was used to examine the protective role of mitochondria during repeated stimulation and the impact of mitochondrial dysfunction on ECC resulting from progressive OXPHOS inhibition. Pathological myoplasmic calcium accumulation occurred through distinct mechanisms in the model in the case of either electron transport chain, F1F0 ATP synthase, or adenine nucleotide transporter impairments. To investigate the effect of each impairment on force, a model of calcium-stimulated apoptosis was utilized to capture dysfunction-induced reductions in muscle mass, driving whole muscle force loss. The model presented in this study can be used to examine the role of mitochondria in the regulation of calcium, ATP, and force generation during voluntary contraction.
Collapse
Affiliation(s)
- Sageanne Senneff
- School of Electrical and Electronic Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | - Madeleine M. Lowery
- School of Electrical and Electronic Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
5
|
Nieblas B, Pérez-Treviño P, García N. Role of mitochondria-associated endoplasmic reticulum membranes in insulin sensitivity, energy metabolism, and contraction of skeletal muscle. Front Mol Biosci 2022; 9:959844. [PMID: 36275635 PMCID: PMC9585326 DOI: 10.3389/fmolb.2022.959844] [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: 06/02/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Abstract
Skeletal muscle has a critical role in the regulation of the energy balance of the organism, particularly as the principal tissue responsible for insulin-stimulated glucose disposal and as the major site of peripheral insulin resistance (IR), which has been related to accumulation of lipid intermediates, reduced oxidative capacity of mitochondria and endoplasmic reticulum (ER) stress. These organelles form contact sites, known as mitochondria-associated ER membranes (MAMs). This interconnection seems to be involved in various cellular processes, including Ca2+ transport and energy metabolism; therefore, MAMs could play an important role in maintaining cellular homeostasis. Evidence suggests that alterations in MAMs may contribute to IR. However, the evidence does not refer to a specific subcellular location, which is of interest due to the fact that skeletal muscle is constituted by oxidative and glycolytic fibers as well as different mitochondrial populations that appear to respond differently to stimuli and pathological conditions. In this review, we show the available evidence of possible differential responses in the formation of MAMs in skeletal muscle as well as its role in insulin signaling and the beneficial effect it could have in the regulation of energetic metabolism and muscular contraction.
Collapse
Affiliation(s)
- Bianca Nieblas
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Nuevo León, México
- Experimental Medicine and Advanced Therapies, The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Nuevo León, México
| | - Perla Pérez-Treviño
- Experimental Medicine and Advanced Therapies, The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Nuevo León, México
| | - Noemí García
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Nuevo León, México
- Experimental Medicine and Advanced Therapies, The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Nuevo León, México
- *Correspondence: Noemí García,
| |
Collapse
|
6
|
Mechanical Disturbance of Osteoclasts Induces ATP Release That Leads to Protein Synthesis in Skeletal Muscle through an Akt-mTOR Signaling Pathway. Int J Mol Sci 2022; 23:ijms23169444. [PMID: 36012713 PMCID: PMC9408906 DOI: 10.3390/ijms23169444] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Muscle and bone are tightly integrated through mechanical and biochemical signals. Osteoclasts are cells mostly related to pathological bone loss; however, they also start physiological bone remodeling. Therefore, osteoclast signals released during bone remodeling could improve both bone and skeletal muscle mass. Extracellular ATP is an autocrine/paracrine signaling molecule released by bone and muscle cells. Then, in the present work, it was hypothesized that ATP is a paracrine mediator released by osteoclasts and leads to skeletal muscle protein synthesis. RAW264.7-derived osteoclasts were co-cultured in Transwell® chambers with flexor digitorum brevis (FDB) muscle isolated from adult BalbC mice. The osteoclasts at the upper chamber were mechanically stimulated by controlled culture medium perturbation, resulting in a two-fold increase in protein synthesis in FDB muscle at the lower chamber. Osteoclasts released ATP to the extracellular medium in response to mechanical stimulation, proportional to the magnitude of the stimulus and partly dependent on the P2X7 receptor. On the other hand, exogenous ATP promoted Akt phosphorylation (S473) in isolated FDB muscle in a time- and concentration-dependent manner. ATP also induced phosphorylation of proteins downstream Akt: mTOR (S2448), p70S6K (T389) and 4E-BP1 (T37/46). Exogenous ATP increased the protein synthesis rate in FDB muscle 2.2-fold; this effect was blocked by Suramin (general P2X/P2Y antagonist), LY294002 (phosphatidylinositol 3 kinase inhibitor) and Rapamycin (mTOR inhibitor). These blockers, as well as apyrase (ATP metabolizing enzyme), also abolished the induction of FDB protein synthesis evoked by mechanical stimulation of osteoclasts in the co-culture model. Therefore, the present findings suggest that mechanically stimulated osteoclasts release ATP, leading to protein synthesis in isolated FDB muscle, by activating the P2-PI3K-Akt-mTOR pathway. These results open a new area for research and clinical interest in bone-to-muscle crosstalk in adaptive processes related to muscle use/disuse or in musculoskeletal pathologies.
Collapse
|
7
|
Jaque-Fernández F, Jorquera G, Troc-Gajardo J, Pietri-Rouxel F, Gentil C, Buvinic S, Allard B, Jaimovich E, Jacquemond V, Casas M. Pannexin-1 and CaV1.1 show reciprocal interaction during excitation-contraction and excitation-transcription coupling in skeletal muscle. J Gen Physiol 2021; 153:212695. [PMID: 34636893 PMCID: PMC8515650 DOI: 10.1085/jgp.202012635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/24/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023] Open
Abstract
One of the most important functions of skeletal muscle is to respond to nerve stimuli by contracting. This function ensures body movement but also participates in other important physiological roles, like regulation of glucose homeostasis. Muscle activity is closely regulated to adapt to different demands and shows a plasticity that relies on both transcriptional activity and nerve stimuli. These two processes, both dependent on depolarization of the plasma membrane, have so far been regarded as separated and independent processes due to a lack of evidence of common protein partners or molecular mechanisms. In this study, we reveal intimate functional interactions between the process of excitation-induced contraction and the process of excitation-induced transcriptional activity in skeletal muscle. We show that the plasma membrane voltage-sensing protein CaV1.1 and the ATP-releasing channel Pannexin-1 (Panx1) regulate each other in a reciprocal manner, playing roles in both processes. Specifically, knockdown of CaV1.1 produces chronically elevated extracellular ATP concentrations at rest, consistent with disruption of the normal control of Panx1 activity. Conversely, knockdown of Panx1 affects not only activation of transcription but also CaV1.1 function on the control of muscle fiber contraction. Altogether, our results establish the presence of bidirectional functional regulations between the molecular machineries involved in the control of contraction and transcription induced by membrane depolarization of adult muscle fibers. Our results are important for an integrative understanding of skeletal muscle function and may impact our understanding of several neuromuscular diseases.
Collapse
Affiliation(s)
- Francisco Jaque-Fernández
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Gonzalo Jorquera
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jennifer Troc-Gajardo
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - France Pietri-Rouxel
- Université Pierre et Marie Curie, Université Paris 06, Institut National de la Santé et de la Recherche Médicale/Centre National de la Recherche Scientifique/Institut de Myologie/Centre de Recherche en Myologie, Groupement hospitalier universitaire Pitié Salpêtrière, Paris, France
| | - Christel Gentil
- Université Pierre et Marie Curie, Université Paris 06, Institut National de la Santé et de la Recherche Médicale/Centre National de la Recherche Scientifique/Institut de Myologie/Centre de Recherche en Myologie, Groupement hospitalier universitaire Pitié Salpêtrière, Paris, France
| | - Sonja Buvinic
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Bruno Allard
- Université Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR-5310, Institut National de la Santé et de la Recherche Médicale U-1217, Institut NeuroMyoGène, Lyon, France
| | - Enrique Jaimovich
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Vincent Jacquemond
- Université Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique UMR-5310, Institut National de la Santé et de la Recherche Médicale U-1217, Institut NeuroMyoGène, Lyon, France
| | - Mariana Casas
- Programa de Fisiología y Biofísica, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
8
|
Lilliu E, Koenig S, Koenig X, Frieden M. Store-Operated Calcium Entry in Skeletal Muscle: What Makes It Different? Cells 2021; 10:cells10092356. [PMID: 34572005 PMCID: PMC8468011 DOI: 10.3390/cells10092356] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/03/2021] [Accepted: 09/04/2021] [Indexed: 01/26/2023] Open
Abstract
Current knowledge on store-operated Ca2+ entry (SOCE) regarding its localization, kinetics, and regulation is mostly derived from studies performed in non-excitable cells. After a long time of relative disinterest in skeletal muscle SOCE, this mechanism is now recognized as an essential contributor to muscle physiology, as highlighted by the muscle pathologies that are associated with mutations in the SOCE molecules STIM1 and Orai1. This review mainly focuses on the peculiar aspects of skeletal muscle SOCE that differentiate it from its counterpart found in non-excitable cells. This includes questions about SOCE localization and the movement of respective proteins in the highly organized skeletal muscle fibers, as well as the diversity of expressed STIM isoforms and their differential expression between muscle fiber types. The emerging evidence of a phasic SOCE, which is activated during EC coupling, and its physiological implication is described as well. The specific issues related to the use of SOCE modulators in skeletal muscles are discussed. This review highlights the complexity of SOCE activation and its regulation in skeletal muscle, with an emphasis on the most recent findings and the aim to reach a current picture of this mesmerizing phenomenon.
Collapse
Affiliation(s)
- Elena Lilliu
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Stéphane Koenig
- Department of Cell Physiology and Metabolism, University of Geneva, 1201 Geneva, Switzerland;
| | - Xaver Koenig
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria;
- Correspondence: (X.K.); (M.F.)
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, University of Geneva, 1201 Geneva, Switzerland;
- Correspondence: (X.K.); (M.F.)
| |
Collapse
|
9
|
Nemirovskaya TL. ATP-Dependent Pathways of Regulation of Skeletal Muscle Signaling and Their Interaction with Gene Expression under Unloading: The Role of “Slow” Calcium. Biophysics (Nagoya-shi) 2021. [DOI: 10.1134/s0006350921040163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
10
|
Ma B, Zhang L, Li J, Xing T, Jiang Y, Gao F. Dietary taurine supplementation ameliorates muscle loss in chronic heat stressed broilers via suppressing the perk signaling and reversing endoplasmic reticulum-stress-induced apoptosis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:2125-2134. [PMID: 32978773 DOI: 10.1002/jsfa.10835] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Heat stress seriously affects animal health and induces enormous financial losses in poultry production. Exploring the appropriate means for ameliorating unfavorable effects caused by heat stress is essential. We investigated whether taurine supplementation could attenuate breast muscle loss in chronic heat-stressed broilers, as well as its mechanism. We designed three groups: a normal control group (22 °C), a heat stress group (32 °C) and a taurine treatment group (32 °C, basal diet + 5 g·kg-1 taurine). RESULTS We found that taurine significantly moderated the decreases of breast muscle mass and yield, as well as the increases of serum aspartate aminotransferase activity and serum urine acid level in chronic heat-stressed broilers. Additionally, supplementary taurine significantly alleviated elevations of the cytoplasm Ca2+ concentration, protein expressions of GRP78 and p-PERK, mRNA expressions of Ca2+ channels (RyR1, IP3R3) and endoplasmic reticulum (ER) stress factors (GRP78, GRP94, PERK, EIF2α, ATF4, IRE1, XBP1, ATF6 and CHOP), apoptosis (Caspase-3 and TUNEL), protein catabolism, and the reduction of taurine transporter (TauT) mRNA expression in the breast muscle induced by chronic heat stress. CONCLUSION Supplementary taurine could attenuate chronic heat stress-induced breast muscle loss via reversing ER stress-induced apoptosis and suppressing protein catabolism. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Bingbing Ma
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Lin Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Jiaolong Li
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Tong Xing
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| | - Yun Jiang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Feng Gao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
11
|
Quezada ER, Díaz-Vegas A, Jaimovich E, Casas M. Changes in Gene Expression of the MCU Complex Are Induced by Electrical Stimulation in Adult Skeletal Muscle. Front Physiol 2021; 11:601313. [PMID: 33574764 PMCID: PMC7870689 DOI: 10.3389/fphys.2020.601313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/27/2020] [Indexed: 11/29/2022] Open
Abstract
The slow calcium transient triggered by low-frequency electrical stimulation (ES) in adult muscle fibers and regulated by the extracellular ATP/IP3/IP3R pathway has been related to muscle plasticity. A regulation of muscular tropism associated with the MCU has also been described. However, the role of transient cytosolic calcium signals and signaling pathways related to muscle plasticity over the regulation of gene expression of the MCU complex (MCU, MICU1, MICU2, and EMRE) in adult skeletal muscle is completely unknown. In the present work, we show that 270 0.3-ms-long pulses at 20-Hz ES (and not at 90 Hz) transiently decreased the mRNA levels of the MCU complex in mice flexor digitorum brevis isolated muscle fibers. Importantly, when ATP released after 20-Hz ES is hydrolyzed by the enzyme apyrase, the repressor effect of 20 Hz on mRNA levels of the MCU complex is lost. Accordingly, the exposure of muscle fibers to 30 μM exogenous ATP produces the same effect as 20-Hz ES. Moreover, the use of apyrase in resting conditions (without ES) increased mRNA levels of MCU, pointing out the importance of extracellular ATP concentration over MCU mRNA levels. The use of xestospongin B (inhibitor of IP3 receptors) also prevented the decrease of mRNA levels of MCU, MICU1, MICU2, and EMRE mediated by a low-frequency ES. Our results show that the MCU complex can be regulated by electrical stimuli in a frequency-dependent manner. The changes observed in mRNA levels may be related to changes in the mitochondria, associated with the phenotypic transition from a fast- to a slow-type muscle, according to the described effect of this stimulation frequency on muscle phenotype. The decrease in mRNA levels of the MCU complex by exogenous ATP and the increase in MCU levels when basal ATP is reduced with the enzyme apyrase indicate that extracellular ATP may be a regulator of the MCU complex. Moreover, our results suggest that this regulation is part of the axes linking low-frequency stimulation with ATP/IP3/IP3R.
Collapse
Affiliation(s)
- Esteban R Quezada
- Center for Exercise, Metabolism, and Cancer, Physiology and Biophysics Program, Biomedical Sciences Institute (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alexis Díaz-Vegas
- Center for Exercise, Metabolism, and Cancer, Physiology and Biophysics Program, Biomedical Sciences Institute (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Enrique Jaimovich
- Center for Exercise, Metabolism, and Cancer, Physiology and Biophysics Program, Biomedical Sciences Institute (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Mariana Casas
- Center for Exercise, Metabolism, and Cancer, Physiology and Biophysics Program, Biomedical Sciences Institute (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
12
|
Dos Santos M, Backer S, Saintpierre B, Izac B, Andrieu M, Letourneur F, Relaix F, Sotiropoulos A, Maire P. Single-nucleus RNA-seq and FISH identify coordinated transcriptional activity in mammalian myofibers. Nat Commun 2020; 11:5102. [PMID: 33037211 PMCID: PMC7547110 DOI: 10.1038/s41467-020-18789-8] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle fibers are large syncytia but it is currently unknown whether gene expression is coordinately regulated in their numerous nuclei. Here we show by snRNA-seq and snATAC-seq that slow, fast, myotendinous and neuromuscular junction myonuclei each have different transcriptional programs, associated with distinct chromatin states and combinations of transcription factors. In adult mice, identified myofiber types predominantly express either a slow or one of the three fast isoforms of Myosin heavy chain (MYH) proteins, while a small number of hybrid fibers can express more than one MYH. By snRNA-seq and FISH, we show that the majority of myonuclei within a myofiber are synchronized, coordinately expressing only one fast Myh isoform with a preferential panel of muscle-specific genes. Importantly, this coordination of expression occurs early during post-natal development and depends on innervation. These findings highlight a previously undefined mechanism of coordination of gene expression in a syncytium.
Collapse
Affiliation(s)
| | - Stéphanie Backer
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France
| | | | - Brigitte Izac
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France
| | - Muriel Andrieu
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France
| | - Franck Letourneur
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France
| | - Frederic Relaix
- Université Paris-Est Creteil, INSERM U955 IMRB., 94000, Creteil, France
| | | | - Pascal Maire
- Université de Paris, Institut Cochin, INSERM, CNRS., 75014, Paris, France.
| |
Collapse
|
13
|
Debattisti V, Horn A, Singh R, Seifert EL, Hogarth MW, Mazala DA, Huang KT, Horvath R, Jaiswal JK, Hajnóczky G. Dysregulation of Mitochondrial Ca 2+ Uptake and Sarcolemma Repair Underlie Muscle Weakness and Wasting in Patients and Mice Lacking MICU1. Cell Rep 2020; 29:1274-1286.e6. [PMID: 31665639 PMCID: PMC7007691 DOI: 10.1016/j.celrep.2019.09.063] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/07/2019] [Accepted: 09/20/2019] [Indexed: 01/29/2023] Open
Abstract
Muscle function is regulated by Ca2+, which mediates excitation-contraction coupling, energy metabolism, adaptation to exercise, and sarcolemmal repair. Several of these actions rely on Ca2+ delivery to the mitochondrial matrix via the mitochondrial Ca2+ uniporter, the pore of which is formed by mitochondrial calcium uniporter (MCU). MCU's gatekeeping and cooperative activation are controlled by MICU1. Loss-of-protein mutation in MICU1 causes a neuromuscular disease. To determine the mechanisms underlying the muscle impairments, we used MICU1 patient cells and skeletal muscle-specific MICU1 knockout mice. Both these models show a lower threshold for MCU-mediated Ca2+ uptake. Lack of MICU1 is associated with impaired mitochondrial Ca2+ uptake during excitation-contraction, aerobic metabolism impairment, muscle weakness, fatigue, and myofiber damage during physical activity. MICU1 deficit compromises mitochondrial Ca2+ uptake during sarcolemmal injury, which causes ineffective repair of the damaged myofibers. Thus, dysregulation of mitochondrial Ca2+ uptake hampers myofiber contractile function, likely through energy metabolism and membrane repair.
Collapse
Affiliation(s)
- Valentina Debattisti
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam Horn
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue Northwest, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Raghavendra Singh
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Erin L Seifert
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Marshall W Hogarth
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue Northwest, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Davi A Mazala
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue Northwest, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Kai Ting Huang
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rita Horvath
- Wellcome Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue Northwest, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA.
| | - György Hajnóczky
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Sarcoplasmic reticulum and calcium signaling in muscle cells: Homeostasis and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 350:197-264. [PMID: 32138900 DOI: 10.1016/bs.ircmb.2019.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The sarco/endoplasmic reticulum is an extensive, dynamic and heterogeneous membranous network that fulfills multiple homeostatic functions. Among them, it compartmentalizes, stores and releases calcium within the intracellular space. In the case of muscle cells, calcium released from the sarco/endoplasmic reticulum in the vicinity of the contractile machinery induces cell contraction. Furthermore, sarco/endoplasmic reticulum-derived calcium also regulates gene transcription in the nucleus, energy metabolism in mitochondria and cytosolic signaling pathways. These diverse and overlapping processes require a highly complex fine-tuning that the sarco/endoplasmic reticulum provides by means of its numerous tubules and cisternae, specialized domains and contacts with other organelles. The sarco/endoplasmic reticulum also possesses a rich calcium-handling machinery, functionally coupled to both contraction-inducing stimuli and the contractile apparatus. Such is the importance of the sarco/endoplasmic reticulum for muscle cell physiology, that alterations in its structure, function or its calcium-handling machinery are intimately associated with the development of cardiometabolic diseases. Cardiac hypertrophy, insulin resistance and arterial hypertension are age-related pathologies with a common mechanism at the muscle cell level: the accumulation of damaged proteins at the sarco/endoplasmic reticulum induces a stress response condition termed endoplasmic reticulum stress, which impairs proper organelle function, ultimately leading to pathogenesis.
Collapse
|
15
|
Del Campo A, Contreras-Hernández I, Castro-Sepúlveda M, Campos CA, Figueroa R, Tevy MF, Eisner V, Casas M, Jaimovich E. Muscle function decline and mitochondria changes in middle age precede sarcopenia in mice. Aging (Albany NY) 2019; 10:34-55. [PMID: 29302020 PMCID: PMC5811241 DOI: 10.18632/aging.101358] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/30/2017] [Indexed: 01/02/2023]
Abstract
Sarcopenia is the degenerative loss of muscle mass and strength with aging. Although a role of mitochondrial metabolism in muscle function and in the development of many diseases has been described, the role of mitochondrial topology and dynamics in the process of muscle aging is not fully understood. This work shows a time line of changes in both mitochondrial distribution and skeletal muscle function during mice lifespan. We isolated muscle fibers from flexor digitorum brevis of mice of different ages. A fusion-like phenotype of mitochondria, together with a change in orientation perpendicular to the fiber axis was evident in the Adult group compared to Juvenile and Older groups. Moreover, an increase in the contact area between sarcoplasmic reticulum and mitochondria was evident in the same group. Together with the morphological changes, mitochondrial Ca2+ resting levels were reduced at age 10-14 months and significantly increased in the Older group. This was consistent with a reduced number of mitochondria-to-jSR pairs in the Older group compared to the Juvenile. Our results support the idea of several age-dependent changes in mitochondria that are accentuated in midlife prior to a complete sarcopenic phenotype.
Collapse
Affiliation(s)
- Andrea Del Campo
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ignacio Contreras-Hernández
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Mauricio Castro-Sepúlveda
- Department of Cellular and Molecular Biology, School of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian A Campos
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Reinaldo Figueroa
- Center for Genomics and Bioinformatics, Universidad Mayor de Chile, Santiago, Chile
| | - María Florencia Tevy
- Center for Genomics and Bioinformatics, Universidad Mayor de Chile, Santiago, Chile
| | - Verónica Eisner
- Department of Cellular and Molecular Biology, School of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Casas
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Physiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Center for Exercise, Metabolism and Cancer, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
16
|
Bosutti A, Bernareggi A, Massaria G, D'Andrea P, Taccola G, Lorenzon P, Sciancalepore M. A "noisy" electrical stimulation protocol favors muscle regeneration in vitro through release of endogenous ATP. Exp Cell Res 2019; 381:121-128. [PMID: 31082374 DOI: 10.1016/j.yexcr.2019.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/17/2019] [Accepted: 05/09/2019] [Indexed: 02/04/2023]
Abstract
An in vitro system of electrical stimulation was used to explore whether an innovative "noisy" stimulation protocol derived from human electromyographic recordings (EMGstim) could promote muscle regeneration. EMGstim was delivered to cultured mouse myofibers isolated from Flexor Digitorum Brevis, preserving their satellite cells. In response to EMGstim, immunostaining for the myogenic regulatory factor myogenin, revealed an increased percentage of elongated myogenin-positive cells surrounding the myofibers. Conditioned medium collected from EMGstim-treated cell cultures, promoted satellite cells differentiation in unstimulated myofiber cell cultures, suggesting that extracellular soluble factors could mediate the process. Interestingly, the myogenic effect of EMGstim was mimicked by exogenously applied ATP (0.1 μM), reduced by the ATP diphosphohydrolase apyrase and prevented by blocking endogenous ATP release with carbenoxolone. In conclusion, our results show that "noisy" electrical stimulations favor muscle progenitor cell differentiation most likely via the release of endogenous ATP from contracting myofibres. Our data also suggest that "noisy" stimulation protocols could be potentially more efficient than regular stimulations to promote in vivo muscle regeneration after traumatic injury or in neuropathological diseases.
Collapse
Affiliation(s)
- Alessandra Bosutti
- Department of Life Sciences and Centre for Neuroscience B.R.A.I.N., University of Trieste, Via A. Fleming 22, I-34127, Trieste, Italy
| | - Annalisa Bernareggi
- Department of Life Sciences and Centre for Neuroscience B.R.A.I.N., University of Trieste, Via A. Fleming 22, I-34127, Trieste, Italy
| | - Gabriele Massaria
- Department of Life Sciences and Centre for Neuroscience B.R.A.I.N., University of Trieste, Via A. Fleming 22, I-34127, Trieste, Italy; Area Science Park, Padriciano, 99, I-34149, Trieste, Italy
| | - Paola D'Andrea
- Department of Life Sciences and Centre for Neuroscience B.R.A.I.N., University of Trieste, Via A. Fleming 22, I-34127, Trieste, Italy
| | - Giuliano Taccola
- Department of Neuroscience, SISSA, Via Bonomea 265, 34136, Trieste, Italy; SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory), Istituto di Medicina Fisica e Riabilitazione (IMFR), Via Gervasutta 48, 33100, Udine, Italy
| | - Paola Lorenzon
- Department of Life Sciences and Centre for Neuroscience B.R.A.I.N., University of Trieste, Via A. Fleming 22, I-34127, Trieste, Italy
| | - Marina Sciancalepore
- Department of Life Sciences and Centre for Neuroscience B.R.A.I.N., University of Trieste, Via A. Fleming 22, I-34127, Trieste, Italy.
| |
Collapse
|
17
|
Valladares D, Utreras-Mendoza Y, Campos C, Morales C, Diaz-Vegas A, Contreras-Ferrat A, Westermeier F, Jaimovich E, Marchi S, Pinton P, Lavandero S. IP 3 receptor blockade restores autophagy and mitochondrial function in skeletal muscle fibers of dystrophic mice. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3685-3695. [PMID: 30251688 DOI: 10.1016/j.bbadis.2018.08.042] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 08/06/2018] [Accepted: 08/30/2018] [Indexed: 12/14/2022]
Abstract
Duchenne muscular dystrophy (DMD) is characterized by a severe and progressive destruction of muscle fibers associated with altered Ca2+ homeostasis. We have previously shown that the IP3 receptor (IP3R) plays a role in elevating basal cytoplasmic Ca2+ and that pharmacological blockade of IP3R restores muscle function. Moreover, we have shown that the IP3R pathway negatively regulates autophagy by controlling mitochondrial Ca2+ levels. Nevertheless, it remains unclear whether IP3R is involved in abnormal mitochondrial Ca2+ levels, mitochondrial dynamics, or autophagy and mitophagy observed in adult DMD skeletal muscle. Here, we show that the elevated basal autophagy and autophagic flux levels were normalized when IP3R was downregulated in mdx fibers. Pharmacological blockade of IP3R in mdx fibers restored both increased mitochondrial Ca2+ levels and mitochondrial membrane potential under resting conditions. Interestingly, mdx mitochondria changed from a fission to an elongated state after IP3R knockdown, and the elevated mitophagy levels in mdx fibers were normalized. To our knowledge, this is the first study associating IP3R1 activity with changes in autophagy, mitochondrial Ca2+ levels, mitochondrial membrane potential, mitochondrial dynamics, and mitophagy in adult mouse skeletal muscle. Moreover, these results suggest that increased IP3R activity in mdx fibers plays an important role in the pathophysiology of DMD. Overall, these results lead us to propose the use of specific IP3R blockers as a new pharmacological treatment for DMD, given their ability to restore both autophagy/mitophagy and mitochondrial function.
Collapse
Affiliation(s)
- Denisse Valladares
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Escuela de Kinesiologia, Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile.
| | - Yildy Utreras-Mendoza
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Cristian Campos
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Camilo Morales
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Alexis Diaz-Vegas
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Ariel Contreras-Ferrat
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Francisco Westermeier
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Enrique Jaimovich
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Chile
| | - Saverio Marchi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
18
|
Díaz-Vegas AR, Cordova A, Valladares D, Llanos P, Hidalgo C, Gherardi G, De Stefani D, Mammucari C, Rizzuto R, Contreras-Ferrat A, Jaimovich E. Mitochondrial Calcium Increase Induced by RyR1 and IP3R Channel Activation After Membrane Depolarization Regulates Skeletal Muscle Metabolism. Front Physiol 2018; 9:791. [PMID: 29988564 PMCID: PMC6026899 DOI: 10.3389/fphys.2018.00791] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/06/2018] [Indexed: 11/13/2022] Open
Abstract
Aim: We hypothesize that both type-1 ryanodine receptor (RyR1) and IP3-receptor (IP3R) calcium channels are necessary for the mitochondrial Ca2+ increase caused by membrane depolarization induced by potassium (or by electrical stimulation) of single skeletal muscle fibers; this calcium increase would couple muscle fiber excitation to an increase in metabolic output from mitochondria (excitation-metabolism coupling). Methods: Mitochondria matrix and cytoplasmic Ca2+ levels were evaluated in fibers isolated from flexor digitorium brevis muscle using plasmids for the expression of a mitochondrial Ca2+ sensor (CEPIA3mt) or a cytoplasmic Ca2+ sensor (RCaMP). The role of intracellular Ca2+ channels was evaluated using both specific pharmacological inhibitors (xestospongin B for IP3R and Dantrolene for RyR1) and a genetic approach (shIP3R1-RFP). O2 consumption was detected using Seahorse Extracellular Flux Analyzer. Results: In isolated muscle fibers cell membrane depolarization increased both cytoplasmic and mitochondrial Ca2+ levels. Mitochondrial Ca2+ uptake required functional inositol IP3R and RyR1 channels. Inhibition of either channel decreased basal O2 consumption rate but only RyR1 inhibition decreased ATP-linked O2 consumption. Cell membrane depolarization-induced Ca2+ signals in sub-sarcolemmal mitochondria were accompanied by a reduction in mitochondrial membrane potential; Ca2+ signals propagated toward intermyofibrillar mitochondria, which displayed increased membrane potential. These results are compatible with slow, Ca2+-dependent propagation of mitochondrial membrane potential from the surface toward the center of the fiber. Conclusion: Ca2+-dependent changes in mitochondrial membrane potential have different kinetics in the surface vs. the center of the fiber; these differences are likely to play a critical role in the control of mitochondrial metabolism, both at rest and after membrane depolarization as part of an “excitation-metabolism” coupling process in skeletal muscle fibers.
Collapse
Affiliation(s)
- Alexis R Díaz-Vegas
- Muscle Physiology Laboratory, Center of Studies in Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Alex Cordova
- Biomedical Neuroscience Institute, Center of Studies in Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Denisse Valladares
- Muscle Physiology Laboratory, Center of Studies in Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Exercise and Movement Science Laboratory, Universidad Finis Terrae, Santiago, Chile
| | - Paola Llanos
- Muscle Physiology Laboratory, Center of Studies in Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Institute for Research in Dental Science, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Center of Studies in Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Gaia Gherardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Diego De Stefani
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ariel Contreras-Ferrat
- Muscle Physiology Laboratory, Center of Studies in Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Muscle Physiology Laboratory, Center of Studies in Exercise, Metabolism and Cancer, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| |
Collapse
|
19
|
Pham TL, St-Pierre ME, Ravel-Chapuis A, Parks TEC, Langlois S, Penuela S, Jasmin BJ, Cowan KN. Expression of Pannexin 1 and Pannexin 3 during skeletal muscle development, regeneration, and Duchenne muscular dystrophy. J Cell Physiol 2018; 233:7057-7070. [PMID: 29744875 DOI: 10.1002/jcp.26629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 03/30/2018] [Indexed: 01/17/2023]
Abstract
Pannexin 1 (Panx1) and Pannexin 3 (Panx3) are single membrane channels recently implicated in myogenic commitment, as well as myoblast proliferation and differentiation in vitro. However, their expression patterns during skeletal muscle development and regeneration had yet to be investigated. Here, we show that Panx1 levels increase during skeletal muscle development becoming highly expressed together with Panx3 in adult skeletal muscle. In adult mice, Panx1 and Panx3 were differentially expressed in fast- and slow-twitch muscles. We also report that Panx1/PANX1 and Panx3/PANX3 are co-expressed in mouse and human satellite cells, which play crucial roles in skeletal muscle regeneration. Interestingly, Panx1 and Panx3 levels were modulated in muscle degeneration/regeneration, similar to the pattern seen during skeletal muscle development. As Duchenne muscular dystrophy is characterized by skeletal muscle degeneration and impaired regeneration, we next used mild and severe mouse models of this disease and found a significant dysregulation of Panx1 and Panx3 levels in dystrophic skeletal muscles. Together, our results are the first demonstration that Panx1 and Panx3 are differentially expressed amongst skeletal muscle types with their levels being highly modulated during skeletal muscle development, regeneration, and dystrophy. These findings suggest that Panx1 and Panx3 channels may play important and distinct roles in healthy and diseased skeletal muscles.
Collapse
Affiliation(s)
- Tammy L Pham
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Marie-Eve St-Pierre
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Tara E C Parks
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Stéphanie Langlois
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Surgery, Division of Pediatric Surgery, University of Ottawa, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Kyle N Cowan
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Surgery, Division of Pediatric Surgery, University of Ottawa, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Navarro-Marquez M, Torrealba N, Troncoso R, Vásquez-Trincado C, Rodriguez M, Morales PE, Villalobos E, Eura Y, Garcia L, Chiong M, Klip A, Jaimovich E, Kokame K, Lavandero S. Herpud1 impacts insulin-dependent glucose uptake in skeletal muscle cells by controlling the Ca2+-calcineurin-Akt axis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1653-1662. [DOI: 10.1016/j.bbadis.2018.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 01/08/2023]
|
21
|
Szewczyk A, Gehl J, Daczewska M, Saczko J, Frandsen SK, Kulbacka J. Calcium electroporation for treatment of sarcoma in preclinical studies. Oncotarget 2018; 9:11604-11618. [PMID: 29545923 PMCID: PMC5837766 DOI: 10.18632/oncotarget.24352] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/25/2018] [Indexed: 01/08/2023] Open
Abstract
Calcium electroporation (CaEP) describes the use of electric pulses (electroporation) to transiently permeabilize cells to allow supraphysiological doses of calcium to enter the cytosol. Calcium electroporation has successfully been investigated for treatment of cutaneous metastases in a clinical study. This preclinical study explores the possible use of calcium electroporation for treatment of sarcoma. A normal murine muscle cell line (C2C12), and a human rhabdomyosarcoma cell line (RD) were used in the undifferentiated and differentiated state. Electroporation was performed using 8 pulses of 100 μs at 600–1000 V/cm; with calcium (0, 0.5, 1, and 5 mM). Viability was examined by MTS assay, intracellular calcium levels were measured, and expression of plasma membrane calcium ATPase (PMCA) was investigated using western blotting. Calcium/sodium exchanger (NCX1), ryanodine receptor (RyR1) expression and cytoskeleton structure (zyxin/actin) were assessed by immunofluorescence. CaEP efficiency on RD tumors was tested in vivo in immuno-deficient mice. CaEP was significantly more efficient in RD than in normal cells. Intracellular Ca2+ levels after CaEP increased significantly in RD, whereas a lower increase was seen in normal cells. CaEP caused decreased expression of PMCA and NCX1 in malignant cells and RyR1 in both cell lines whereas normal cells exhibited increased expression of NCX1 after CaEP. Calcium electroporation also affected cytoskeleton structure in malignant cells. This study showed that calcium electroporation is tolerated significantly better in normal muscle cells than sarcoma cells and as an inexpensive and simple cancer treatment this could potentially be used in connection with sarcoma surgery for local treatment.
Collapse
Affiliation(s)
- Anna Szewczyk
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Wroclaw, Poland
| | - Julie Gehl
- Center for Experimental Drug and Gene Electrotransfer (CEDGE), Department of Clinical Oncology and Palliative Care, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Malgorzata Daczewska
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Wroclaw, Poland
| | - Jolanta Saczko
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
| | - Stine Krog Frandsen
- Center for Experimental Drug and Gene Electrotransfer (CEDGE), Department of Clinical Oncology and Palliative Care, Zealand University Hospital, Roskilde, Denmark.,Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Julita Kulbacka
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
22
|
Sun Q, Peng Y, Qi W, Kim Y, Clark JM, Kim D, Park Y. Permethrin decreased insulin-stimulated AKT phosphorylation dependent on extracellular signal-regulated kinase-1 (ERK), but not AMP-activated protein kinase α (AMPKα), in C2C12 myotubes. Food Chem Toxicol 2017; 109:95-101. [PMID: 28866332 DOI: 10.1016/j.fct.2017.08.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 12/16/2022]
Abstract
Previously 10 μM permethrin (38.7% cis and 59.4% trans isomers), a pyrethroid insecticide widely used in agriculture and household products for pest control, was reported to reduce insulin-stimulated glucose uptake and phosphorylation of protein kinase B (p-AKT) in C2C12 mouse myotubes. The underlying mechanisms on how permethrin decreases insulin-stimulated AKT phosphorylation, however, are unknown. Thus, the goal of this study was to determine the possible mechanism(s) through which permethrin reduced insulin-stimulated AKT phosphorylation in C2C12 myotubes. Permethrin treatment, at 10 μM, decreased insulin-stimulated membrane glucose transporter type 4 (GLUT4) and AKT phosphorylation, and increased insulin receptor substrate 1 (IRS1) Ser307 phosphorylation in the presence of insulin. The inactivation of AKT by permethrin was independent of AMPKα. ERK inactivation by U0126, however, restored insulin-stimulated AKT phosphorylation, which was decreased by permethrin treatment. These results suggest that permethrin decreased insulin-stimulated AKT phosphorylation via ERK activation, but not by AMPKα inactivation.
Collapse
Affiliation(s)
| | - Ye Peng
- Department of Food Science, United States
| | - Weipeng Qi
- Department of Food Science, United States
| | - Yoo Kim
- Department of Food Science, United States
| | - John M Clark
- Department of Veterinary and Animal Sciences, United States
| | - Daeyoung Kim
- Department of Mathematics and Statistics, University of Massachusetts, Amherst, MA, 01003, United States
| | | |
Collapse
|
23
|
Santulli G, Nakashima R, Yuan Q, Marks AR. Intracellular calcium release channels: an update. J Physiol 2017; 595:3041-3051. [PMID: 28303572 PMCID: PMC5430224 DOI: 10.1113/jp272781] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/20/2017] [Indexed: 12/19/2022] Open
Abstract
Ryanodine receptors (RyRs) and inositol 1,4,5-trisphosphate receptors (IP3 Rs) are calcium (Ca2+ ) release channels on the endo/sarcoplasmic reticulum (ER/SR). Here we summarize the latest advances in the field, describing the recently discovered mechanistic roles of intracellular Ca2+ release channels in the regulation of mitochondrial fitness and endothelial function, providing novel therapeutic options for the treatment of heart failure, hypertension, and diabetes mellitus.
Collapse
Affiliation(s)
- Gaetano Santulli
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
| | - Ryutaro Nakashima
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
| | - Qi Yuan
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
| | - Andrew R. Marks
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
- Department of MedicineColumbia UniversityNew YorkNYUSA
| |
Collapse
|
24
|
A chemical chaperone improves muscle function in mice with a RyR1 mutation. Nat Commun 2017; 8:14659. [PMID: 28337975 PMCID: PMC5376670 DOI: 10.1038/ncomms14659] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 01/20/2017] [Indexed: 12/28/2022] Open
Abstract
Mutations in the RYR1 gene cause severe myopathies. Mice with an I4895T mutation in the type 1 ryanodine receptor/Ca2+ release channel (RyR1) display muscle weakness and atrophy, but the underlying mechanisms are unclear. Here we show that the I4895T mutation in RyR1 decreases the amplitude of the sarcoplasmic reticulum (SR) Ca2+ transient, resting cytosolic Ca2+ levels, muscle triadin content and calsequestrin (CSQ) localization to the junctional SR, and increases endoplasmic reticulum (ER) stress/unfolded protein response (UPR) and mitochondrial ROS production. Treatment of mice carrying the I4895T mutation with a chemical chaperone, sodium 4-phenylbutyrate (4PBA), reduces ER stress/UPR and improves muscle function, but does not restore SR Ca2+ transients in I4895T fibres to wild type levels, suggesting that decreased SR Ca2+ release is not the major driver of the myopathy. These findings suggest that 4PBA, an FDA-approved drug, has potential as a therapeutic intervention for RyR1 myopathies that are associated with ER stress. Mutations in the RyR1 channel cause core myopathies. Here the authors show that ER stress and the unfolded protein response underlie the pathology caused by a common RyR1 channel mutation, and show that treatment with a chemical chaperone restores muscle function in mice.
Collapse
|
25
|
Morales PE, Bucarey JL, Espinosa A. Muscle Lipid Metabolism: Role of Lipid Droplets and Perilipins. J Diabetes Res 2017; 2017:1789395. [PMID: 28676863 PMCID: PMC5476901 DOI: 10.1155/2017/1789395] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/19/2017] [Accepted: 04/26/2017] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle is one of the main regulators of carbohydrate and lipid metabolism in our organism, and therefore, it is highly susceptible to changes in glucose and fatty acid (FA) availability. Skeletal muscle is an extremely complex tissue: its metabolic capacity depends on the type of fibers it is made up of and the level of stimulation it undergoes, such as acute or chronic contraction. Obesity is often associated with increased FA levels, which leads to the accumulation of toxic lipid intermediates, oxidative stress, and autophagy in skeletal fibers. This lipotoxicity is one of the most common causes of insulin resistance (IR). In this scenario, the "isolation" of certain lipids in specific cell compartments, through the action of the specific lipid droplet, perilipin (PLIN) family of proteins, is conceived as a lifeguard compensatory strategy. In this review, we summarize the cellular mechanism underlying lipid mobilization and metabolism inside skeletal muscle, focusing on the function of lipid droplets, the PLIN family of proteins, and how these entities are modified in exercise, obesity, and IR conditions.
Collapse
Affiliation(s)
- Pablo Esteban Morales
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Jose Luis Bucarey
- CIDIS-AC, Escuela de Medicina, Universidad de Valparaiso, Valparaiso, Chile
| | - Alejandra Espinosa
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- *Alejandra Espinosa:
| |
Collapse
|
26
|
Arias-Calderón M, Almarza G, Díaz-Vegas A, Contreras-Ferrat A, Valladares D, Casas M, Toledo H, Jaimovich E, Buvinic S. Characterization of a multiprotein complex involved in excitation-transcription coupling of skeletal muscle. Skelet Muscle 2016; 6:15. [PMID: 27069569 PMCID: PMC4827232 DOI: 10.1186/s13395-016-0087-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/19/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Electrical activity regulates the expression of skeletal muscle genes by a process known as "excitation-transcription" (E-T) coupling. We have demonstrated that release of adenosine 5'-triphosphate (ATP) during depolarization activates membrane P2X/P2Y receptors, being the fundamental mediators between electrical stimulation, slow intracellular calcium transients, and gene expression. We propose that this signaling pathway would require the proper coordination between the voltage sensor (dihydropyridine receptor, DHPR), pannexin 1 channels (Panx1, ATP release conduit), nucleotide receptors, and other signaling molecules. The goal of this study was to assess protein-protein interactions within the E-T machinery and to look for novel constituents in order to characterize the signaling complex. METHODS Newborn derived myotubes, adult fibers, or triad fractions from rat or mouse skeletal muscles were used. Co-immunoprecipitation, 2D blue native SDS/PAGE, confocal microscopy z-axis reconstruction, and proximity ligation assays were combined to assess the physical proximity of the putative complex interactors. An L6 cell line overexpressing Panx1 (L6-Panx1) was developed to study the influence of some of the complex interactors in modulation of gene expression. RESULTS Panx1, DHPR, P2Y2 receptor (P2Y2R), and dystrophin co-immunoprecipitated in the different preparations assessed. 2D blue native SDS/PAGE showed that DHPR, Panx1, P2Y2R and caveolin-3 (Cav3) belong to the same multiprotein complex. We observed co-localization and protein-protein proximity between DHPR, Panx1, P2Y2R, and Cav3 in adult fibers and in the L6-Panx1 cell line. We found a very restricted location of Panx1 and Cav3 in a putative T-tubule zone near the sarcolemma, while DHPR was highly expressed all along the transverse (T)-tubule. By Panx1 overexpression, extracellular ATP levels were increased both at rest and after electrical stimulation. Basal mRNA levels of the early gene cfos and the oxidative metabolism markers citrate synthase and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) were significantly increased by Panx1 overexpression. Interleukin 6 expression evoked by 20-Hz electrical stimulation (270 pulses, 0.3 ms each) was also significantly upregulated in L6-Panx1 cells. CONCLUSIONS We propose the existence of a relevant multiprotein complex that coordinates events involved in E-T coupling. Unveiling the molecular actors involved in the regulation of gene expression will contribute to the understanding and treatment of skeletal muscle disorders due to wrong-expressed proteins, as well as to improve skeletal muscle performance.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Animals
- Animals, Newborn
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Caveolin 3/genetics
- Caveolin 3/metabolism
- Cell Line
- Connexins/genetics
- Connexins/metabolism
- Dystrophin/genetics
- Dystrophin/metabolism
- Electric Stimulation
- Gene Expression Regulation
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Multiprotein Complexes
- Muscle Contraction
- Muscle Fibers, Skeletal/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Protein Binding
- Rats, Wistar
- Receptors, Purinergic P2Y2/genetics
- Receptors, Purinergic P2Y2/metabolism
- Transcription, Genetic
- Transcriptional Activation
- Transfection
Collapse
Affiliation(s)
- Manuel Arias-Calderón
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
- />Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492 Santiago, Chile
| | - Gonzalo Almarza
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Alexis Díaz-Vegas
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Ariel Contreras-Ferrat
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Denisse Valladares
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Mariana Casas
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Héctor Toledo
- />Programa de Biología Molecular y Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Enrique Jaimovich
- />Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
- />Programa de Biología Molecular y Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, 8380453 Chile
| | - Sonja Buvinic
- />Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492 Santiago, Chile
| |
Collapse
|
27
|
Hernández-Ochoa EO, Vanegas C, Iyer SR, Lovering RM, Schneider MF. Alternating bipolar field stimulation identifies muscle fibers with defective excitability but maintained local Ca(2+) signals and contraction. Skelet Muscle 2016; 6:6. [PMID: 26855765 PMCID: PMC4743112 DOI: 10.1186/s13395-016-0076-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/05/2016] [Indexed: 11/25/2022] Open
Abstract
Background Most cultured enzymatically dissociated adult myofibers exhibit spatially uniform (UNI) contractile responses and Ca2+ transients over the entire myofiber in response to electric field stimuli of either polarity applied via bipolar electrodes. However, some myofibers only exhibit contraction and Ca2+ transients at alternating (ALT) ends in response to alternating polarity field stimulation. Here, we present for the first time the methodology for identification of ALT myofibers in primary cultures and isolated muscles, as well as a study of their electrophysiological properties. Results We used high-speed confocal microscopic Ca2+ imaging, electric field stimulation, microelectrode recordings, immunostaining, and confocal microscopy to characterize the properties of action potential-induced Ca2+ transients, contractility, resting membrane potential, and staining of T-tubule voltage-gated Na+ channel distribution applied to cultured adult myofibers. Here, we show for the first time, with high temporal and spatial resolution, that normal control myofibers with UNI responses can be converted to ALT response myofibers by TTX addition or by removal of Na+ from the bathing medium, with reappearance of the UNI response on return of Na+. Our results suggest disrupted excitability as the cause of ALT behavior and indicate that the ALT response is due to local depolarization-induced Ca2+ release, whereas the UNI response is triggered by action potential propagation over the entire myofiber. Consistent with this interpretation, local depolarizing monopolar stimuli give uniform (propagated) responses in UNI myofibers, but only local responses at the electrode in ALT myofibers. The ALT responses in electrically inexcitable myofibers are consistent with expectations of current spread between bipolar stimulating electrodes, entering (hyperpolarizing) one end of a myofiber and leaving (depolarizing) the other end of the myofiber. ALT responses were also detected in some myofibers within intact isolated whole muscles from wild-type and MDX mice, demonstrating that ALT responses can be present before enzymatic dissociation. Conclusions We suggest that checking for ALT myofiber responsiveness by looking at the end of a myofiber during alternating polarity stimuli provides a test for compromised excitability of myofibers, and could be used to identify inexcitable, damaged or diseased myofibers by ALT behavior in healthy and diseased muscle. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0076-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erick O Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, 108 N. Greene Street, Baltimore, MD 21201 USA
| | - Camilo Vanegas
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, 108 N. Greene Street, Baltimore, MD 21201 USA
| | - Shama R Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, 108 N. Greene Street, Baltimore, MD 21201 USA
| |
Collapse
|
28
|
Mishra P, Varuzhanyan G, Pham AH, Chan DC. Mitochondrial Dynamics is a Distinguishing Feature of Skeletal Muscle Fiber Types and Regulates Organellar Compartmentalization. Cell Metab 2015; 22:1033-44. [PMID: 26603188 PMCID: PMC4670593 DOI: 10.1016/j.cmet.2015.09.027] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/08/2015] [Accepted: 09/23/2015] [Indexed: 01/18/2023]
Abstract
Skeletal muscle fibers differentiate into specific fiber types with distinct metabolic properties determined by their reliance on oxidative phosphorylation (OXPHOS). Using in vivo approaches, we find that OXPHOS-dependent fibers, compared to glycolytic fibers, contain elongated mitochondrial networks with higher fusion rates that are dependent on the mitofusins Mfn1 and Mfn2. Switching of a glycolytic fiber to an oxidative IIA type is associated with elongation of mitochondria, suggesting that mitochondrial fusion is linked to metabolic state. Furthermore, we reveal that mitochondrial proteins are compartmentalized to discrete domains centered around their nuclei of origin. The domain dimensions are dependent on fiber type and are regulated by the mitochondrial dynamics proteins Mfn1, Mfn2, and Mff. Our results indicate that mitochondrial dynamics is tailored to fiber type physiology and provides a rationale for the segmental defects characteristic of aged and diseased muscle fibers.
Collapse
Affiliation(s)
- Prashant Mishra
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Grigor Varuzhanyan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Anh H Pham
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
29
|
Georgiev T, Svirin M, Jaimovich E, Fink RHA. Localized nuclear and perinuclear Ca(2+) signals in intact mouse skeletal muscle fibers. Front Physiol 2015; 6:263. [PMID: 26483696 PMCID: PMC4586431 DOI: 10.3389/fphys.2015.00263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/07/2015] [Indexed: 11/13/2022] Open
Abstract
Nuclear Ca2+ is important for the regulation of several nuclear processes such as gene expression. Localized Ca2+ signals (LCSs) in skeletal muscle fibers of mice have been mainly studied as Ca2+ release events from the sarcoplasmic reticulum. Their location with regard to cell nuclei has not been investigated. Our study is based on the hypothesis that LCSs associated with nuclei are present in skeletal muscle fibers of adult mice. Therefore, we carried out experiments addressing this question and we found novel Ca2+ signals associated with nuclei of skeletal muscle fibers (with possibly attached satellite cells). We measured localized nuclear and perinuclear Ca2+ signals (NLCSs and PLCSs) alongside cytosolic localized Ca2+ signals (CLCSs) during a hypertonic treatment. We also observed NLCSs under isotonic conditions. The NLCSs and PLCSs are Ca2+ signals in the range of micrometer [FWHM (full width at half maximum): 2.75 ± 0.27 μm (NLCSs) and 2.55 ± 0.17 μm (PLCSs), S.E.M.]. Additionally, global nuclear Ca2+ signals (NGCSs) were observed. To investigate which type of Ca2+ channels contribute to the Ca2+ signals associated with nuclei in skeletal muscle fibers, we performed measurements with the RyR blocker dantrolene, the DHPR blocker nifedipine or the IP3R blocker Xestospongin C. We observed Ca2+ signals associated with nuclei in the presence of each blocker. Nifedipine and dantrolene had an inhibitory effect on the fraction of fibers with PLCSs. The situation for the fraction of fibers with NLCSs is more complex indicating that RyR is less important for the generation of NLCSs compared to the generation of PLCSs. The fraction of fibers with NLCSs and PLCSs is not reduced in the presence of Xestospongin C. The localized perinuclear and intranuclear Ca2+ signals may be a powerful tool for the cell to regulate adaptive processes as gene expression. The intranuclear Ca2+ signals may be particularly interesting in this respect.
Collapse
Affiliation(s)
- Tihomir Georgiev
- Medical Biophysics Unit, Institut für Physiologie und Pathophysiologie, Ruprecht Karls Universität Heidelberg, Germany ; Facultad de Medicina, Center for Molecular Studies of the Cell, Universidad de Chile Santiago de Chile, Chile
| | - Mikhail Svirin
- Medical Biophysics Unit, Institut für Physiologie und Pathophysiologie, Ruprecht Karls Universität Heidelberg, Germany
| | - Enrique Jaimovich
- Facultad de Medicina, Center for Molecular Studies of the Cell, Universidad de Chile Santiago de Chile, Chile
| | - Rainer H A Fink
- Medical Biophysics Unit, Institut für Physiologie und Pathophysiologie, Ruprecht Karls Universität Heidelberg, Germany
| |
Collapse
|
30
|
Phosphoinositides in Ca(2+) signaling and excitation-contraction coupling in skeletal muscle: an old player and newcomers. J Muscle Res Cell Motil 2015; 36:491-9. [PMID: 26377756 DOI: 10.1007/s10974-015-9422-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/09/2015] [Indexed: 10/23/2022]
Abstract
Since the postulate, 30 years ago, that phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P 2) as the precursor of inositol 1,4,5-trisphosphate (Ins(1,4,5)P 3) would be critical for skeletal muscle excitation-contraction (EC) coupling, the issue of whether phosphoinositides (PtdInsPs) may have something to do with Ca(2+) signaling in muscle raised limited interest, if any. In recent years however, the PtdInsP world has expanded considerably with new functions for PtdIns(4,5)P 2 but also with functions for the other members of the PtdInsP family. In this context, the discovery that genetic deficiency in a PtdInsP phosphatase has dramatic consequences on Ca(2+) homeostasis in skeletal muscle came unanticipated and opened up new perspectives in regards to how PtdInsPs modulate muscle Ca(2+) signaling under normal and disease conditions. This review intends to make an update of the established, the questioned, and the unknown regarding the role of PtdInsPs in skeletal muscle Ca(2+) homeostasis and EC coupling, with very specific emphasis given to Ca(2+) signals in differentiated skeletal muscle fibers.
Collapse
|
31
|
Enríquez V, Granados S, Arias MP, Calderón JC. Muscle Fiber Types of Gluteus Medius in the Colombian Creole Horse. J Equine Vet Sci 2015. [DOI: 10.1016/j.jevs.2015.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Berthier C, Kutchukian C, Bouvard C, Okamura Y, Jacquemond V. Depression of voltage-activated Ca2+ release in skeletal muscle by activation of a voltage-sensing phosphatase. J Gen Physiol 2015; 145:315-30. [PMID: 25825170 PMCID: PMC4380211 DOI: 10.1085/jgp.201411309] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 03/05/2015] [Indexed: 12/30/2022] Open
Abstract
Phosphoinositides act as signaling molecules in numerous cellular transduction processes, and phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) regulates the function of several types of plasma membrane ion channels. We investigated the potential role of PtdIns(4,5)P2 in Ca(2+) homeostasis and excitation-contraction (E-C) coupling of mouse muscle fibers using in vivo expression of the voltage-sensing phosphatases (VSPs) Ciona intestinalis VSP (Ci-VSP) or Danio rerio VSP (Dr-VSP). Confocal images of enhanced green fluorescent protein-tagged Dr-VSP revealed a banded pattern consistent with VSP localization within the transverse tubule membrane. Rhod-2 Ca(2+) transients generated by 0.5-s-long voltage-clamp depolarizing pulses sufficient to elicit Ca(2+) release from the sarcoplasmic reticulum (SR) but below the range at which VSPs are activated were unaffected by the presence of the VSPs. However, in Ci-VSP-expressing fibers challenged by 5-s-long depolarizing pulses, the Ca(2+) level late in the pulse (3 s after initiation) was significantly lower at 120 mV than at 20 mV. Furthermore, Ci-VSP-expressing fibers showed a reversible depression of Ca(2+) release during trains, with the peak Ca(2+) transient being reduced by ∼30% after the application of 10 200-ms-long pulses to 100 mV. A similar depression was observed in Dr-VSP-expressing fibers. Cav1.1 Ca(2+) channel-mediated current was unaffected by Ci-VSP activation. In fibers expressing Ci-VSP and a pleckstrin homology domain fused with monomeric red fluorescent protein (PLCδ1PH-mRFP), depolarizing pulses elicited transient changes in mRFP fluorescence consistent with release of transverse tubule-bound PLCδ1PH domain into the cytosol; the voltage sensitivity of these changes was consistent with that of Ci-VSP activation, and recovery occurred with a time constant in the 10-s range. Our results indicate that the PtdIns(4,5)P2 level is tightly maintained in the transverse tubule membrane of the muscle fibers, and that VSP-induced depletion of PtdIns(4,5)P2 impairs voltage-activated Ca(2+) release from the SR. Because Ca(2+) release is thought to be independent from InsP3 signaling, the effect likely results from an interaction between PtdIns(4,5)P2 and a protein partner of the E-C coupling machinery.
Collapse
Affiliation(s)
- Christine Berthier
- Centre National de la Recherche Scientifique UMR 5534, Université Lyon 1, Centre de Génétique et de Physiologie Moléculaire et Cellulaire, 69100 Villeurbanne, France
| | - Candice Kutchukian
- Centre National de la Recherche Scientifique UMR 5534, Université Lyon 1, Centre de Génétique et de Physiologie Moléculaire et Cellulaire, 69100 Villeurbanne, France
| | - Clément Bouvard
- Centre National de la Recherche Scientifique UMR 5534, Université Lyon 1, Centre de Génétique et de Physiologie Moléculaire et Cellulaire, 69100 Villeurbanne, France
| | - Yasushi Okamura
- Laboratory of Integrative Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Vincent Jacquemond
- Centre National de la Recherche Scientifique UMR 5534, Université Lyon 1, Centre de Génétique et de Physiologie Moléculaire et Cellulaire, 69100 Villeurbanne, France
| |
Collapse
|
33
|
Park DR, Park KH, Kim BJ, Yoon CS, Kim UH. Exercise ameliorates insulin resistance via Ca2+ signals distinct from those of insulin for GLUT4 translocation in skeletal muscles. Diabetes 2015; 64:1224-34. [PMID: 25409702 DOI: 10.2337/db14-0939] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Muscle contraction and insulin induce glucose uptake in skeletal muscle through GLUT4 membrane translocation. Beneficial effects of exercise on glucose homeostasis in insulin-resistant individuals are known to be due to their distinct mechanism between contraction and insulin action on glucose uptake in skeletal muscle. However, the underlying mechanisms are not clear. Here we show that in skeletal muscle, distinct Ca(2+) second messengers regulate GLUT4 translocation by contraction and insulin treatment; d-myo-inositol 1,4,5-trisphosphate/nicotinic acid adenine dinucleotide phosphate (NAADP) and cyclic ADP-ribose/NAADP are main players for insulin- and contraction-induced glucose uptake, respectively. Different patterns of phosphorylation of AMPK and Ca(2+)/calmodulin-dependent protein kinase II were shown in electrical stimuli (ES)- and insulin-induced glucose uptake pathways. ES-induced Ca(2+) signals and glucose uptake are dependent on glycolysis, which influences formation of NAD(P)-derived signaling messengers, whereas insulin-induced signals are not. High-fat diet (HFD) induced a defect in only insulin-mediated, but not ES-mediated, Ca(2+) signaling for glucose uptake, which is related to a specifically lower NAADP formation. Exercise decreases blood glucose levels in HFD-induced insulin resistance mice via NAADP formation. Thus we conclude that different usage of Ca(2+) signaling in contraction/insulin-stimulated glucose uptake in skeletal muscle may account for the mechanism by which exercise ameliorates glucose homeostasis in individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Dae-Ryoung Park
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Korea National Creative Research Laboratory for Ca Signaling Network, Chonbuk National University, Jeonju, Korea
| | - Kwang-Hyun Park
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Korea National Creative Research Laboratory for Ca Signaling Network, Chonbuk National University, Jeonju, Korea
| | - Byung-Ju Kim
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Korea National Creative Research Laboratory for Ca Signaling Network, Chonbuk National University, Jeonju, Korea
| | - Chung-Su Yoon
- Department of Physical Education, Chonbuk National University, Jeonju, Korea
| | - Uh-Hyun Kim
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Korea National Creative Research Laboratory for Ca Signaling Network, Chonbuk National University, Jeonju, Korea Institute of Cardiovascular Research, Chonbuk National University, Jeonju, Korea
| |
Collapse
|
34
|
Llanos P, Contreras-Ferrat A, Georgiev T, Osorio-Fuentealba C, Espinosa A, Hidalgo J, Hidalgo C, Jaimovich E. The cholesterol-lowering agent methyl-β-cyclodextrin promotes glucose uptake via GLUT4 in adult muscle fibers and reduces insulin resistance in obese mice. Am J Physiol Endocrinol Metab 2015; 308:E294-305. [PMID: 25491723 DOI: 10.1152/ajpendo.00189.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Insulin stimulates glucose uptake in adult skeletal muscle by promoting the translocation of GLUT4 glucose transporters to the transverse tubule (T-tubule) membranes, which have particularly high cholesterol levels. We investigated whether T-tubule cholesterol content affects insulin-induced glucose transport. Feeding mice a high-fat diet (HFD) for 8 wk increased by 30% the T-tubule cholesterol content of triad-enriched vesicular fractions from muscle tissue compared with triads from control mice. Additionally, isolated muscle fibers (flexor digitorum brevis) from HFD-fed mice showed a 40% decrease in insulin-stimulated glucose uptake rates compared with fibers from control mice. In HFD-fed mice, four subcutaneous injections of MβCD, an agent reported to extract membrane cholesterol, improved their defective glucose tolerance test and normalized their high fasting glucose levels. The preincubation of isolated muscle fibers with relatively low concentrations of MβCD increased both basal and insulin-induced glucose uptake in fibers from controls or HFD-fed mice and decreased Akt phosphorylation without altering AMPK-mediated signaling. In fibers from HFD-fed mice, MβCD improved insulin sensitivity even after Akt or CaMK II inhibition and increased membrane GLUT4 content. Indinavir, a GLUT4 antagonist, prevented the stimulatory effects of MβCD on glucose uptake. Addition of MβCD elicited ryanodine receptor-mediated calcium signals in isolated fibers, which were essential for glucose uptake. Our findings suggest that T-tubule cholesterol content exerts a critical regulatory role on insulin-stimulated GLUT4 translocation and glucose transport and that partial cholesterol removal from muscle fibers may represent a useful strategy to counteract insulin resistance.
Collapse
Affiliation(s)
- Paola Llanos
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Santiago, Chile;
| | - Ariel Contreras-Ferrat
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Tihomir Georgiev
- Medical Biophysics, Institute of Physiology und Pathophysiology, Ruprecht Karls Universität, Heidelberg, Germany
| | | | - Alejandra Espinosa
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Jorge Hidalgo
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Physiology and Biophysics Program, Institute of Biomedical Sciences (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Physiology and Biophysics Program, Institute of Biomedical Sciences (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile; Biomedical Neuroscience Institute, Facultad de Medicina, Universidad de Chile, Santiago, Chile; and
| | - Enrique Jaimovich
- Center for Molecular Studies of the Cell, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Cell and Molecular Biology Program, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
35
|
Casas M, Buvinic S, Jaimovich E. ATP signaling in skeletal muscle: from fiber plasticity to regulation of metabolism. Exerc Sport Sci Rev 2014; 42:110-6. [PMID: 24949845 DOI: 10.1249/jes.0000000000000017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Tetanic electrical stimulation releases adenosine triphosphate (ATP) from muscle fibers through pannexin-1 channels in a frequency-dependent manner; extracellular ATP activates signals that ultimately regulate gene expression and is able to increase glucose transport through activation of P2Y receptors, phosphatidylinositol 3-kinase, Akt, and AS160. We hypothesize that this mechanism is an important link between exercise and the regulation of muscle fiber plasticity and metabolism.
Collapse
Affiliation(s)
- Mariana Casas
- 1Center for Molecular Studies of the Cell, Biomedical Sciences Institute, Faculty of Medicine, Universidad de Chile; and 2Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | | | | |
Collapse
|
36
|
Eisner V, Lenaers G, Hajnóczky G. Mitochondrial fusion is frequent in skeletal muscle and supports excitation-contraction coupling. ACTA ACUST UNITED AC 2014; 205:179-95. [PMID: 24751540 PMCID: PMC4003250 DOI: 10.1083/jcb.201312066] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mitochondrial fusion is frequent in skeletal muscle, and its disruption jeopardizes excitation–contraction coupling and may contribute to the pathology of myopathies. Genetic targeting experiments indicate a fundamental role for mitochondrial fusion proteins in mammalian physiology. However, owing to the multiple functions of fusion proteins, their related phenotypes are not necessarily caused by altered mitochondrial fusion. Perhaps the biggest mystery is presented by skeletal muscle, where mostly globular-shaped mitochondria are densely packed into the narrow intermyofilamental space, limiting the interorganellar interactions. We show here that mitochondria form local networks and regularly undergo fusion events to share matrix content in skeletal muscle fibers. However, fusion events are less frequent and more stable in the fibers than in nondifferentiated myoblasts. Complementation among muscle mitochondria was suppressed by both in vivo genetic perturbations and chronic alcohol consumption that cause myopathy. An Mfn1-dependent pathway is revealed whereby fusion inhibition weakens the metabolic reserve of mitochondria to cause dysregulation of calcium oscillations during prolonged stimulation. Thus, fusion dynamically connects skeletal muscle mitochondria and its prolonged loss jeopardizes bioenergetics and excitation–contraction coupling, providing a potential pathomechanism contributing to myopathies.
Collapse
Affiliation(s)
- Verónica Eisner
- MitoCare Center, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | | | | |
Collapse
|
37
|
Bustamante M, Fernández-Verdejo R, Jaimovich E, Buvinic S. Electrical stimulation induces IL-6 in skeletal muscle through extracellular ATP by activating Ca(2+) signals and an IL-6 autocrine loop. Am J Physiol Endocrinol Metab 2014; 306:E869-82. [PMID: 24518675 PMCID: PMC3989743 DOI: 10.1152/ajpendo.00450.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interleukin-6 (IL-6) is an important myokine that is highly expressed in skeletal muscle cells upon exercise. We assessed IL-6 expression in response to electrical stimulation (ES) or extracellular ATP as a known mediator of the excitation-transcription mechanism in skeletal muscle. We examined whether the canonical signaling cascade downstream of IL-6 (IL-6/JAK2/STAT3) also responds to muscle cell excitation, concluding that IL-6 influences its own expression through a positive loop. Either ES or exogenous ATP (100 μM) increased both IL-6 expression and p-STAT3 levels in rat myotubes, a process inhibited by 100 μM suramin and 2 U/ml apyrase. ATP also evoked IL-6 expression in both isolated skeletal fibers and extracts derived from whole FDB muscles. ATP increased IL-6 release up to 10-fold. STAT3 activation evoked by ATP was abolished by the JAK2 inhibitor HBC. Blockade of secreted IL-6 with a neutralizing antibody or preincubation with the STAT3 inhibitor VIII reduced STAT3 activation evoked by extracellular ATP by 70%. Inhibitor VIII also reduced by 70% IL-6 expression evoked by ATP, suggesting a positive IL-6 loop. In addition, ATP increased up to 60% the protein levels of SOCS3, a negative regulator of the IL-6 signaling pathway. On the other hand, intracellular calcium chelation or blockade of IP3-dependent calcium signals abolished STAT3 phosphorylation evoked by either extracellular ATP or ES. These results suggest that expression of IL-6 in stimulated skeletal muscle cells is mediated by extracellular ATP and nucleotide receptors, involving IP3-dependent calcium signals as an early step that triggers a positive IL-6 autocrine loop.
Collapse
Affiliation(s)
- Mario Bustamante
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; and
| | | | | | | |
Collapse
|
38
|
Calderón JC, Bolaños P, Caputo C. The excitation-contraction coupling mechanism in skeletal muscle. Biophys Rev 2014; 6:133-160. [PMID: 28509964 PMCID: PMC5425715 DOI: 10.1007/s12551-013-0135-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 12/06/2013] [Indexed: 12/27/2022] Open
Abstract
First coined by Alexander Sandow in 1952, the term excitation-contraction coupling (ECC) describes the rapid communication between electrical events occurring in the plasma membrane of skeletal muscle fibres and Ca2+ release from the SR, which leads to contraction. The sequence of events in twitch skeletal muscle involves: (1) initiation and propagation of an action potential along the plasma membrane, (2) spread of the potential throughout the transverse tubule system (T-tubule system), (3) dihydropyridine receptors (DHPR)-mediated detection of changes in membrane potential, (4) allosteric interaction between DHPR and sarcoplasmic reticulum (SR) ryanodine receptors (RyR), (5) release of Ca2+ from the SR and transient increase of Ca2+ concentration in the myoplasm, (6) activation of the myoplasmic Ca2+ buffering system and the contractile apparatus, followed by (7) Ca2+ disappearance from the myoplasm mediated mainly by its reuptake by the SR through the SR Ca2+ adenosine triphosphatase (SERCA), and under several conditions movement to the mitochondria and extrusion by the Na+/Ca2+ exchanger (NCX). In this text, we review the basics of ECC in skeletal muscle and the techniques used to study it. Moreover, we highlight some recent advances and point out gaps in knowledge on particular issues related to ECC such as (1) DHPR-RyR molecular interaction, (2) differences regarding fibre types, (3) its alteration during muscle fatigue, (4) the role of mitochondria and store-operated Ca2+ entry in the general ECC sequence, (5) contractile potentiators, and (6) Ca2+ sparks.
Collapse
Affiliation(s)
- Juan C Calderón
- Physiology and Biochemistry Research Group-Physis, Department of Physiology and Biochemistry, Faculty of Medicine, University of Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia.
- Laboratory of Cellular Physiology, Centre of Biophysics and Biochemistry, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela.
- Departamento de Fisiología y Bioquímica, Grupo de Investigación en Fisiología y Bioquímica-Physis, Facultad de Medicina, Universidad de Antioquia, Calle 70 No 52-21, Medellín, Colombia.
| | - Pura Bolaños
- Laboratory of Cellular Physiology, Centre of Biophysics and Biochemistry, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela
| | - Carlo Caputo
- Laboratory of Cellular Physiology, Centre of Biophysics and Biochemistry, Venezuelan Institute for Scientific Research (IVIC), Caracas, Venezuela
| |
Collapse
|
39
|
Valladares D, Almarza G, Contreras A, Pavez M, Buvinic S, Jaimovich E, Casas M. Electrical stimuli are anti-apoptotic in skeletal muscle via extracellular ATP. Alteration of this signal in Mdx mice is a likely cause of dystrophy. PLoS One 2013; 8:e75340. [PMID: 24282497 PMCID: PMC3839923 DOI: 10.1371/journal.pone.0075340] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/11/2013] [Indexed: 12/20/2022] Open
Abstract
ATP signaling has been shown to regulate gene expression in skeletal muscle and to be altered in models of muscular dystrophy. We have previously shown that in normal muscle fibers, ATP released through Pannexin1 (Panx1) channels after electrical stimulation plays a role in activating some signaling pathways related to gene expression. We searched for a possible role of ATP signaling in the dystrophy phenotype. We used muscle fibers from flexor digitorum brevis isolated from normal and mdx mice. We demonstrated that low frequency electrical stimulation has an anti-apoptotic effect in normal muscle fibers repressing the expression of Bax, Bim and PUMA. Addition of exogenous ATP to the medium has a similar effect. In dystrophic fibers, the basal levels of extracellular ATP were higher compared to normal fibers, but unlike control fibers, they do not present any ATP release after low frequency electrical stimulation, suggesting an uncoupling between electrical stimulation and ATP release in this condition. Elevated levels of Panx1 and decreased levels of Cav1.1 (dihydropyridine receptors) were found in triads fractions prepared from mdx muscles. Moreover, decreased immunoprecipitation of Cav1.1 and Panx1, suggest uncoupling of the signaling machinery. Importantly, in dystrophic fibers, exogenous ATP was pro-apoptotic, inducing the transcription of Bax, Bim and PUMA and increasing the levels of activated Bax and cytosolic cytochrome c. These evidence points to an involvement of the ATP pathway in the activation of mechanisms related with cell death in muscular dystrophy, opening new perspectives towards possible targets for pharmacological therapies.
Collapse
Affiliation(s)
- Denisse Valladares
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gonzalo Almarza
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ariel Contreras
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mario Pavez
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sonja Buvinic
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Ciencias Básicas y Comunitarias, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mariana Casas
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
40
|
Osorio-Fuentealba C, Contreras-Ferrat AE, Altamirano F, Espinosa A, Li Q, Niu W, Lavandero S, Klip A, Jaimovich E. Electrical stimuli release ATP to increase GLUT4 translocation and glucose uptake via PI3Kγ-Akt-AS160 in skeletal muscle cells. Diabetes 2013; 62:1519-26. [PMID: 23274898 PMCID: PMC3636621 DOI: 10.2337/db12-1066] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Skeletal muscle glucose uptake in response to exercise is preserved in insulin-resistant conditions, but the signals involved are debated. ATP is released from skeletal muscle by contractile activity and can autocrinely signal through purinergic receptors, and we hypothesized it may influence glucose uptake. Electrical stimulation, ATP, and insulin each increased fluorescent 2-NBD-Glucose (2-NBDG) uptake in primary myotubes, but only electrical stimulation and ATP-dependent 2-NBDG uptake were inhibited by adenosine-phosphate phosphatase and by purinergic receptor blockade (suramin). Electrical stimulation transiently elevated extracellular ATP and caused Akt phosphorylation that was additive to insulin and inhibited by suramin. Exogenous ATP transiently activated Akt and, inhibiting phosphatidylinositol 3-kinase (PI3K) or Akt as well as dominant-negative Akt mutant, reduced ATP-dependent 2-NBDG uptake and Akt phosphorylation. ATP-dependent 2-NBDG uptake was also inhibited by the G protein βγ subunit-interacting peptide βark-ct and by the phosphatidylinositol 3-kinase-γ (PI3Kγ) inhibitor AS605240. ATP caused translocation of GLUT4myc-eGFP to the cell surface, mechanistically mediated by increased exocytosis involving AS160/Rab8A reduced by dominant-negative Akt or PI3Kγ kinase-dead mutants, and potentiated by myristoylated PI3Kγ. ATP stimulated 2-NBDG uptake in normal and insulin-resistant adult muscle fibers, resembling the reported effect of exercise. Hence, the ATP-induced pathway may be tapped to bypass insulin resistance.
Collapse
Affiliation(s)
- Cesar Osorio-Fuentealba
- Center for Molecular Studies of the Cell, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Ariel E. Contreras-Ferrat
- Center for Molecular Studies of the Cell, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Francisco Altamirano
- Center for Molecular Studies of the Cell, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Alejandra Espinosa
- Center for Molecular Studies of the Cell, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
- School of Medical Technology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Qing Li
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Wenyan Niu
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Sergio Lavandero
- Center for Molecular Studies of the Cell, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
- Faculty of Pharmaceutical and Chemical Sciences, Universidad de Chile, Santiago, Chile
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Enrique Jaimovich
- Center for Molecular Studies of the Cell, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
- Corresponding author: Enrique Jaimovich,
| |
Collapse
|
41
|
Valdés JA, Flores S, Fuentes EN, Osorio-Fuentealba C, Jaimovich E, Molina A. IGF-1 induces IP3-dependent calcium signal involved in the regulation of myostatin gene expression mediated by NFAT during myoblast differentiation. J Cell Physiol 2013; 228:1452-63. [DOI: 10.1002/jcp.24298] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 11/27/2012] [Indexed: 12/20/2022]
|
42
|
Jorquera G, Altamirano F, Contreras-Ferrat A, Almarza G, Buvinic S, Jacquemond V, Jaimovich E, Casas M. Cav1.1 controls frequency-dependent events regulating adult skeletal muscle plasticity. J Cell Sci 2013; 126:1189-98. [PMID: 23321639 DOI: 10.1242/jcs.116855] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
An important pending question in neuromuscular biology is how skeletal muscle cells decipher the stimulation pattern coming from motoneurons to define their phenotype as slow or fast twitch muscle fibers. We have previously shown that voltage-gated L-type calcium channel (Cav1.1) acts as a voltage sensor for activation of inositol (1,4,5)-trisphosphate [Ins(1,4,5)P₃]-dependent Ca(2+) signals that regulates gene expression. ATP released by muscle cells after electrical stimulation through pannexin-1 channels plays a key role in this process. We show now that stimulation frequency determines both ATP release and Ins(1,4,5)P₃ production in adult skeletal muscle and that Cav1.1 and pannexin-1 colocalize in the transverse tubules. Both ATP release and increased Ins(1,4,5)P₃ was seen in flexor digitorum brevis fibers stimulated with 270 pulses at 20 Hz, but not at 90 Hz. 20 Hz stimulation induced transcriptional changes related to fast-to-slow muscle fiber phenotype transition that required ATP release. Addition of 30 µM ATP to fibers induced the same transcriptional changes observed after 20 Hz stimulation. Myotubes lacking the Cav1.1-α1 subunit released almost no ATP after electrical stimulation, showing that Cav1.1 has a central role in this process. In adult muscle fibers, ATP release and the transcriptional changes produced by 20 Hz stimulation were blocked by both the Cav1.1 antagonist nifedipine (25 µM) and by the Cav1.1 agonist (-)S-BayK 8644 (10 µM). We propose a new role for Cav1.1, independent of its calcium channel activity, in the activation of signaling pathways allowing muscle fibers to decipher the frequency of electrical stimulation and to activate specific transcriptional programs that define their phenotype.
Collapse
Affiliation(s)
- Gonzalo Jorquera
- Centro de Estudios Moleculares de Célula, ICBM, Facultad de Medicina, Universidad de Chile, Independencia 1027-8380453, Santiago, Chile
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Tjondrokoesoemo A, Li N, Lin PH, Pan Z, Ferrante CJ, Shirokova N, Brotto M, Weisleder N, Ma J. Type 1 inositol (1,4,5)-trisphosphate receptor activates ryanodine receptor 1 to mediate calcium spark signaling in adult mammalian skeletal muscle. J Biol Chem 2012; 288:2103-9. [PMID: 23223241 DOI: 10.1074/jbc.m112.425975] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Functional coupling between inositol (1,4,5)-trisphosphate receptor (IP(3)R) and ryanodine receptor (RyR) represents a critical component of intracellular Ca(2+) signaling in many excitable cells; however, the role of this mechanism in skeletal muscle remains elusive. In skeletal muscle, RyR-mediated Ca(2+) sparks are suppressed in resting conditions, whereas application of transient osmotic stress can trigger activation of Ca(2+) sparks that are restricted to the periphery of the fiber. Here we show that onset of these spatially confined Ca(2+) sparks involves interaction between activation of IP(3)R and RyR near the sarcolemmal membrane. Pharmacological prevention of IP(3) production or inhibition of IP(3)R channel activity abolishes stress-induced Ca(2+) sparks in skeletal muscle. Although genetic ablation of the type 2 IP(3)R does not appear to affect Ca(2+) sparks in skeletal muscle, specific silencing of the type 1 IP(3)R leads to ablation of stress-induced Ca(2+) sparks. Our data indicate that membrane-delimited signaling involving cross-talk between IP(3)R1 and RyR1 contributes to Ca(2+) spark activation in skeletal muscle.
Collapse
Affiliation(s)
- Andoria Tjondrokoesoemo
- Department of Physiology and Biophysics, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Cea LA, Riquelme MA, Cisterna BA, Puebla C, Vega JL, Rovegno M, Sáez JC. Connexin- and pannexin-based channels in normal skeletal muscles and their possible role in muscle atrophy. J Membr Biol 2012; 245:423-36. [PMID: 22850938 DOI: 10.1007/s00232-012-9485-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/28/2012] [Indexed: 12/13/2022]
Abstract
Precursor cells of skeletal muscles express connexins 39, 43 and 45 and pannexin1. In these cells, most connexins form two types of membrane channels, gap junction channels and hemichannels, whereas pannexin1 forms only hemichannels. All these channels are low-resistance pathways permeable to ions and small molecules that coordinate developmental events. During late stages of skeletal muscle differentiation, myofibers become innervated and stop expressing connexins but still express pannexin1 hemichannels that are potential pathways for the ATP release required for potentiation of the contraction response. Adult injured muscles undergo regeneration, and connexins are reexpressed and form membrane channels. In vivo, connexin reexpression occurs in undifferentiated cells that form new myofibers, favoring the healing process of injured muscle. However, differentiated myofibers maintained in culture for 48 h or treated with proinflammatory cytokines for less than 3 h also reexpress connexins and only form functional hemichannels at the cell surface. We propose that opening of these hemichannels contributes to drastic changes in electrochemical gradients, including reduction of membrane potential, increases in intracellular free Ca(2+) concentration and release of diverse metabolites (e.g., NAD(+) and ATP) to the extracellular milieu, contributing to multiple metabolic and physiologic alterations that characterize muscles undergoing atrophy in several acquired and genetic human diseases. Consequently, inhibition of connexin hemichannels expressed by injured or denervated skeletal muscles might reduce or prevent deleterious changes triggered by conditions that promote muscle atrophy.
Collapse
Affiliation(s)
- Luis A Cea
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile,
| | | | | | | | | | | | | |
Collapse
|
45
|
Blaauw B, Del Piccolo P, Rodriguez L, Hernandez Gonzalez VH, Agatea L, Solagna F, Mammano F, Pozzan T, Schiaffino S. No evidence for inositol 1,4,5-trisphosphate-dependent Ca2+ release in isolated fibers of adult mouse skeletal muscle. ACTA ACUST UNITED AC 2012; 140:235-41. [PMID: 22802359 PMCID: PMC3409103 DOI: 10.1085/jgp.201110747] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The presence and role of functional inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs) in adult skeletal muscle are controversial. The current consensus is that, in adult striated muscle, the relative amount of IP3Rs is too low and the kinetics of Ca2+ release from IP3R is too slow compared with ryanodine receptors to contribute to the Ca2+ transient during excitation–contraction coupling. However, it has been suggested that IP3-dependent Ca2+ release may be involved in signaling cascades leading to regulation of muscle gene expression. We have reinvestigated IP3-dependent Ca2+ release in isolated flexor digitorum brevis (FDB) muscle fibers from adult mice. Although Ca2+ transients were readily induced in cultured C2C12 muscle cells by (a) UTP stimulation, (b) direct injection of IP3, or (c) photolysis of membrane-permeant caged IP3, no statistically significant change in calcium signal was detected in adult FDB fibers. We conclude that the IP3–IP3R system does not appear to affect global calcium levels in adult mouse skeletal muscle.
Collapse
Affiliation(s)
- Bert Blaauw
- Venetian Institute of Molecular Medicine, 35129 Padova, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Analysis of spontaneous and nerve-evoked calcium transients in intact extraocular muscles in vitro. Exp Eye Res 2012; 100:73-85. [PMID: 22579493 DOI: 10.1016/j.exer.2012.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Revised: 03/29/2012] [Accepted: 04/18/2012] [Indexed: 12/22/2022]
Abstract
Extraocular muscles (EOMs) have unique calcium handling properties, yet little is known about the dynamics of calcium events underlying ultrafast and tonic contractions in myofibers of intact EOMs. Superior oblique EOMs of juvenile chickens were dissected with their nerve attached, maintained in oxygenated Krebs buffer, and loaded with fluo-4. Spontaneous and nerve stimulation-evoked calcium transients were recorded and, following calcium imaging, some EOMs were double-labeled with rhodamine-conjugated alpha-bungarotoxin (rhBTX) to identify EOM myofiber types. EOMs showed two main types of spontaneous calcium transients, one slow type (calcium waves with 1/2(max) duration of 2-12 s, velocity of 25-50 μm/s) and two fast "flash-like" types (Type 1, 30-90 ms; Type 2, 90-150 ms 1/2(max) duration). Single pulse nerve stimulation evoked fast calcium transients identical to the fast (Type 1) calcium transients. Calcium waves were accompanied by a local myofiber contraction that followed the calcium transient wavefront. The magnitude of calcium-wave induced myofiber contraction far exceeded those of movement induced by nerve stimulation and associated fast calcium transients. Tetrodotoxin eliminated nerve-evoked transients, but not spontaneous transients. Alpha-bungarotoxin eliminated both spontaneous and nerve-evoked fast calcium transients, but not calcium waves, and caffeine increased wave activity. Calcium waves were observed in myofibers lacking spontaneous or evoked fast transients, suggestive of multiply-innervated myofibers, and this was confirmed by double-labeling with rhBTX. We propose that the abundant spontaneous calcium transients and calcium waves with localized contractions that do not depend on innervation may contribute to intrinsic generation of tonic functions of EOMs.
Collapse
|
47
|
Altamirano F, López JR, Henríquez C, Molinski T, Allen PD, Jaimovich E. Increased resting intracellular calcium modulates NF-κB-dependent inducible nitric-oxide synthase gene expression in dystrophic mdx skeletal myotubes. J Biol Chem 2012; 287:20876-87. [PMID: 22549782 DOI: 10.1074/jbc.m112.344929] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder caused by dystrophin mutations, characterized by chronic inflammation and severe muscle wasting. Dystrophic muscles exhibit activated immune cell infiltrates, up-regulated inflammatory gene expression, and increased NF-κB activity, but the contribution of the skeletal muscle cell to this process has been unclear. The aim of this work was to study the pathways that contribute to the increased resting calcium ([Ca(2+)](rest)) observed in mdx myotubes and its possible link with up-regulation of NF-κB and pro-inflammatory gene expression in dystrophic muscle cells. [Ca(2+)](rest) was higher in mdx than in WT myotubes (308 ± 6 versus 113 ± 2 nm, p < 0.001). In mdx myotubes, both the inhibition of Ca(2+) entry (low Ca(2+) solution, Ca(2+)-free solution, and Gd(3+)) and blockade of either ryanodine receptors or inositol 1,4,5-trisphosphate receptors reduced [Ca(2+)](rest). Basal activity of NF-κB was significantly up-regulated in mdx versus WT myotubes. There was an increased transcriptional activity and p65 nuclear localization, which could be reversed when [Ca(2+)](rest) was reduced. Levels of mRNA for TNFα, IL-1β, and IL-6 were similar in WT and mdx myotubes, whereas inducible nitric-oxide synthase (iNOS) expression was increased 5-fold. Reducing [Ca(2+)](rest) using different strategies reduced iNOS gene expression presumably as a result of decreased activation of NF-κB. We propose that NF-κB, modulated by increased [Ca(2+)](rest), is constitutively active in mdx myotubes, and this mechanism can account for iNOS overexpression and the increase in reactive nitrogen species that promote damage in dystrophic skeletal muscle cells.
Collapse
Affiliation(s)
- Francisco Altamirano
- From the Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile
| | | | | | | | | | | |
Collapse
|
48
|
Measurement of calcium release due to inositol trisphosphate receptors in skeletal muscle. Methods Mol Biol 2012; 798:383-93. [PMID: 22130849 DOI: 10.1007/978-1-61779-343-1_22] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Calcium transients elicited by IP(3) receptors upon electrical stimulation of skeletal muscle cells (slow calcium signals) are often hard to visualize due to their relatively small amplitude compared to the large transient originated from ryanodine receptors associated to excitation-contraction coupling. The study of slow calcium transients, however, is relevant due to their function in regulation of muscle gene expression and in the process of excitation-transcription coupling. Discussed here are the procedures used to record slow calcium signals from both cultured mouse myotubes and from cultured adult skeletal muscle fibers.
Collapse
|
49
|
Vicencio JM, Estrada M, Galvis D, Bravo R, Contreras AE, Rotter D, Szabadkai G, Hill JA, Rothermel BA, Jaimovich E, Lavandero S. Anabolic androgenic steroids and intracellular calcium signaling: a mini review on mechanisms and physiological implications. Mini Rev Med Chem 2011; 11:390-8. [PMID: 21443511 DOI: 10.2174/138955711795445880] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 01/21/2011] [Indexed: 02/05/2023]
Abstract
Increasing evidence suggests that nongenomic effects of testosterone and anabolic androgenic steroids (AAS) operate concertedly with genomic effects. Classically, these responses have been viewed as separate and independent processes, primarily because nongenomic responses are faster and appear to be mediated by membrane androgen receptors, whereas long-term genomic effects are mediated through cytosolic androgen receptors regulating transcriptional activity. Numerous studies have demonstrated increases in intracellular Ca2+ in response to AAS. These Ca2+ mediated responses have been seen in a diversity of cell types, including osteoblasts, platelets, skeletal muscle cells, cardiac myocytes and neurons. The versatility of Ca2+ as a second messenger provides these responses with a vast number of pathophysiological implications. In cardiac cells, testosterone elicits voltage-dependent Ca2+ oscillations and IP3R-mediated Ca2+ release from internal stores, leading to activation of MAPK and mTOR signaling that promotes cardiac hypertrophy. In neurons, depending upon concentration, testosterone can provoke either physiological Ca2+ oscillations, essential for synaptic plasticity, or sustained, pathological Ca2+ transients that lead to neuronal apoptosis. We propose therefore, that Ca2+ acts as an important point of crosstalk between nongenomic and genomic AAS signaling, representing a central regulator that bridges these previously thought to be divergent responses.
Collapse
Affiliation(s)
- J M Vicencio
- Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences/Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|