1
|
Franco-Obregón A, Tai YK. Are Aminoglycoside Antibiotics TRPing Your Metabolic Switches? Cells 2024; 13:1273. [PMID: 39120305 PMCID: PMC11311832 DOI: 10.3390/cells13151273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
Transient receptor potential (TRP) channels are broadly implicated in the developmental programs of most tissues. Amongst these tissues, skeletal muscle and adipose are noteworthy for being essential in establishing systemic metabolic balance. TRP channels respond to environmental stimuli by supplying intracellular calcium that instigates enzymatic cascades of developmental consequence and often impinge on mitochondrial function and biogenesis. Critically, aminoglycoside antibiotics (AGAs) have been shown to block the capacity of TRP channels to conduct calcium entry into the cell in response to a wide range of developmental stimuli of a biophysical nature, including mechanical, electromagnetic, thermal, and chemical. Paradoxically, in vitro paradigms commonly used to understand organismal muscle and adipose development may have been led astray by the conventional use of streptomycin, an AGA, to help prevent bacterial contamination. Accordingly, streptomycin has been shown to disrupt both in vitro and in vivo myogenesis, as well as the phenotypic switch of white adipose into beige thermogenic status. In vivo, streptomycin has been shown to disrupt TRP-mediated calcium-dependent exercise adaptations of importance to systemic metabolism. Alternatively, streptomycin has also been used to curb detrimental levels of calcium leakage into dystrophic skeletal muscle through aberrantly gated TRPC1 channels that have been shown to be involved in the etiology of X-linked muscular dystrophies. TRP channels susceptible to AGA antagonism are critically involved in modulating the development of muscle and adipose tissues that, if administered to behaving animals, may translate to systemwide metabolic disruption. Regenerative medicine and clinical communities need to be made aware of this caveat of AGA usage and seek viable alternatives, to prevent contamination or infection in in vitro and in vivo paradigms, respectively.
Collapse
Affiliation(s)
- Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zürich, 8057 Zürich, Switzerland
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Institute of Health Technology and Innovation (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- BICEPS Lab (Biolonic Currents Electromagnetic Pulsing Systems), National University of Singapore, Singapore 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
2
|
Nguyen J, Gilbert PM. Decoding the forces that shape muscle stem cell function. Curr Top Dev Biol 2024; 158:279-306. [PMID: 38670710 DOI: 10.1016/bs.ctdb.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Skeletal muscle is a force-producing organ composed of muscle tissues, connective tissues, blood vessels, and nerves, all working in synergy to enable movement and provide support to the body. While robust biomechanical descriptions of skeletal muscle force production at the body or tissue level exist, little is known about force application on microstructures within the muscles, such as cells. Among various cell types, skeletal muscle stem cells reside in the muscle tissue environment and play a crucial role in driving the self-repair process when muscle damage occurs. Early evidence indicates that the fate and function of skeletal muscle stem cells are controlled by both biophysical and biochemical factors in their microenvironments, but much remains to accomplish in quantitatively describing the biophysical muscle stem cell microenvironment. This book chapter aims to review current knowledge on the influence of biophysical stresses and landscape properties on muscle stem cells in heath, aging, and diseases.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Tranter JD, Kumar A, Nair VK, Sah R. Mechanosensing in Metabolism. Compr Physiol 2023; 14:5269-5290. [PMID: 38158369 PMCID: PMC11681368 DOI: 10.1002/cphy.c230005] [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] [Indexed: 01/03/2024]
Abstract
Electrical mechanosensing is a process mediated by specialized ion channels, gated directly or indirectly by mechanical forces, which allows cells to detect and subsequently respond to mechanical stimuli. The activation of mechanosensitive (MS) ion channels, intrinsically gated by mechanical forces, or mechanoresponsive (MR) ion channels, indirectly gated by mechanical forces, results in electrical signaling across lipid bilayers, such as the plasma membrane. While the functions of mechanically gated channels within a sensory context (e.g., proprioception and touch) are well described, there is emerging data demonstrating functions beyond touch and proprioception, including mechanoregulation of intracellular signaling and cellular/systemic metabolism. Both MR and MS ion channel signaling have been shown to contribute to the regulation of metabolic dysfunction, including obesity, insulin resistance, impaired insulin secretion, and inflammation. This review summarizes our current understanding of the contributions of several MS/MR ion channels in cell types implicated in metabolic dysfunction, namely, adipocytes, pancreatic β-cells, hepatocytes, and skeletal muscle cells, and discusses MS/MR ion channels as possible therapeutic targets. © 2024 American Physiological Society. Compr Physiol 14:5269-5290, 2024.
Collapse
Affiliation(s)
- John D. Tranter
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ashutosh Kumar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Vinayak K. Nair
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Cardiovascular Research, Washington University, St. Louis, Missouri, USA
- St. Louis VA Medical Center, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Abstract
All cells in the body are exposed to physical force in the form of tension, compression, gravity, shear stress, or pressure. Cells convert these mechanical cues into intracellular biochemical signals; this process is an inherent property of all cells and is essential for numerous cellular functions. A cell's ability to respond to force largely depends on the array of mechanical ion channels expressed on the cell surface. Altered mechanosensing impairs conscious senses, such as touch and hearing, and unconscious senses, like blood pressure regulation and gastrointestinal (GI) activity. The GI tract's ability to sense pressure changes and mechanical force is essential for regulating motility, but it also underlies pain originating in the GI tract. Recent identification of the mechanically activated ion channels Piezo1 and Piezo2 in the gut and the effects of abnormal ion channel regulation on cellular function indicate that these channels may play a pathogenic role in disease. Here, we discuss our current understanding of mechanically activated Piezo channels in the pathogenesis of pancreatic and GI diseases, including pancreatitis, diabetes mellitus, irritable bowel syndrome, GI tumors, and inflammatory bowel disease. We also describe how Piezo channels could be important targets for treating GI diseases.
Collapse
|
5
|
Mechanotransduction for Muscle Protein Synthesis via Mechanically Activated Ion Channels. Life (Basel) 2023; 13:life13020341. [PMID: 36836698 PMCID: PMC9962945 DOI: 10.3390/life13020341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Cell mechanotransduction, the ability to detect physical forces and convert them into a series of biochemical events, is important for a wide range of physiological processes. Cells express an array of mechanosensors transducing physical forces into intracellular signaling cascades, including ion channels. Ion channels that can be directly activated by mechanical cues are known as mechanically activated (MA), or stretch-activated (SA), channels. In response to repeated exposures to mechanical stimulation in the form of resistance training, enhanced protein synthesis and fiber hypertrophy are elicited in skeletal muscle, whereas a lack of mechanical stimuli due to inactivity/mechanical unloading leads to reduced muscle protein synthesis and fiber atrophy. To date, the role of MA channels in the transduction of mechanical load to intracellular signaling pathways regulating muscle protein synthesis is poorly described. This review article will discuss MA channels in striated muscle, their regulation, and putative roles in the anabolic processes in muscle cells/fibers in response to mechanical stimuli.
Collapse
|
6
|
A preliminary study on the role of Piezo1 channels in myokine release from cultured mouse myotubes. Biochem Biophys Res Commun 2022; 623:148-153. [DOI: 10.1016/j.bbrc.2022.07.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/14/2022] [Indexed: 11/20/2022]
|
7
|
Yoshihara T, Dobashi S, Takaragawa M, Naito H. Effect of losartan treatment on Smad signaling and recovery from hindlimb unloading-induced soleus muscle atrophy in female rats. Eur J Pharmacol 2022; 931:175223. [PMID: 35988789 DOI: 10.1016/j.ejphar.2022.175223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/08/2022] [Accepted: 08/16/2022] [Indexed: 11/29/2022]
Abstract
Losartan, an angiotensin II type 1 receptor blocker, exerts protective effect on soleus muscle atrophy in female rats. Thus, we aimed to examine the effect of losartan treatment on the recovery of atrophied soleus muscles. Female Wistar rats were subjected to hindlimb unloading for 7 d and then reloading for 7 d with either phosphate-buffered saline (PBS; n = 9) or losartan (40 mg/kg/day; n = 9). The soleus muscles were removed at rest (sedentary control [SED]; n = 9), after 7 d of hindlimb unloading (HU; n = 9), and after 7 d of reloading (HUR-PBS or HUR-LOS; n = 9 each). The absolute and relative weights, and fiber cross-sectional area (CSA) of the soleus muscles of rats in the HU group were significantly reduced as compared to those of the rats in the SED group at 7 d post-hindlimb unloading. Seven days of reloading significantly increased the muscle weights of rats in the HUR-PBS and HUR-LOS groups, with the recovery rate of the absolute muscle weight and type I fiber CSA being significantly higher in the HUR-LOS group (6.1% and 10.1%, respectively) than in the HUR-PBS group (4.7% and 5.2%, respectively) (p < 0.05). Moreover, the absolute and relative muscle weight in HUR-PBS were lower than SED; however, no significant difference was observed between the SED and HUR-LOS groups. CSAs of type I and IIa fiber were significantly higher in the HUR-LOS group than in the HU group. Losartan administration during reloading resulted in increased Smad1/5/8 and mTOR signaling and decreased Smad2/3 signaling and protein ubiquitination, facilitating the recovery of atrophied soleus muscle. Therefore, losartan administration-induced muscle recovery may partially be attributed to enhanced Smad1/5/8 and mTOR signaling activation, and reduced activation of canonical TGF-β signaling (Smad2/3) in the soleus muscle.
Collapse
Affiliation(s)
- Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan.
| | - Shohei Dobashi
- Institute of Health and Sports Science & Medicine, Juntendo University, Chiba, Japan
| | - Mizuki Takaragawa
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan; Institute of Health and Sports Science & Medicine, Juntendo University, Chiba, Japan
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan; Institute of Health and Sports Science & Medicine, Juntendo University, Chiba, Japan
| |
Collapse
|
8
|
García-Castañeda M, Michelucci A, Zhao N, Malik S, Dirksen RT. Postdevelopmental knockout of Orai1 improves muscle pathology in a mouse model of Duchenne muscular dystrophy. J Gen Physiol 2022; 154:213383. [PMID: 35939054 PMCID: PMC9365874 DOI: 10.1085/jgp.202213081] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), an X-linked disorder caused by loss-of-function mutations in the dystrophin gene, is characterized by progressive muscle degeneration and weakness. Enhanced store-operated Ca2+ entry (SOCE), a Ca2+ influx mechanism coordinated by STIM1 sensors of luminal Ca2+ within the sarcoplasmic reticulum (SR) and Ca2+-permeable Orai1 channels in the sarcolemma, is proposed to contribute to Ca2+-mediated muscle damage in DMD. To directly determine the impact of Orai1-dependent SOCE on the dystrophic phenotype, we crossed mdx mice with tamoxifen-inducible, muscle-specific Orai1 knockout mice (mdx-Orai1 KO mice). Both constitutive and SOCE were significantly increased in flexor digitorum brevis fibers from mdx mice, while SOCE was absent in fibers from both Orai1 KO and mdx-Orai1 KO mice. Compared with WT mice, fibers from mdx mice exhibited (1) increased resting myoplasmic Ca2+ levels, (2) reduced total releasable Ca2+ store content, and (3) a prolonged rate of electrically evoked Ca2+ transient decay. These effects were partially normalized in fibers from mdx-Orai1 KO mice. Intact extensor digitorum longus muscles from mdx mice exhibited a significant reduction of maximal specific force, which was rescued in muscles from mdx-Orai1 KO mice. Finally, during exposure to consecutive eccentric contractions, muscles from mdx mice displayed a more pronounced decline in specific force compared with that of WT mice, which was also significantly attenuated by Orai1 ablation. Together, these results indicate that enhanced Orai1-dependent SOCE exacerbates the dystrophic phenotype and that Orai1 deficiency improves muscle pathology by both normalizing Ca2+ homeostasis and promoting sarcolemmal integrity/stability.
Collapse
Affiliation(s)
- Maricela García-Castañeda
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Antonio Michelucci
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY,Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Nan Zhao
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Robert T. Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
9
|
Kurth F, Tai YK, Parate D, van Oostrum M, Schmid YRF, Toh SJ, Yap JLY, Wollscheid B, Othman A, Dittrich PS, Franco-Obregón A. Cell-Derived Vesicles as TRPC1 Channel Delivery Systems for the Recovery of Cellular Respiratory and Proliferative Capacities. ACTA ACUST UNITED AC 2020; 4:e2000146. [PMID: 32875708 DOI: 10.1002/adbi.202000146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/28/2020] [Indexed: 11/07/2022]
Abstract
Pulsed electromagnetic fields (PEMFs) are capable of specifically activating a TRPC1-mitochondrial axis underlying cell expansion and mitohormetic survival adaptations. This study characterizes cell-derived vesicles (CDVs) generated from C2C12 murine myoblasts and shows that they are equipped with the sufficient molecular machinery to confer mitochondrial respiratory capacity and associated proliferative responses upon their fusion with recipient cells. CDVs derived from wild type C2C12 myoblasts include the cation-permeable transient receptor potential (TRP) channels, TRPC1 and TRPA1, and directly respond to PEMF exposure with TRPC1-mediated calcium entry. By contrast, CDVs derived from C2C12 muscle cells in which TRPC1 has been genetically knocked-down using CRISPR/Cas9 genome editing, do not. Wild type C2C12-derived CDVs are also capable of restoring PEMF-induced proliferative and mitochondrial activation in two C2C12-derived TRPC1 knockdown clonal cell lines in accordance to their endogenous degree of TRPC1 suppression. C2C12 wild type CDVs respond to menthol with calcium entry and accumulation, likewise verifying TRPA1 functional gating and further corroborating compartmental integrity. Proteomic and lipidomic analyses confirm the surface membrane origin of the CDVs providing an initial indication of the minimal cellular machinery required to recover mitochondrial function. CDVs hence possess the potential of restoring respiratory and proliferative capacities to senescent cells and tissues.
Collapse
Affiliation(s)
- Felix Kurth
- Department of Biosystems Science and Engineering, Bioanalytics Group, ETH Zurich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Dinesh Parate
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Marc van Oostrum
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Otto-Stern-Weg 3, Zurich, 8093, Switzerland
| | - Yannick R F Schmid
- Department of Biosystems Science and Engineering, Bioanalytics Group, ETH Zurich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Shi Jie Toh
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Jasmine Lye Yee Yap
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Bernd Wollscheid
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Otto-Stern-Weg 3, Zurich, 8093, Switzerland
| | - Alaa Othman
- Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, Otto-Stern-Weg 3, Zurich, 8093, Switzerland.,Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland.,Institute for Clinical Chemistry, University Hospital Zurich, Zurich, 8091, Switzerland
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, Bioanalytics Group, ETH Zurich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore.,Institute for Health Innovation & Technology, iHealthtech, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| |
Collapse
|
10
|
Yap JLY, Tai YK, Fröhlich J, Fong CHH, Yin JN, Foo ZL, Ramanan S, Beyer C, Toh SJ, Casarosa M, Bharathy N, Kala MP, Egli M, Taneja R, Lee CN, Franco-Obregón A. Ambient and supplemental magnetic fields promote myogenesis via a TRPC1-mitochondrial axis: evidence of a magnetic mitohormetic mechanism. FASEB J 2019; 33:12853-12872. [PMID: 31518158 DOI: 10.1096/fj.201900057r] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We show that both supplemental and ambient magnetic fields modulate myogenesis. A lone 10 min exposure of myoblasts to 1.5 mT amplitude supplemental pulsed magnetic fields (PEMFs) accentuated in vitro myogenesis by stimulating transient receptor potential (TRP)-C1-mediated calcium entry and downstream nuclear factor of activated T cells (NFAT)-transcriptional and P300/CBP-associated factor (PCAF)-epigenetic cascades, whereas depriving myoblasts of ambient magnetic fields slowed myogenesis, reduced TRPC1 expression, and silenced NFAT-transcriptional and PCAF-epigenetic cascades. The expression levels of peroxisome proliferator-activated receptor γ coactivator 1α, the master regulator of mitochondriogenesis, was also enhanced by brief PEMF exposure. Accordingly, mitochondriogenesis and respiratory capacity were both enhanced with PEMF exposure, paralleling TRPC1 expression and pharmacological sensitivity. Clustered regularly interspaced short palindromic repeats-Cas9 knockdown of TRPC1 precluded proliferative and mitochondrial responses to supplemental PEMFs, whereas small interfering RNA gene silencing of TRPM7 did not, coinciding with data that magnetoreception did not coincide with the expression or function of other TRP channels. The aminoglycoside antibiotics antagonized and down-regulated TRPC1 expression and, when applied concomitantly with PEMF exposure, attenuated PEMF-stimulated calcium entry, mitochondrial respiration, proliferation, differentiation, and epigenetic directive in myoblasts, elucidating why the developmental potential of magnetic fields may have previously escaped detection. Mitochondrial-based survival adaptations were also activated upon PEMF stimulation. Magnetism thus deploys an authentic myogenic directive that relies on an interplay between mitochondria and TRPC1 to reach fruition.-Yap, J. L. Y., Tai, Y. K., Fröhlich, J., Fong, C. H. H., Yin, J. N., Foo, Z. L., Ramanan, S., Beyer, C., Toh, S. J., Casarosa, M., Bharathy, N., Kala, M. P., Egli, M., Taneja, R., Lee, C. N., Franco-Obregón, A. Ambient and supplemental magnetic fields promote myogenesis via a TRPC1-mitochondrial axis: evidence of a magnetic mitohormetic mechanism.
Collapse
Affiliation(s)
- Jasmine Lye Yee Yap
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Jürg Fröhlich
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute for Electromagnetic Fields, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Charlene Hui Hua Fong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Jocelyn Naixin Yin
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Zi Ling Foo
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Sharanya Ramanan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Christian Beyer
- Institute for Electromagnetic Fields, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland.,Centre Suisse d'Électronique et de Microtechnique (CSEM SA), Neuchâtel, Switzerland
| | - Shi Jie Toh
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore
| | - Marco Casarosa
- Department of Experimental and Clinical Biomedical Sciences Mario Serio, University of Florence, Florence, Italy
| | - Narendra Bharathy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | - Monica Palanichamy Kala
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Marcel Egli
- Institute of Medical Engineering, Lucerne University of Applied Sciences and Arts, Hergiswil, Switzerland; and
| | - Reshma Taneja
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chuen Neng Lee
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute for Health Innovation and Technology, iHealthtech, National University of Singapore, Singapore
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,BioIonic Currents Electromagnetic Pulsing Systems (BICEPS) Laboratory, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute for Health Innovation and Technology, iHealthtech, National University of Singapore, Singapore
| |
Collapse
|
11
|
An Anabolic Signaling Response of Rat Soleus Muscle to Eccentric Contractions Following Hindlimb Unloading: A Potential Role of Stretch-Activated Ion Channels. Int J Mol Sci 2019; 20:ijms20051165. [PMID: 30866432 PMCID: PMC6429234 DOI: 10.3390/ijms20051165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/02/2019] [Accepted: 03/04/2019] [Indexed: 12/03/2022] Open
Abstract
Mechanisms that convert a mechanical signal into a biochemical response in an atrophied skeletal muscle remain poorly understood. The aims of the study were to evaluate a temporal response of anabolic signaling and protein synthesis (PS) to eccentric contractions (EC) in rat soleus during hindlimb unloading (HU); and to assess a possible role of stretch-activated ion channels (SAC) in the propagation of a mechanical signal to mTORC1 following HU. Following HU, an isolated soleus was subjected to EC. Upon completion of EC, muscles were collected for western blot analyses to determine the content/phosphorylation of the key anabolic markers. We found that a degree of EC-induced p70S6K phosphorylation and the rate of PS in the soleus of 3- and 7-day unloaded rats was significantly less than that in control. A decrease in EC-induced phosphorylation of p70S6K, RPS6 and PS in the 7-day unloaded soleus treated with SAC inhibitor did not differ from that of the 7-day unloaded soleus without SAC blockade. The results of the study suggest that (i) HU results in a blunted anabolic response to a bout of EC, (ii) attenuation of mTORC1-signaling and PS in response to EC in unloaded soleus may be associated with inactivation of SAC.
Collapse
|
12
|
Mirzoev TM, Tyganov SA, Petrova IO, Shenkman BS. Acute recovery from disuse atrophy: the role of stretch-activated ion channels in the activation of anabolic signaling in skeletal muscle. Am J Physiol Endocrinol Metab 2019; 316:E86-E95. [PMID: 30457911 DOI: 10.1152/ajpendo.00261.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The aim of the study was to 1) measure time-course alternations in the rate of protein synthesis (PS) and phosphorylation status of the key anabolic markers, and 2) find out the role of stretch-activated ion channels (SACs) in the activation of anabolic signaling in the rat soleus during an acute reloading following disuse atrophy. Wistar rats were subjected to 14-day hindlimb suspension (HS) followed by 6, 12, and 24 h of reloading. To examine the role of SAC in the reloading-induced activation of anabolic signaling, the rats were treated with gadolinium (Gd3+), a SAC blocker. The content of signaling proteins was determined by Western blot. c-Myc mRNA expression was assessed by RT-PCR. After 24-h reloading, the PS rate was elevated by 44% versus control. After 6-h reloading, the p-70-kDa ribosomal protein S6 kinase (p70S6k) and translation initiation factor 4E-binding protein 1 (4E-BP1) did not differ from control; however, 12-h reloading resulted in an upregulation of both p70s6k and 4E-BP1 phosphorylation versus control. The phosphorylation of AKT (Ser473) and glycogen synthase kinase-3β (Ser9) was reduced after HS and then completely restored by 12-h reloading. c-Myc was significantly upregulated during the entire reloading. Gd3+ treatment during reloading (12 h) prevented a full phosphorylation of p70S6k, rpS6, 4E-BP1, as well as PS activation. The results of the study suggest that 1) enhanced PS during the acute recovery from HS may be associated with the activation of ribosome biogenesis as well as mammalian target of rapamycin complex 1 (mTORC1)-dependent signaling pathways, and 2) functional SACs are necessary for complete activation of mTORC1 signaling in rat soleus during acute recovery from HS.
Collapse
Affiliation(s)
- Timur M Mirzoev
- Myology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Sergey A Tyganov
- Myology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Irina O Petrova
- Myology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| | - Boris S Shenkman
- Myology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences , Moscow , Russia
| |
Collapse
|
13
|
Li EW, McKee-Muir OC, Gilbert PM. Cellular Biomechanics in Skeletal Muscle Regeneration. Curr Top Dev Biol 2018; 126:125-176. [DOI: 10.1016/bs.ctdb.2017.08.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
14
|
Sturgeon RM, Magoski NS. Diacylglycerol-mediated regulation of Aplysia bag cell neuron excitability requires protein kinase C. J Physiol 2016; 594:5573-92. [PMID: 27198498 DOI: 10.1113/jp272152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/17/2016] [Indexed: 01/15/2023] Open
Abstract
KEY POINTS In Aplysia, reproduction is initiated by the bag cell neurons and a prolonged period of enhanced excitability known as the afterdischarge. Phosphoinositide turnover is upregulated during the afterdischarge resulting in the hydrolysis of phosphatidylinositol-4,5-bisphosphate by phospholipase C (PLC) and the release of diacylglycerol (DAG) and inositol trisphosphate (IP3 ). In whole-cell voltage-clamped cultured bag cell neurons, 1-oleoyl-2-acetyl-sn-glycerol (OAG), a synthetic DAG analogue, activates a dose-dependent, transient, inward current (IOAG ) that is enhanced by IP3 , mimicked by PLC activation and dependent on basal protein kinase C (PKC) activity. OAG depolarizes bag cell neurons and triggers action potential firing in culture, and prolongs electrically stimulated afterdischarges in intact bag cell neuron clusters ex vivo. Although PKC alone cannot activate the current, it is required for IOAG ; this is the first description of required obligate PKC activity working in concert with PLC, DAG and IP3 to maintain the depolarization required for prolonged excitability in Aplysia reproduction. ABSTRACT Following synaptic input, the bag cell neurons of Aplysia undergo a long-term afterdischarge of action potentials to secrete egg-laying hormone and initiate reproduction. Early in the afterdischarge, phospholipase C (PLC) hydrolyses phosphatidylinositol-4,5-bisphosphate into inositol trisphosphate (IP3 ) and diacylglycerol (DAG). In Aplysia, little is known about the action of DAG, or any interaction with IP3 ; thus, we examined the effects of a synthetic DAG analogue, 1-oleoyl-2-acetyl-sn-glycerol (OAG), on whole-cell voltage-clamped cultured bag cell neurons. OAG induced a large, prolonged, Ca(2+) -permeable, concentration-dependent inward current (IOAG ) that reversed at ∼-20 mV and was enhanced by intracellular IP3 . A similar current was evoked by either another DAG analogue, 1,2-dioctanoyl-sn-glycerol (DOG), or activating PLC with N-(3-trifluoromethylphenyl)-2,4,6-trimethylbenzenesulfonamide (m-3M3FBS). IOAG was reduced by the general cation channel blockers Gd(3+) or flufenamic acid. Work in other systems indicated that OAG activates channels independently of protein kinase C (PKC); however, we found pretreating bag cell neurons with any of the PKC inhibitors bisindolylmaleimide, sphinganine, or H7, attenuated IOAG . However, stimulating PKC with phorbol 12-myristate 13-acetate (PMA) did not evoke current or enhance IOAG ; moreover, unlike PMA, OAG failed to trigger PKC, as confirmed by an independent bioassay. Finally, OAG or m-3M3FBS depolarized cultured neurons, and while OAG did not provoke afterdischarges from bag cell neurons in the nervous system, it did double the duration of synaptically elicited afterdischarges. To our knowledge, this is the first report of obligate PKC activity for IOAG gating. An interaction between phosphoinositol metabolites and PKC could control the cation channel to influence afterdischarge duration.
Collapse
Affiliation(s)
- Raymond M Sturgeon
- Department of Biomedical and Molecular Sciences, Physiology Graduate Program, Queen's University, Kingston, ON, Canada, K7L 3N6
| | - Neil S Magoski
- Department of Biomedical and Molecular Sciences, Physiology Graduate Program, Queen's University, Kingston, ON, Canada, K7L 3N6.
| |
Collapse
|
15
|
Goodman CA, Hornberger TA, Robling AG. Bone and skeletal muscle: Key players in mechanotransduction and potential overlapping mechanisms. Bone 2015; 80:24-36. [PMID: 26453495 PMCID: PMC4600534 DOI: 10.1016/j.bone.2015.04.014] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 03/18/2015] [Accepted: 04/07/2015] [Indexed: 12/16/2022]
Abstract
The development and maintenance of skeletal muscle and bone mass is critical for movement, health and issues associated with the quality of life. Skeletal muscle and bone mass are regulated by a variety of factors that include changes in mechanical loading. Moreover, bone mass is, in large part, regulated by muscle-derived mechanical forces and thus by changes in muscle mass/strength. A thorough understanding of the cellular mechanism(s) responsible for mechanotransduction in bone and skeletal muscle is essential for the development of effective exercise and pharmaceutical strategies aimed at increasing, and/or preventing the loss of, mass in these tissues. Thus, in this review we will attempt to summarize the current evidence for the major molecular mechanisms involved in mechanotransduction in skeletal muscle and bone. By examining the differences and similarities in mechanotransduction between these two tissues, it is hoped that this review will stimulate new insights and ideas for future research and promote collaboration between bone and muscle biologists.(1).
Collapse
Affiliation(s)
- Craig A Goodman
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA; Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Australia; Institute of Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, VIC, Australia.
| | - Troy A Hornberger
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Alexander G Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
16
|
Kurth F, Franco-Obregón A, Casarosa M, Küster SK, Wuertz-Kozak K, Dittrich PS. Transient receptor potential vanilloid 2-mediated shear-stress responses in C2C12 myoblasts are regulated by serum and extracellular matrix. FASEB J 2015. [PMID: 26207028 DOI: 10.1096/fj.15-275396] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The developmental sensitivity of skeletal muscle to mechanical forces is unparalleled in other tissues. Calcium entry via reputedly mechanosensitive transient receptor potential (TRP) channel classes has been shown to play an essential role in both the early proliferative stage and subsequent differentiation of skeletal muscle myoblasts, particularly TRP canonical (TRPC) 1 and TRP vanilloid (TRPV) 2. Here we show that C2C12 murine myoblasts respond to fluid flow-induced shear stress with increments in cytosolic calcium that are largely initiated by the mechanosensitive opening of TRPV2 channels. Response to fluid flow was augmented by growth in low extracellular serum concentration (5 vs. 20% fetal bovine serum) by greater than 9-fold and at 18 h in culture, coincident with the greatest TRPV2 channel expression under identical conditions (P < 0.02). Fluid flow responses were also enhanced by substrate functionalization with laminin, rather than with fibronectin, agreeing with previous findings that the gating of TRPV2 is facilitated by laminin. Fluid flow-induced calcium increments were blocked by ruthenium red (27%) and SKF-96365 (38%), whereas they were unaltered by 2-aminoethoxydiphenyl borate, further corroborating that TRPV2 channels play a predominant role in fluid flow mechanosensitivity over that of TRPC1 and TRP melastatin (TRPM) 7.
Collapse
Affiliation(s)
- Felix Kurth
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Alfredo Franco-Obregón
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Marco Casarosa
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Simon K Küster
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Karin Wuertz-Kozak
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Petra S Dittrich
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| |
Collapse
|
17
|
Morrissey JB, Cheng RY, Davoudi S, Gilbert PM. Biomechanical Origins of Muscle Stem Cell Signal Transduction. J Mol Biol 2015; 428:1441-54. [PMID: 26004541 DOI: 10.1016/j.jmb.2015.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/03/2015] [Accepted: 05/06/2015] [Indexed: 10/23/2022]
Abstract
Skeletal muscle, the most abundant and widespread tissue in the human body, contracts upon receiving electrochemical signals from the nervous system to support essential functions such as thermoregulation, limb movement, blinking, swallowing and breathing. Reconstruction of adult muscle tissue relies on a pool of mononucleate, resident muscle stem cells, known as "satellite cells", expressing the paired-box transcription factor Pax7 necessary for their specification during embryonic development and long-term maintenance during adult life. Satellite cells are located around the myofibres in a niche at the interface of the basal lamina and the host fibre plasma membrane (i.e., sarcolemma), at a very low frequency. Upon damage to the myofibres, quiescent satellite cells are activated and give rise to a population of transient amplifying myogenic progenitor cells, which eventually exit the cell cycle permanently and fuse to form new myofibres and regenerate the tissue. A subpopulation of satellite cells self-renew and repopulate the niche, poised to respond to future demands. Harnessing the potential of satellite cells relies on a complete understanding of the molecular mechanisms guiding their regulation in vivo. Over the past several decades, studies revealed many signal transduction pathways responsible for satellite cell fate decisions, but the niche cues driving the activation and silencing of these pathways are less clear. Here we explore the scintillating possibility that considering the dynamic changes in the biophysical properties of the skeletal muscle, namely stiffness, and the stretch and shear forces to which a myofibre can be subjected to may provide missing information necessary to gain a full understanding of satellite cell niche regulation.
Collapse
Affiliation(s)
- James B Morrissey
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada M5S3G9; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada M5S3E1
| | - Richard Y Cheng
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada M5S3G9; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada M5S3E1
| | - Sadegh Davoudi
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada M5S3G9; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada M5S3E1
| | - Penney M Gilbert
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada M5S3G9; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada M5S3E1.
| |
Collapse
|
18
|
Tan N, Lansman JB. Utrophin regulates modal gating of mechanosensitive ion channels in dystrophic skeletal muscle. J Physiol 2014; 592:3303-23. [PMID: 24879867 DOI: 10.1113/jphysiol.2014.274332] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Dystrophin is a large, submembrane cytoskeletal protein, absence of which causes Duchenne muscular dystrophy. Utrophin is a dystrophin homologue found in both muscle and brain whose physiological function is unknown. Recordings of single-channel activity were made from membrane patches on skeletal muscle from mdx, mdx/utrn(+/-) heterozygotes and mdx/utrn(-/-) double knockout mice to investigate the role of these cytoskeletal proteins in mechanosensitive (MS) channel gating. We find complex, gene dose-dependent effects of utrophin depletion in dystrophin-deficient mdx muscle: (1) increased MS channel open probability, (2) a shift of MS channel gating to larger pressures, (3) appearance of modal gating of MS channels and small conductance channels and (4) expression of large conductance MS channels. We suggest a physical model in which utrophin acts as a scaffolding protein that stabilizes lipid microdomains and clusters MS channel subunits. Depletion of utrophin disrupts domain composition in a manner that favours open channel area expansion, as well as allowing diffusion and aggregation of additional MS channel subunits.
Collapse
Affiliation(s)
- Nhi Tan
- Department of Cellular and Molecular Pharmacology, School of Medicine, University of California, San Francisco, San Francisco, CA, 94143-0450, USA
| | - Jeffry B Lansman
- Department of Cellular and Molecular Pharmacology, School of Medicine, University of California, San Francisco, San Francisco, CA, 94143-0450, USA
| |
Collapse
|
19
|
Crocetti S, Beyer C, Unternährer S, Benavides Damm T, Schade-Kampmann G, Hebeisen M, Di Berardino M, Fröhlich J, Franco-Obregón A. Impedance flow cytometry gauges proliferative capacity by detecting TRPC1 expression. Cytometry A 2014; 85:525-36. [DOI: 10.1002/cyto.a.22461] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 12/10/2013] [Accepted: 03/03/2014] [Indexed: 01/13/2023]
Affiliation(s)
| | - Christian Beyer
- Electromagnetics in Medicine and Biology Group, Laboratory for Electromagnetic Fields and Microwave Electronics; ETH Zürich Switzerland
| | | | - Tatiana Benavides Damm
- Institute for Biomechanics; ETH Zürich Switzerland
- CC Aerospace Biomedical Science & Technology, Space Biology Group, Luzern University of Applied Sciences and Arts; Hergiswil Switzerland
| | | | - Monika Hebeisen
- Leister Process Technologies; Axetris Division; Kaegiswil Switzerland
| | | | - Jürg Fröhlich
- Electromagnetics in Medicine and Biology Group, Laboratory for Electromagnetic Fields and Microwave Electronics; ETH Zürich Switzerland
| | - Alfredo Franco-Obregón
- Institute for Biomechanics; ETH Zürich Switzerland
- Department of Surgery; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
| |
Collapse
|
20
|
Harisseh R, Chatelier A, Magaud C, Déliot N, Constantin B. Involvement of TRPV2 and SOCE in calcium influx disorder in DMD primary human myotubes with a specific contribution of α1-syntrophin and PLC/PKC in SOCE regulation. Am J Physiol Cell Physiol 2013; 304:C881-94. [DOI: 10.1152/ajpcell.00182.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Calcium homeostasis is critical for several vital functions in excitable and nonexcitable cells and has been shown to be impaired in many pathologies including Duchenne muscular dystrophy (DMD). Various studies using murine models showed the implication of calcium entry in the dystrophic phenotype. However, alteration of store-operated calcium entry (SOCE) and transient receptor potential vanilloid 2 (TRPV2)-dependant cation entry has not been investigated yet in human skeletal muscle cells. We pharmacologically characterized basal and store-operated cation entries in primary cultures of myotubes prepared from muscle of normal and DMD patients and found, for the first time, an increased SOCE in DMD myotubes. Moreover, this increase cannot be explained by an over expression of the well-known SOCE actors: TRPC1/4, Orai1, and stromal interaction molecule 1 (STIM1) mRNA and proteins. Thus we investigated the modes of regulation of this cation entry. We firstly demonstrated the important role of the scaffolding protein α1-syntrophin, which regulates SOCE in primary human myotubes through its PDZ domain. We also studied the implication of phospholipase C (PLC) and protein kinase C (PKC) in SOCE and showed that their inhibition restores normal levels of SOCE in DMD human myotubes. In addition, the involvement of TRPV2 in calcium deregulation in DMD human myotubes was explored. We showed an abnormal elevation of TRPV2-dependant cation entry in dystrophic primary human myotubes compared with normal ones. These findings show that calcium homeostasis mishandling in DMD myotubes depends on SOCE under the influence of Ca2+/PLC/PKC pathway and α1-syntrophin regulation as well as on TRPV2-dependant cation influx.
Collapse
Affiliation(s)
- Rania Harisseh
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| | - Aurélien Chatelier
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| | - Christophe Magaud
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| | - Nadine Déliot
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| | - Bruno Constantin
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| |
Collapse
|
21
|
Benavides Damm T, Richard S, Tanner S, Wyss F, Egli M, Franco-Obregón A. Calcium-dependent deceleration of the cell cycle in muscle cells by simulated microgravity. FASEB J 2013; 27:2045-54. [PMID: 23363573 DOI: 10.1096/fj.12-218693] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Of all our mechanosensitive tissues, skeletal muscle is the most developmentally responsive to physical activity. Conversely, restricted mobility due to injury or disease results in muscle atrophy. Gravitational force is another form of mechanical input with profound developmental consequences. The mechanical unloading resulting from the reduced gravitational force experienced during spaceflight results in oxidative muscle loss. We examined the early stages of myogenesis under conditions of simulated microgravity (SM). C2C12 mouse myoblasts in SM proliferated more slowly (2.23× less) as a result of their being retained longer within the G2/M phase of the cell cycle (2.10× more) relative to control myoblasts at terrestrial gravity. Blocking calcium entry via TRP channels with SKF-96365 (10-20 μM) accumulated myoblasts within the G2/M phase of the cell cycle and retarded their proliferation. On the genetic level, SM resulted in the reduced expression of TRPC1 and IGF-1 isoforms, transcriptional events regulated by calcium downstream of mechanical input. A decrease in TRPC1-mediated calcium entry thus appears to be a pivotal event in the muscle atrophy brought on by gravitational mechanical unloading. Hence, relieving the constant force of gravity on cells might prove one valid experimental approach to expose the underlying mechanisms modulating mechanically regulated developmental programs.
Collapse
|
22
|
Vasquez I, Tan N, Boonyasampant M, Koppitch KA, Lansman JB. Partial opening and subconductance gating of mechanosensitive ion channels in dystrophic skeletal muscle. J Physiol 2012; 590:6167-85. [PMID: 22966155 PMCID: PMC3530124 DOI: 10.1113/jphysiol.2012.240044] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 09/03/2012] [Indexed: 12/31/2022] Open
Abstract
We recorded the activity of single mechanosensitive (MS) ion channels in skeletal muscle from the mdx mouse, a deletion mutant that lacks the cytoskeletal protein, dystrophin. Experiments were designed to examine the influence of dystrophin, a major component of skeletal muscle costameres, on the behaviour of single MS channels. In the majority of recordings from cell-attached patches, MS channels have a conductance of ∼23 pS. Recordings from some patches, however, showed a smaller conductance channel of ∼7-14 pS. Large and small conductance channels were detected in a single patch and showed serial, non-random gating, suggesting different opening levels of a single channel. Analysis of the distribution of current amplitudes within the open channel showed MS channels fluctuate between subconductance levels. MS channels in dystrophic muscle spend ∼60% of the time at smaller subconductance levels, often failing to reach the fully open level. Applying pressure to the membrane of mdx fibres increases in a graded manner occupancy of the fully open state, while reducing occupancy of subconductance levels. Recordings also show partial openings of MS channels in both wild-type and mdx muscle that fail to reach the fully open state. Partial openings occur at a higher frequency in mdx muscle and reflect occupancy of subconductance levels seen during complete activations. In muscle from mdx/utrn(-/-) double knockout mice, MS channels also spend more time at subconductance levels than the fully open state. Conductance variability of MS channels may represent gating of a heteromeric protein composed of different channel subunits. The results also show that partial opening and prolonged burst duration are distinct mechanisms that contribute to excess Ca(2+) entry in dystrophic muscle.
Collapse
Affiliation(s)
- Ivan Vasquez
- Department of Cellular & Molecular Pharmacology, School of Medicine, University of California, San Francisco, CA 94143-0450, USA
| | | | | | | | | |
Collapse
|
23
|
Ho TC, Horn NA, Huynh T, Kelava L, Lansman JB. Evidence TRPV4 contributes to mechanosensitive ion channels in mouse skeletal muscle fibers. Channels (Austin) 2012; 6:246-54. [PMID: 22785252 DOI: 10.4161/chan.20719] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We recorded the activity of single mechanosensitive (MS) ion channels from membrane patches on single muscle fibers isolated from mice. We investigated the actions of various TRP (transient receptor potential) channel blockers on MS channel activity. 2-aminoethoxydiphenyl borate (2-APB) neither inhibited nor facilitated single channel activity at submillimolar concentrations. The absence of an effect of 2-APB indicates MS channels are not composed purely of TRPC or TRPV1, 2 or 3 proteins. Exposing patches to 1-oleolyl-2-acetyl-sn-glycerol (OAG), a potent activator of TRPC channels, also had no effect on MS channel activity. In addition, flufenamic acid and spermidine had no effect on the activity of single MS channels. By contrast, SKF-96365 and ruthenium red blocked single-channel currents at micromolar concentrations. SKF-96365 produced a rapid block of the open channel current. The blocking rate depended linearly on blocker concentration, while the unblocking rate was independent of concentration, consistent with a simple model of open channel block. A fit to the concentration-dependence of block gave k(on) = 13 x 10 ( 6) M (-1) s (-1) and k(off) = 1609 sec (-1) with K(D) = ~124 µM. Block by ruthenium red was complex, involving both reduction of the amplitude of the single-channel current and increased occupancy of subconductance levels. The reduction in current amplitude with increasing concentration of ruthenium red gave a K(D) = ~49 µM. The high sensitivity of MS channels to block by ruthenium red suggests MS channels in skeletal muscle contain TRPV subunits. Recordings from skeletal muscle isolated from TRPV4 knockout mice failed to show MS channel activity, consistent with a contribution of TRPV4. In addition, exposure to hypo-osmotic solutions increases opening of MS channels in muscle. Our results provide evidence TRPV4 contributes to MS channels in skeletal muscle.
Collapse
Affiliation(s)
- Tiffany C Ho
- Department of Cellular and Molecular Pharmacology, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
24
|
Matsumura CY, Taniguti APT, Pertille A, Neto HS, Marques MJ. Stretch-activated calcium channel protein TRPC1 is correlated with the different degrees of the dystrophic phenotype in mdx mice. Am J Physiol Cell Physiol 2011; 301:C1344-50. [DOI: 10.1152/ajpcell.00056.2011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In Duchenne muscular dystrophy (DMD) and in the mdx mouse model of DMD, the lack of dystrophin is related to enhanced calcium influx and muscle degeneration. Stretch-activated channels (SACs) might be directly involved in the pathology of DMD, and transient receptor potential cation channels have been proposed as likely candidates of SACs. We investigated the levels of transient receptor potential canonical channel 1 (TRPC1) and the effects of streptomycin, a SAC blocker, in muscles showing different degrees of the dystrophic phenotype. Mdx mice (18 days old, n = 16) received daily intraperitoneal injections of streptomycin (182 mg/kg body wt) for 18 days, followed by removal of the diaphragm, sternomastoid (STN), biceps brachii, and tibialis anterior muscles. Control mdx mice ( n = 37) were injected with saline. Western blot analysis showed higher levels of TRPC1 in diaphragm muscle compared with STN and limb muscles. Streptomycin reduced creatine kinase and prevented exercise-induced increases of total calcium and Evans blue dye uptake in diaphragm and in STN muscles. It is suggested that different levels of the stretch-activated calcium channel protein TRPC1 may contribute to the different degrees of the dystrophic phenotype seen in mdx mice. Early treatment designed to regulate the activity of these channels may ameliorate the progression of dystrophy in the most affected muscle, the diaphragm.
Collapse
Affiliation(s)
- Cíntia Yuri Matsumura
- Departamento de Anatomia, Biologia Celular, Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Ana Paula Tiemi Taniguti
- Departamento de Anatomia, Biologia Celular, Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Adriana Pertille
- Departamento de Anatomia, Biologia Celular, Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Humberto Santo Neto
- Departamento de Anatomia, Biologia Celular, Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Maria Julia Marques
- Departamento de Anatomia, Biologia Celular, Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
25
|
Saitoh M, Kuroda R, Muranaka Y, Uto N, Murai J, Kuroda H. Asymmetric inhibition of spicule formation in sea urchin embryos with low concentrations of gadolinium ion. Dev Growth Differ 2010; 52:735-46. [DOI: 10.1111/j.1440-169x.2010.01210.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Alexandre J, Lassalles JP. Hydrostatic and osmotic pressure activated channel in plant vacuole. Biophys J 2010; 60:1326-36. [PMID: 19431814 DOI: 10.1016/s0006-3495(91)82170-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The vacuolar membrane of red beet vacuoles contains a channel which was not gated by voltage or Ca(2+) ions. Its unit conductance was 20 pS in 200 mM symmetrical KCl solutions. It was stretch activated: the conductance remained constant but the probability of opening was increased by suction or pressure applied to a membrane patch. A 1.5-kNm(-2) suction applied to isolated patches or a 0.08-kNm(-2) pressure applied to a 45-mum diameter vacuole induced an e-fold change in the mean current. A 75% inhibition of the channel current was obtained with 10 muM Gd(3+) on the cytoplasmic side. The channel was more permeable for K(+) than for Cl(-) (P(K)/P(Cl) approximately 3). A possible clustering for this channel was suggested by the recordings of the patch current. The channel properties were not significantly affected by a change in sorbitol osmolality in the solutions under isoosmotic conditions, between 0.6 and 1 mol/kg sorbitol. However, the channel was very sensitive to an osmotic gradient. A 0.2-mol/kg sorbitol gradient induced a two-fold increase in unit conductance and a thirty-fold increase in the mean patch current of the channel. A current was measured, when the osmotic gradient was the only driving force applied to the vacuolar membrane. The hydrostatic and osmotic pressure (HOP) activated channel described in this paper could be gated in vivo condition by a change in osmolality, without the need of a change in the turgor pressure in the cell. The HOP channel represents a possible example of an osmoreceptor for plant cells.
Collapse
Affiliation(s)
- J Alexandre
- Laboratoire "Echanges Cellulaires," URA CNRS 203, Faculté des Sciences de Rouen, BP 118, 76134 Mont-Saint-Aignan Cedex, France
| | | |
Collapse
|
27
|
Martin V, Dousset E, Laurin J, Gondin J, Gautier M, Decherchi P. Group III and IV muscle afferent discharge patterns after repeated lengthening and shortening actions. Muscle Nerve 2009; 40:827-37. [PMID: 19626674 DOI: 10.1002/mus.21368] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The purpose of this study was to test the hypothesis that group III and IV muscle afferent activity would differ after concentric- and eccentric-type fatiguing tasks. Tibialis anterior afferent activities from adult rats were measured in three conditions: before and after a rest period (C), and after concentric (CC) or eccentric (EC) exercise. Specific activators were used to elicit increases in afferent discharge rates, i.e., electrically induced fatigue (EIF), or potassium chloride (KCl) and lactic acid (LA) injections. After the rest period (POST-condition), the control group displayed a pattern of response to stimuli similar to that obtained in baseline condition (PRE-condition). However, responses were significantly different in the exercise groups: afferent responses were blunted in the CC group and were almost suppressed in the EC group. These results demonstrate that the type of muscular contraction involved in the fatiguing task can affect group III and IV afferent fiber activity differently and, potentially, can differentially affect the regulation of central motor command.
Collapse
Affiliation(s)
- Vincent Martin
- INSERM U 902/EA 3872, Unité de Biologie Intégrative des Adaptations à l'Exercice, Université d'Evry-Val-d'Essonne/Genopole, Evry, France
| | | | | | | | | | | |
Collapse
|
28
|
Sabourin J, Lamiche C, Vandebrouck A, Magaud C, Rivet J, Cognard C, Bourmeyster N, Constantin B. Regulation of TRPC1 and TRPC4 cation channels requires an alpha1-syntrophin-dependent complex in skeletal mouse myotubes. J Biol Chem 2009; 284:36248-36261. [PMID: 19812031 DOI: 10.1074/jbc.m109.012872] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The dystrophin-associated protein complex (DAPC) is essential for skeletal muscle, and the lack of dystrophin in Duchenne muscular dystrophy results in a reduction of DAPC components such as syntrophins and in fiber necrosis. By anchoring various molecules, the syntrophins may confer a role in cell signaling to the DAPC. Calcium disorders and abnormally elevated cation influx in dystrophic muscle cells have suggested that the DAPC regulates some sarcolemmal cationic channels. We demonstrated previously that mini-dystrophin and alpha1-syntrophin restore normal cation entry in dystrophin-deficient myotubes and that sarcolemmal TRPC1 channels associate with dystrophin and the bound PDZ domain of alpha1-syntrophin. This study shows that small interfering RNA (siRNA) silencing of alpha1-syntrophin dysregulated cation influx in myotubes. Moreover, deletion of the PDZ-containing domain prevented restoration of normal cation entry by alpha1-syntrophin transfection in dystrophin-deficient myotubes. TRPC1 and TRPC4 channels are expressed at the sarcolemma of muscle cells; forced expression or siRNA silencing showed that cation influx regulated by alpha1-syntrophin is supported by TRPC1 and TRPC4. A molecular association was found between TRPC1 and TRPC4 channels and the alpha1-syntrophin-dystrophin complex. TRPC1 and TRPC4 channels may form sarcolemmal channels anchored to the DAPC, and alpha1-syntrophin is necessary to maintain the normal regulation of TRPC-supported cation entry in skeletal muscle. Cation channels with DAPC form a signaling complex that modulates cation entry and may be crucial for normal calcium homeostasis in skeletal muscles.
Collapse
Affiliation(s)
- Jessica Sabourin
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Coralie Lamiche
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Aurelie Vandebrouck
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Christophe Magaud
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Jerome Rivet
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Christian Cognard
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Nicolas Bourmeyster
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France
| | - Bruno Constantin
- Institut de Physiologie et Biologie Cellulaires, UMR CNRS 6187, Université de Poitiers, 86022 Poitiers, France.
| |
Collapse
|
29
|
Spangenburg EE. Changes in muscle mass with mechanical load: possible cellular mechanismsThis paper is one of a selection of papers published in this Special Issue, entitled 14th International Biochemistry of Exercise Conference – Muscles as Molecular and Metabolic Machines, and has undergone the Journal’s usual peer review process. Appl Physiol Nutr Metab 2009; 34:328-35. [DOI: 10.1139/h09-010] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Understanding the mechanisms that regulate skeletal muscle mass has remained a focus of numerous researchers for many years. Recent investigations have begun to elucidate cellular signaling mechanisms that regulate skeletal muscle hypertrophy, with significant effort being focused on the Akt/mammalian target of rapamycin (mTOR) signaling pathway. The Akt/mTOR pathway plays a major role in regulating the initiation of protein synthesis after the onset of mechanical loading of skeletal muscle. Although a number of downstream substrates for Akt/mTOR have been elucidated, very little is known about the upstream mechanisms that mechanical load employs to activate the Akt/mTOR signaling pathway. Thus, the purpose of this review is to discuss potential mechanisms that may contribute to the activation of the Akt/mTOR signaling mechanism in mechanically loaded skeletal muscle.
Collapse
Affiliation(s)
- Espen E. Spangenburg
- University of Maryland, School of Public Health, Department of Kinesiology, College Park, MD 20742, USA (e-mail: )
| |
Collapse
|
30
|
Germinario E, Esposito A, Midrio M, Peron S, Palade PT, Betto R, Danieli-Betto D. High-frequency fatigue of skeletal muscle: role of extracellular Ca(2+). Eur J Appl Physiol 2008; 104:445-53. [PMID: 18560877 DOI: 10.1007/s00421-008-0796-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2008] [Indexed: 10/21/2022]
Abstract
The present study evaluated whether Ca(2+) entry operates during fatigue of skeletal muscle. The involvement of different skeletal muscle membrane calcium channels and of the Na(+)/Ca(2+) exchanger (NCX) has been examined. The decline of force was analysed in vitro in mouse soleus and EDL muscles submitted to 60 and 110 Hz continuous stimulation, respectively. Stimulation with this high-frequency fatigue (HFF) protocol, in Ca(2+)-free conditions, caused in soleus muscle a dramatic increase of fatigue, while in the presence of high Ca(2+) fatigue was reduced. In EDL muscle, HFF was not affected by external Ca(2+) levels either way, suggesting that external Ca(2+) plays a general protective role only in soleus. Calciseptine, a specific antagonist of the cardiac isoform (alpha1C) of the dihydropyridine receptor, gadolinium, a blocker of both stretch-activated and store-operated Ca(2+) channels, as well as inhibitors of P2X receptors did not affect the development of HFF. Conversely, the Ca(2+) ionophore A23187 increased the protective action of extracellular Ca(2+). KB-R7943, a selective inhibitor of the reverse mode of NCX, produced an effect similar to that of Ca(2+)-free solution. These results indicate that a transmembrane Ca(2+) influx, mainly through NCX, may play a protective role during HFF development in soleus muscle.
Collapse
Affiliation(s)
- Elena Germinario
- Department of Human Anatomy and Physiology, University of Padova, Via Marzolo 3, 35131 Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
31
|
Gardam KE, Geiger JE, Hickey CM, Hung AY, Magoski NS. Flufenamic acid affects multiple currents and causes intracellular Ca2+ release in Aplysia bag cell neurons. J Neurophysiol 2008; 100:38-49. [PMID: 18436631 DOI: 10.1152/jn.90265.2008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Flufenamic acid (FFA) is a nonsteroidal antiinflammatory agent, commonly used to block nonselective cation channels. We previously reported that FFA potentiated, rather than inhibited, a cation current in Aplysia bag cell neurons. Prompted by this paradoxical result, the present study examined the effects of FFA on membrane currents and intracellular Ca2+ in cultured bag cell neurons. Under whole cell voltage clamp, FFA evoked either outward (I out) or inward (I in) currents. I out had a rapid onset, was inhibited by the K+ channel blocker, tetraethylammonium, and was associated with both an increase in membrane conductance and a negative shift in the whole cell current reversal potential. I in developed more slowly, was inhibited by the cation channel blocker, Gd3+, and was concomitant with both an increased conductance and positive shift in reversal potential. FFA also enhanced the use-dependent inactivation and caused a positive-shift in the activation curve of the voltage-dependent Ca2+ current. Furthermore, as measured by ratiometric imaging, FFA produced a rise in intracellular Ca2+ that persisted in the absence of extracellular Ca2+ and was reduced by depleting either the endoplasmic reticulum and/or mitochondrial stores. Ca2+ appeared to be involved in the activation of I in, as strong intracellular Ca2+ buffering effectively eliminated I in but did not alter I out. Finally, the effects of FFA were likely not due to block of cyclooxygenase given that the general cyclooxygenase inhibitor, indomethacin, failed to evoke either current. That FFA influences a number of neuronal properties needs to be taken into consideration when employing it as a cation channel antagonist.
Collapse
Affiliation(s)
- Kate E Gardam
- Department of Physiology and Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|
32
|
Sonobe T, Inagaki T, Poole DC, Kano Y. Intracellular calcium accumulation following eccentric contractions in rat skeletal muscle in vivo: role of stretch-activated channels. Am J Physiol Regul Integr Comp Physiol 2008; 294:R1329-37. [DOI: 10.1152/ajpregu.00815.2007] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although the accumulation of intracellular calcium ions ([Ca2+]i) is associated with muscle damage, little is known regarding the temporal profile of muscle [Ca2+]iunder in vivo conditions, and, specifically, the effects of different contraction types [e.g., isometric (ISO); eccentric (ECC)] on [Ca2+]iremain to be determined. The following hypotheses were tested. 1) For 90 min at rest, an in vivo vs. in vitro preparation would better maintain initial [Ca2+]i. 2) Compared with ISO, ECC contractions (50 contractions, 10 sets, 5-min interval) would lead to a greater increase of [Ca2+]i. 3) Elevated [Ca2+]iduring ECC would be reduced or prevented by the stretch-activated ion channel blockers streptomycin and gadolinium (Gd3+). Spinotrapezius muscles of Wistar rats were exteriorized (in vivo) or excised (in vitro). [Ca2+]iwas evaluated by loading the muscle with fura 2-AM using fluorescence imaging. [Ca2+]irose progressively beyond 40 min at rest under in vitro but not in vivo conditions during the 90-min protocol. In vivo [Ca2+]iincreased more rapidly during ECC (first set) than ISO (fifth set) ( P < 0.05 vs. precontraction values). The peak level of [Ca2+]iwas increased by 21.5% (ISO) and 42.8% (ECC) after 10 sets (both P < 0.01). Streptomycin and Gd3+abolished the majority of [Ca2+]iincrease during ECC (69 and 86% reduction, respectively; P < 0.01 from peak [Ca2+]iof ECC). In conclusion, in vivo quantitative analyses demonstrated that ECC contractions elevate [Ca2+]isignificantly more than ISO contractions and that stretch-activated channels may play a permissive role in this response.
Collapse
|
33
|
Taghli-Lamallem O, Akasaka T, Hogg G, Nudel U, Yaffe D, Chamberlain JS, Ocorr K, Bodmer R. Dystrophin deficiency in Drosophila reduces lifespan and causes a dilated cardiomyopathy phenotype. Aging Cell 2008; 7:237-49. [PMID: 18221418 DOI: 10.1111/j.1474-9726.2008.00367.x] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
A number of studies have been conducted recently on the model organism Drosophila to determine the function of genes involved in human disease, including those implicated in neurological disorders, cancer and metabolic and cardiovascular diseases. The simple structure and physiology of the Drosophila heart tube together with the available genetics provide a suitable in vivo assay system for studying cardiac gene functions. In our study, we focus on analysis of the role of dystrophin (Dys) in heart physiology. As in humans, the Drosophila dys gene encodes multiple isoforms, of which the large isoforms (DLPs) and a truncated form (Dp117) are expressed in the adult heart. Here, we show that the loss of dys function in the heart leads to an age-dependent disruption of the myofibrillar organization within the myocardium as well as to alterations in cardiac performance. dys RNAi-mediated knockdown in the mesoderm also shortens lifespan. Knockdown of all or deletion of the large isoforms increases the heart rate by shortening the diastolic intervals (relaxation phase) of the cardiac cycle. Morphologically, loss of the large DLPs isoforms causes a widening of the cardiac tube and a lower fractional shortening, a phenotype reminiscent of dilated cardiomyopathy. The dilated dys mutant phenotype was reversed by expressing a truncated mammalian form of dys (Dp116). Our results illustrate the utility of Drosophila as a model system to study dilated cardiomyopathy and other muscular-dystrophy-associated phenotypes.
Collapse
MESH Headings
- Age Factors
- Animals
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/mortality
- Cardiomyopathy, Dilated/physiopathology
- Disease Models, Animal
- Drosophila/embryology
- Drosophila/genetics
- Drosophila/metabolism
- Drosophila Proteins/analysis
- Drosophila Proteins/genetics
- Drosophila Proteins/metabolism
- Dystrophin/deficiency
- Dystrophin/genetics
- Heart Defects, Congenital/mortality
- Heart Defects, Congenital/pathology
- Heart Defects, Congenital/physiopathology
- Heart Rate
- Longevity/genetics
- Muscular Dystrophy, Animal/congenital
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/physiopathology
- Myocardial Contraction
- Myocytes, Cardiac/pathology
- Myofibrils/genetics
- Myofibrils/pathology
- Phenotype
- Protein Isoforms
- Sequence Deletion
Collapse
Affiliation(s)
- Ouarda Taghli-Lamallem
- Development and Aging Program, Burnham Institute for Medical Research, 10901 North Torrey Pines Rd, Building 7, Room 7125, La Jolla, Ca 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Roy RR, Zhong H, Khalili N, Kim SJ, Higuchi N, Monti RJ, Grossman E, Hodgson JA, Edgerton VR. Is spinal cord isolation a good model of muscle disuse? Muscle Nerve 2007; 35:312-21. [PMID: 17154282 DOI: 10.1002/mus.20706] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The patterns of normal daily activity that are required to maintain normal skeletal muscle properties remain unknown. The present study was designed to determine whether spinal cord isolation can be used as a reliable experimental model of neuromuscular inactivity, that is, as a baseline for the absence of activity. Electromyograms (EMGs) were recorded from selected hindlimb muscles of unanesthetized rats over 24-hour periods before and 7, 30, 60, and 90 days after surgical isolation of the lumbar spinal cord. Our data indicate that some rat slow muscle fibers pre-surgery were activated for less than 3 hours per day. Spinal cord isolation (SI) reduced the mean daily integrated EMG (IEMG) and daily EMG duration in the primary slow extensor muscle (soleus) to <1% of control, and in the primary fast extensor muscles [medial gastrocnemius (MG) and vastus lateralis (VL)] to <2% of control. These parameters were decreased to <8% and 3% of control, respectively, in a primary fast flexor muscle, the tibialis anterior (TA). From 30 to 90 days post-SI, the mean amplitudes of the spontaneous EMG bursts were relatively normal in the soleus, increased approximately 2-fold in the MG and VL, and increased approximately 4-fold in the TA. Some evidence of the normal antagonistic flexor-extensor relationship was apparent in the brief periods of recorded activity post-SI. These results indicate that SI eliminates nearly all of the normal EMG activity in the hindlimb muscles in the presence of relatively normal muscle innervation and functional intraspinal neural circuitry.
Collapse
Affiliation(s)
- R R Roy
- Brain Research Institute, University of California Los Angeles, 1320 Gonda Neuroscience and Genetics Building, Box 951761, Los Angeles, California 90095-1761, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Vydevska-Chichova M, Mileva K, Radicheva N. Differential changes in myoelectric characteristics of slow and fast fatigable frog muscle fibres during long-lasting activity. J Electromyogr Kinesiol 2007; 17:131-41. [DOI: 10.1016/j.jelekin.2006.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Revised: 12/16/2005] [Accepted: 01/05/2006] [Indexed: 10/24/2022] Open
|
36
|
Hung AY, Magoski NS. Activity-Dependent Initiation of a Prolonged Depolarization in Aplysia Bag Cell Neurons: Role for a Cation Channel. J Neurophysiol 2007; 97:2465-79. [PMID: 17353554 DOI: 10.1152/jn.00941.2006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The translation of prior activity into changes in excitability is essential for memory and the initiation of behavior. After brief synaptic input, the bag cell neurons of Aplysia californica undergo a nearly 30-min afterdischarge to release egg-laying hormone. The present study examines a prolonged depolarization in cultured bag cell neurons. A 5-Hz, 10-s action potential train elicited a depolarization of about 10 mV, which lasted ≤30 min and was reduced by calmodulin kinase inhibition. Very broad action potentials (resulting from TEA application) decreased prolonged depolarization amplitude, indicating that strong Ca2+ influx did not necessarily promote the response. The prolonged depolarization current ( IPD) was recorded after 5-Hz, 10-s trains of square voltage pulses of varying duration (10–150 ms). Despite Ca2+ influx increasing steadily with pulse duration, IPD was most reliably initiated at 100 ms, suggesting a Ca2+ window or limit exists for triggering IPD. Consistent with this, modestly broader action potentials, evoked by lengthening the train current-pulse duration, resulted in smaller prolonged depolarizations. With respect to the properties of IPD, it displayed a linear current–voltage relationship with a reversal potential of about −45 mV that was shifted to approximately −25 mV by lowering internal K+ or about −56 mV by lowering external Na+ and Ca2+. IPD was blocked by Gd3+, but was not antagonized by MDL-123302A, SKF-96365, 2-APB, tetrodotoxin, or flufenamic acid. Optimal Ca2+ influx may activate calmodulin kinase and a voltage-independent, nonselective cation channel to initiate the prolonged depolarization, thereby contributing to the afterdischarge and reproduction.
Collapse
Affiliation(s)
- Anne Y Hung
- Department of Physiology, Queen's University, 4th Floor, Botterell Hall, 18 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | | |
Collapse
|
37
|
|
38
|
Lansman JB, Franco-Obregón A. MECHANOSENSITIVE ION CHANNELS IN SKELETAL MUSCLE: A LINK IN THE MEMBRANE PATHOLOGY OF MUSCULAR DYSTROPHY. Clin Exp Pharmacol Physiol 2006; 33:649-56. [PMID: 16789935 DOI: 10.1111/j.1440-1681.2006.04393.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Mechanosensitive (MS) channels are expressed abundantly in skeletal muscle at all stages of development. In recordings from membrane patches, MS channels are constitutively active at the resting potential. The channels are selective for cations and have a large single-channel conductance (approximately 25 pS in physiological saline) and a high Ca2+ permeability (relative permeability of Ca2+ to K+ (PCa/PK) = 7). 2. Mechanosensitive channel activity recorded from the surface of myotubes from dystrophic mdx mice was substantially greater than the activity recorded from wild-type myotubes. Increased channel activity in the mutant results from the induction in a subpopulation of channels of a novel MS gating mode characterized by markedly prolonged channel openings and inactivation in response to membrane stretch. 3. Membrane stretch or a strong depolarization causes an irreversible switch to the stretch-inactivated gating mode in mdx myotubes. A stretch-induced shift in MS channel gating mode may contribute to stretch-induced elevations in [Ca2+]i during the early stages of disease pathogenesis. 4. Abnormalities of MS channel behaviour are also detected in recordings from patches on flexor digitorum brevis fibres acutely isolated from mdx mice. Mechanosensitive channel opening probability is higher in mdx fibres at all developmental stages. In addition, channel numbers are persistently elevated during postnatal development, failing to undergo a normal process of downregulation during the first 3 postnatal weeks. 5. Two distinct mechanisms may contribute to elevations of [Ca2+]i in dystrophin-deficient skeletal muscle: (i) a membrane stress-dependent switch of MS channels into to a prolonged opening mode; and (ii) a loss of developmental downregulation leading to persistent MS channel expression during postnatal muscle development.
Collapse
Affiliation(s)
- Jeffry B Lansman
- Department of Cellular and Molecular Pharmacology, UCSF School of Medicine, San Francisco, California 94143-0450, USA.
| | | |
Collapse
|
39
|
Wedhas N, Klamut HJ, Dogra C, Srivastava AK, Mohan S, Kumar A. Inhibition of mechanosensitive cation channels inhibits myogenic differentiation by suppressing the expression of myogenic regulatory factors and caspase-3 activity. FASEB J 2006; 19:1986-97. [PMID: 16319142 DOI: 10.1096/fj.05-4198com] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mechanosensitive cation channels (MSC) are ubiquitous in eukaryotic cell types. However, the physiological functions of MSC in several tissues remain in question. In this study we have investigated the role of MSC in skeletal myogenesis. Treatment of C2C12 myoblasts with gadolinium ions (MSC blocker) inhibited myotube formation and the myogenic index in differentiation medium (DM). The enzymatic activity of creatine kinase (CK) and the expression of myosin heavy chain-fast twitch (MyHCf) in C2C12 cultures were also blocked in response to gadolinium. Treatment of C2C12 myoblasts with gadolinium ions did not affect the expression of either cyclin A or cyclin D1 in DM. Other inhibitors of MSC such as streptomycin and GsTMx-4 also suppressed the expression of CK and MyHCf in C2C12 cultures. The inhibitory effect of gadolinium ions on myogenic differentiation was reversible and independent of myogenic cell type. Real-time-polymerase chain reaction analysis revealed that inhibition of MSC decreases the expression of myogenic transcription factors MyoD, myogenin, and Myf-5. Furthermore, the activity of skeletal alpha-actin promoter was suppressed on MSC blockade. Treatment of C2C12 myoblasts with gadolinium ions prevented differentiation-associated cell death and inhibited the cleavage of poly (ADP-ribose) polymerase and activation of caspase-3. On the other hand, delivery of active caspase-3 protein to C2C12 myoblasts reversed the inhibitory effect of gadolinium ions on myogenesis. Our data suggest that inhibition of MSC suppresses myogenic differentiation by inhibiting the caspase-3 activity and the expression of myogenic regulatory factors.
Collapse
Affiliation(s)
- Nia Wedhas
- Molecular Genetics Division, Musculoskeletal Disease Center, Jerry L. Pettis VA Medical Center, Loma Linda, California 92354, USA
| | | | | | | | | | | |
Collapse
|
40
|
Spangenburg EE, McBride TA. Inhibition of stretch-activated channels during eccentric muscle contraction attenuates p70S6K activation. J Appl Physiol (1985) 2005; 100:129-35. [PMID: 16179399 DOI: 10.1152/japplphysiol.00619.2005] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Eccentric contractions (EC) are known to result in muscle hypertrophy, potentially through activation of the Akt-mammalian target of rapamycin-p70 S6 kinase (p70S6K) signaling pathway. Previous work has also demonstrated that EC result in the opening of stretch-activated channels (SAC), and inhibition of these channels resulted in an attenuation of EC-induced muscle hypertrophy. The purpose of this study was to test the hypothesis that a known intracellular pathway directly associated with muscle hypertrophy is coupled to the opening of SAC. Specifically, we measured the activation of the Akt, GSK-3beta, p70S6K, and ribosomal protein S6 following a single bout of EC in the rat tibialis anterior (TA) muscle. The TA muscles performed four sets of six repetitions of EC. In vivo blockade of SAC was performed by a continuous oral treatment with streptomycin in the drinking water (4 g/l) or by intravenous infusion of 80 micromol/kg gadolinium (Gd3+). EC increased the degree of Akt and p70S6K phosphorylation in the TA muscle, whereas in animals in which SAC had been inhibited, there was a reduced capacity for EC to induce Akt or p70S6K phosphorylation. Accompanying this reduced activation of Akt and p70S6K was a failure to phosphorylate GSK-3beta or S6 when SAC were inhibited. The results from these data indicate the necessity of functional SAC for the complete activation of Akt and p70S6K pathway in response to EC.
Collapse
Affiliation(s)
- Espen E Spangenburg
- Exercise Biology Program, Division of Biological Sciences, School of Medicine, University of California-Davis, USA
| | | |
Collapse
|
41
|
Allen DG, Whitehead NP, Yeung EW. Mechanisms of stretch-induced muscle damage in normal and dystrophic muscle: role of ionic changes. J Physiol 2005; 567:723-35. [PMID: 16002444 PMCID: PMC1474216 DOI: 10.1113/jphysiol.2005.091694] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2005] [Accepted: 07/05/2005] [Indexed: 12/22/2022] Open
Abstract
Muscle damage, characterized by prolonged weakness and delayed onset of stiffness and soreness, is common following contractions in which the muscles are stretched. Stretch-induced damage of this sort is more pronounced in the muscular dystrophies and the profound muscle damage observed in these conditions may involve similar pathways. It has been known for many years that damaged muscles accumulate calcium and that elevating calcium in normal muscles simulates many aspects of muscle damage. The changes in intracellular calcium, sodium and pH following stretched contractions are reviewed and the various pathways which have been proposed to allow ion entry are discussed. One possibility is that TRPC1 (transient receptor potential, canonical), a protein which seems to form both a stretch-activated channel and a store-operated channel, is the main source of Ca(2+) entry. The mechanisms by which the changes in intracellular ions contribute to reduced force production, to increased protein breakdown and to increased membrane permeability are considered. A hypothetical scheme for muscle damage which incorporates these ideas is presented.
Collapse
Affiliation(s)
- D G Allen
- School of Medical Sciences, University of Sydney, NSW, Australia.
| | | | | |
Collapse
|
42
|
Macdonald WA, Pedersen TH, Clausen T, Nielsen OB. N-Benzyl-p-toluene sulphonamide allows the recording of trains of intracellular action potentials from nerve-stimulated intact fast-twitch skeletal muscle of the rat. Exp Physiol 2005; 90:815-25. [PMID: 16049056 DOI: 10.1113/expphysiol.2005.031435] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In skeletal muscle, the intracellular recording of trains of action potentials is difficult owing to the movement of the muscle upon stimulation. A potential tool for the removal of muscle movement is the cross-bridge cycle blocker, N-benzyl-p-toluene sulphonamide (BTS), although the effects of BTS on the passive and active membrane properties of intact muscle fibres are not known. Rat extensor digitorum longus (EDL) muscle was used to show that 50 mum BTS reduced tetanic force to approximately 10% of control force, without markedly altering muscle excitability. Incubation with BTS did not alter intracellular K+ content or Na+-K+ pump activity, but produced minor decreases in intracellular Na+ content (7%), resting 22Na+ influx (14%) and excitation-induced 22Na+ influx (29%). Despite these alterations to Na+ fluxes, BTS did not impair muscle excitability, since membrane conductance, resting membrane potential (RMP), rheobase current and the amplitude, overshoot and maximum rate of depolarization of the action potential were all unaltered. However, BTS did induce a small (8%) decrease in the maximum rate of repolarization of the action potential and an increase in the refractory period. The minor effects of BTS on muscle membrane properties did not compromise the ability of the muscle to propagate action potentials, even during tetanic stimulation. In conclusion, BTS can be used successfully to reduce contractility, allowing the intracellular recording of action potentials during both twitch and tetanic contraction of nerve-stimulated muscle, thus making it an excellent tool for the study of electrophysiology in fast-twitch skeletal muscle.
Collapse
Affiliation(s)
- W A Macdonald
- Institute of Physiology and Biophysics, University of Aarhus, Arhus, Denmark.
| | | | | | | |
Collapse
|
43
|
Yeung EW, Whitehead NP, Suchyna TM, Gottlieb PA, Sachs F, Allen DG. Effects of stretch-activated channel blockers on [Ca2+]i and muscle damage in the mdx mouse. J Physiol 2004; 562:367-80. [PMID: 15528244 PMCID: PMC1665499 DOI: 10.1113/jphysiol.2004.075275] [Citation(s) in RCA: 216] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The mdx mouse lacks dystrophin and is a model of human Duchenne muscular dystrophy. Single mdx muscle fibres were isolated and subjected to a series of stretched (eccentric) contractions while measuring intracellular calcium concentration ([Ca(2+)](i)) with fluo-3 and confocal microscopy. Following the stretched contractions there was a slow rise in resting [Ca(2+)](i) and after 30 min both the [Ca(2+)](i) during a tetanus (tetanic [Ca(2+)](i)) and the tetanic force were reduced. Two blockers of stretch-activated channels, streptomycin and the spider venom toxin GsMTx4, prevented the rise of resting [Ca(2+)](i) and partially prevented the decline of tetanic [Ca(2+)](i) and force. Reducing extracellular calcium to zero also prevented the rise in resting [Ca(2+)](i) and prevented some of the decline in tetanic [Ca(2+)](i) and force. Patch-clamping experiments identified a stretch-activated channel in both wild-type and mdx myotubes which was blocked by GsMTx4. These data suggest that blockers of stretch-activated channels can ameliorate the force reduction following stretched contractions by reducing the influx of Ca(2+) into the muscle. We therefore tested whether in intact mdx mice streptomycin, added to the drinking water, was capable of reducing muscle damage. mdx mice show a period of muscle damage from 20 to 40 days of life and fibres which regenerate from this damage display central nuclei. We measured the frequency of central nuclei in control mdx mice compared to streptomycin-treated mdx mice and showed that the incidence of central nuclei was significantly reduced by streptomycin treatment. This result suggests that blockers of stretch-activated channels may protect against muscle damage in the intact mdx mouse.
Collapse
MESH Headings
- Aniline Compounds
- Animals
- Calcium Signaling/drug effects
- Cell Nucleus/drug effects
- Cell Nucleus/pathology
- Electric Stimulation
- Electrophysiology
- Fluorescent Dyes
- In Vitro Techniques
- Intercellular Signaling Peptides and Proteins
- Membrane Potentials/physiology
- Mice
- Mice, Inbred mdx
- Microscopy, Confocal
- Muscle Contraction/physiology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/physiology
- Muscle Spindles/drug effects
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/prevention & control
- Patch-Clamp Techniques
- Peptides/pharmacology
- Protein Synthesis Inhibitors/pharmacology
- Spider Venoms/pharmacology
- Streptomycin/pharmacology
- Xanthenes
Collapse
Affiliation(s)
- Ella W Yeung
- School of Medical Sciences and Institute for Biomedical Research, University of Sydney F13, NSW 2006, Australia
| | | | | | | | | | | |
Collapse
|
44
|
Yeung EW, Allen DG. STRETCH-ACTIVATED CHANNELS IN STRETCH-INDUCED MUSCLE DAMAGE: ROLE IN MUSCULAR DYSTROPHY. Clin Exp Pharmacol Physiol 2004; 31:551-6. [PMID: 15298550 DOI: 10.1111/j.1440-1681.2004.04027.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Stretch-induced muscle injury results in the damage that causes reduced force and increased membrane permeability. This muscle damage is caused, in part, by ionic entry through stretch-activated channels and blocking these channels with Gd3+ or streptomycin reduces the force deficit associated with damage. 2. Dystrophin-deficient muscles are more susceptible to stretch-induced muscle injury and the recovery from injury can be incomplete. We have found that Na+ entry associated with stretch-induced injury is enhanced in dystrophin-deficient muscles and that blockers of stretch-activated channels are capable of preventing ionic entry and reducing muscle damage. 3. A model is presented that proposes links between stretch-induced injury, opening of stretch-activated channels, increased levels of intracellular ions and various forms of muscle damage. Although changes in Na+ accompany stretch-induced muscle injury, we believe that changes in Ca2+ probably have a more central role in the damage process.
Collapse
Affiliation(s)
- Ella W Yeung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | | |
Collapse
|
45
|
Abstract
1. Repeated activity of skeletal muscle causes a variety of changes in its properties: muscles become weaker with intense use (fatigue), may feel sore and weak after repeated contractions involving stretch and can degenerate in some disease conditions. The present review considers the role of early ionic changes in the development of each of these conditions. 2. Single fibre preparations of mouse muscle were used to measure ionic changes following activity induced changes in function. Single fibres were dissected with intact tendons and stimulated to produce force. Fluorescent indicators were microinjected into the fibres to allow simultaneous ionic measurements with determination of mechanical performance. 3. One theory to explain muscle fatigue is that fatigue is caused by the accumulation of lactic acid, producing an intracellular acidosis that inhibits the myofibrillar proteins. In contrast, we found that during repeated tetani there was little or no pH change, but that failure of calcium release was a major contributor to fatigue. Currently, it is proposed that precipitation of calcium and phosphate in the sarcoplasmic reticulum contributes to the failure of calcium release. 4. Muscles can be used to shorten and produce force or they can be used to de-accelerate loads (stretched or eccentric contractions). One day after intense exercise involving stretched contractions, muscles are weak, sore and tender, and this damage can take a week to recover. In this condition, sarcomeres are disorganized and there are increases in resting intracellular Ca2+ and Na+. Recently, we demonstrated that the elevation of Na+ occurs through a stretch-activated channel that can be blocked by either gadolinium or streptomycin. Preventing the increase in [Na+]i with gadolinium also prevented part of the muscle weakness after stretched contractions. 5. Duchenne muscular dystrophy is a lethal degenerative disease of muscles in which the protein dystrophin is absent. Dystrophic muscles are more susceptible to stretch-induced muscle damage and the stretch-activated channel seems to be one pathway for the increases in intracellular Ca2+ and Na+ that are a feature of this disease. We have shown recently that blockers of the stretch-activated channel can minimize some of the short-term damage in muscles from the mdx mouse, which also lacks dystrophin. Currently, we are testing whether blockers of the stretch-activated channels given systemically to mdx mice can protect against some features of the disease.
Collapse
Affiliation(s)
- D G Allen
- School of Biomedical Sciences and Institute for Biomedical Research, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
46
|
Yeung EW, Head SI, Allen DG. Gadolinium reduces short-term stretch-induced muscle damage in isolated mdx mouse muscle fibres. J Physiol 2004; 552:449-58. [PMID: 14561828 PMCID: PMC2343387 DOI: 10.1113/jphysiol.2003.047373] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Duchenne muscular dystrophy is a lethal muscle disease caused by absence of the protein dystrophin which is part of a glycoprotein complex located on the intracellular surface of the surface membrane. The precise function of dystrophin and the reason why its absence causes severe muscle damage are unclear. Stretch-induced muscle damage is well recognised in normal muscle and is more severe in muscles from animals lacking dystrophin (mdx mice). It has been proposed that stretch-induced damage underlies the progression of damage in muscular dystrophy. In the present study we confirm that single fibres from mdx muscle are more susceptible to stretch-induced damage and show that there is an associated rise in intracellular sodium concentration ([Na+]i) which is greater than in wild-type mice. We show that this rise in [Na+]i can be prevented by Gd3+, which is an established blocker of stretch-activated channels. mdx fibres have a higher than normal resting [Na+]i and this is also reduced by Gd3+. If Gd3+ is applied over the period in which [Na+]i rises following stretched contraction, it prevents one component of the reduced force. The other component of reduced force is caused by inhomogeneity of sarcomeres and can be minimised by stretching the muscle to its new optimum length. These experiments show that part of the short-term damage caused by stretch in mdx fibres can be prevented by blocking stretch-activated channels.
Collapse
Affiliation(s)
- Ella W Yeung
- Institute for Biomedical Research and Department of Physiology, University of Sydney F13, NSW 2006, Australia
| | | | | |
Collapse
|
47
|
Yeung EW, Ballard HJ, Bourreau JP, Allen DG. Intracellular sodium in mammalian muscle fibers after eccentric contractions. J Appl Physiol (1985) 2003; 94:2475-82. [PMID: 12588791 DOI: 10.1152/japplphysiol.01128.2002] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The effect of eccentric contractions on intracellular Na(+) concentration ([Na(+)](i)) and its distribution were examined in isolated rat and mouse muscle fiber bundles. [Na(+)](i) was measured with either Na(+)-binding benzofuran isophthalate or sodium green. Ten isometric contractions had no significant effect on force (measured after 5 min of recovery) and caused no significant change in the resting [Na(+)](i) (7.2 +/- 0.5 mM). In contrast 10 eccentric contractions (40% stretch at 4 muscle lengths/s) reduced developed force at 100 Hz to 45 +/- 3% of control and increased [Na(+)](i) to 16.3 +/- 1.6 mM (n = 6; P < 0.001). The rise of [Na(+)](i) occurred over 1-2 min and showed only minimal recovery after 30 min. Confocal images of the distribution of [Na(+)](i) showed a spatially uniform distribution both at rest and after eccentric contractions. Gd(3+) (20 microM) had no effect on resting [Na(+)](i) or control tetanic force but prevented the rise of [Na(+)](i) and reduced the force deficit after eccentric damage. These data suggest that Na(+) entry after eccentric contractions may occur principally through stretch-sensitive channels.
Collapse
Affiliation(s)
- Ella W Yeung
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | | | | | | |
Collapse
|
48
|
Vandebrouck C, Martin D, Colson-Van Schoor M, Debaix H, Gailly P. Involvement of TRPC in the abnormal calcium influx observed in dystrophic (mdx) mouse skeletal muscle fibers. J Cell Biol 2002; 158:1089-96. [PMID: 12235126 PMCID: PMC2173225 DOI: 10.1083/jcb.200203091] [Citation(s) in RCA: 265] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Duchenne muscular dystrophy results from the lack of dystrophin, a cytoskeletal protein associated with the inner surface membrane, in skeletal muscle. The absence of dystrophin induces an abnormal increase of sarcolemmal calcium influx through cationic channels in adult skeletal muscle fibers from dystrophic (mdx) mice. We observed that the activity of these channels was increased after depletion of the stores of calcium with thapsigargin or caffeine. By analogy with the situation observed in nonexcitable cells, we therefore hypothesized that these store-operated channels could belong to the transient receptor potential channel (TRPC) family. We measured the expression of TRPC isoforms in normal and mdx adult skeletal muscles fibers, and among the seven known isoforms, five were detected (TRPC1, 2, 3, 4, and 6) by RT-PCR. Western blot analysis and immunocytochemistry of normal and mdx muscle fibers demonstrated the localization of TRPC1, 4, and 6 proteins at the plasma membrane. Therefore, an antisense strategy was used to repress these TRPC isoforms. In parallel with the repression of the TRPCs, we observed that the occurrence of calcium leak channels was decreased to one tenth of its control value (patch-clamp technique), showing the involvement of TRPC in the abnormal calcium influx observed in dystrophic fibers.
Collapse
Affiliation(s)
- Clarisse Vandebrouck
- Département de Physiologie, Université Catholique de Louvain (UCL 5540), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | | | | | | | | |
Collapse
|
49
|
Vandebrouck C, Duport G, Raymond G, Cognard C. Hypotonic medium increases calcium permeant channels activity in human normal and dystrophic myotubes. Neurosci Lett 2002; 323:239-43. [PMID: 11959428 DOI: 10.1016/s0304-3940(02)00148-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Duchenne muscular dystrophy (DMD) is characterized by the absence of dystrophin and an elevated intracellular calcium level. Single-channel recordings were performed with the cell-attached configuration of the patch-clamp technique. The present study shows, on human co-cultured normal and dystrophic muscle cells, the evidence for an increased activity of calcium permeant cationic mechano-sensitive channels under hypotonic medium stimulation. This activity was particularly enhanced in DMD cells. The hypotonic medium induced drastic changes in the single-channel activity characteristics, which are: a large increase of the calcium over potassium permeability ratio; and a great enhancement of the quantity of current crossing through these channels. These channels could contribute to a significant calcium entry, which could participate in the abnormal calcium homeostasis observed in DMD muscle.
Collapse
MESH Headings
- Adolescent
- Adult
- Calcium/metabolism
- Calcium Channels/drug effects
- Calcium Channels/metabolism
- Cation Transport Proteins/drug effects
- Cation Transport Proteins/metabolism
- Cations/pharmacology
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Cells, Cultured
- Child
- Child, Preschool
- Gadolinium/pharmacology
- Humans
- Hypotonic Solutions/pharmacology
- Membrane Potentials/drug effects
- Membrane Potentials/physiology
- Middle Aged
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
Collapse
Affiliation(s)
- Clarisse Vandebrouck
- Laboratoire de Biomembranes et Signalisation Cellulaire, UMR CNRS/Université de Poitiers 6558, 86022 Poitiers Cedex, France.
| | | | | | | |
Collapse
|
50
|
Lambert MI, Marcus P, Burgess T, Noakes TD. Electro-membrane microcurrent therapy reduces signs and symptoms of muscle damage. Med Sci Sports Exerc 2002; 34:602-7. [PMID: 11932567 DOI: 10.1097/00005768-200204000-00007] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Delayed onset muscle soreness (DOMS) occurs after unaccustomed physical activity or competitive sport, resulting in stiff, painful muscles with impaired function. Acustat electro-membrane microcurrent therapy has been used to treat postoperative pain and soft tissue injury; however, its efficacy in reducing symptoms of muscle damage is not known. METHODS Thirty healthy men were recruited for a double-blind, placebo-controlled trial. The muscles of their nondominant arms were damaged using an eccentric-exercise protocol. Subjects were then randomly assigned to treatment with either Acustat or a matching placebo membrane for 96 h and monitored for a total of 168 h. RESULTS Subjects in both groups experienced severe pain and swelling of the elbow flexors after the eccentric exercise. After 24 h, the elbow joint angle of the placebo group had increased significantly more than those in the Acustat group (13.7 +/- 8.9 degrees vs 7.5 +/- 5.5 degrees; placebo vs Acustat, P < 0.05), possibly as a consequence of the elbow flexor muscles shortening. For the first 48 h after exercise, maximum voluntary contraction of the elbow flexor muscles was significantly impaired in the placebo group by up to 25% (P < 0.05), whereas muscle function was unchanged in the Acustat group. Peak plasma creatine kinase activity was also lower in the Acustat group (peak = 777 +/- 1438 U.L-1) versus the placebo group (peak = 1918 +/- 2067 U.L-1; (P < 0.05). The membranes were well tolerated by the subjects in both groups without any adverse effects. CONCLUSION These data show that treatment of muscle damage with Acustat electro-membrane microcurrent therapy reduces the severity of the symptoms. The mechanisms of action are unknown but are likely related to maintenance of intracellular Ca2+ homeostasis after muscle damaging exercise.
Collapse
Affiliation(s)
- Michael I Lambert
- MRC/UCT Research Unit for Exercise Science and Sports Medicine, P.O. Box 115, Newlands, South Africa.
| | | | | | | |
Collapse
|