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
|
Li J, Zhang Z, Bo H, Zhang Y. Exercise couples mitochondrial function with skeletal muscle fiber type via ROS-mediated epigenetic modification. Free Radic Biol Med 2024; 213:409-425. [PMID: 38295887 DOI: 10.1016/j.freeradbiomed.2024.01.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/21/2024] [Indexed: 02/04/2024]
Abstract
Skeletal muscle is a heterogeneous tissue composed of different types of muscle fibers, demonstrating substantial plasticity. Physiological or pathological stimuli can induce transitions in muscle fiber types. However, the precise regulatory mechanisms behind these transitions remains unclear. This paper reviews the classification and characteristics of muscle fibers, along with the classical mechanisms of muscle fiber type transitions. Additionally, the role of exercise-induced muscle fiber type transitions in disease intervention is reviewed. Epigenetic pathways mediate cellular adaptations and thus represent potential targets for regulating muscle fiber type transitions. This paper focuses on the mechanisms by which epigenetic modifications couple mitochondrial function and contraction characteristics. Reactive Oxygen Species (ROS) are critical signaling regulators for the health-promoting effects of exercise. Finally, we discuss the role of exercise-induced ROS in regulating epigenetic modifications and the transition of muscle fiber types.
Collapse
Affiliation(s)
- Jialin Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin, 301617, China
| | - Ziyi Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin, 301617, China.
| | - Hai Bo
- Department of Military Training Medicines, Logistics University of Chinese People's Armed Police Force, Tianjin, 300162, China.
| | - Yong Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin, 301617, China.
| |
Collapse
|
3
|
Fadel L, Dacic M, Fonda V, Sokolsky BA, Quagliarini F, Rogatsky I, Uhlenhaut NH. Modulating glucocorticoid receptor actions in physiology and pathology: Insights from coregulators. Pharmacol Ther 2023; 251:108531. [PMID: 37717739 PMCID: PMC10841922 DOI: 10.1016/j.pharmthera.2023.108531] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023]
Abstract
Glucocorticoids (GCs) are a class of steroid hormones that regulate key physiological processes such as metabolism, immune function, and stress responses. The effects of GCs are mediated by the glucocorticoid receptor (GR), a ligand-dependent transcription factor that activates or represses the expression of hundreds to thousands of genes in a tissue- and physiological state-specific manner. The activity of GR is modulated by numerous coregulator proteins that interact with GR in response to different stimuli assembling into a multitude of DNA-protein complexes and facilitate the integration of these signals, helping GR to communicate with basal transcriptional machinery and chromatin. Here, we provide a brief overview of the physiological and molecular functions of GR, and discuss the roles of GR coregulators in the immune system, key metabolic tissues and the central nervous system. We also present an analysis of the GR interactome in different cells and tissues, which suggests tissue-specific utilization of GR coregulators, despite widespread functions shared by some of them.
Collapse
Affiliation(s)
- Lina Fadel
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Marija Dacic
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Physiology, Biophysics and Systems Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Vlera Fonda
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Baila A Sokolsky
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Fabiana Quagliarini
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Inez Rogatsky
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| | - N Henriette Uhlenhaut
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany; Metabolic Programming, TUM School of Life Sciences & ZIEL Institute for Food and Health, Gregor11 Mendel-Str. 2, 85354 Freising, Germany.
| |
Collapse
|
4
|
Danowska M, Strączkowski M. The Ca2+/Calmodulin-dependent Calcineurin/NFAT Signaling Pathway in the Pathogenesis of Insulin Resistance in Skeletal Muscle. Exp Clin Endocrinol Diabetes 2023; 131:589-594. [PMID: 37875146 DOI: 10.1055/a-2174-7958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Skeletal muscle is the tissue directly involved in insulin-stimulated glucose uptake. Glucose is the primary energy substrate for contracting muscles, and proper metabolism of glucose is essential for health. Contractile activity and the associated Ca2+signaling regulate functional capacity and muscle mass. A high concentration of Ca2+and the presence of calmodulin (CaM) leads to the activation of calcineurin (CaN), a protein with serine-threonine phosphatase activity. The signaling pathway linked with CaN and transcription factors like the nuclear factor of activated T cells (NFAT) is essential for skeletal muscle development and reprogramming of fast-twitch to slow-twitch fibers. CaN activation may promote metabolic adaptations in muscle cells, resulting in better insulin-stimulated glucose transport. The molecular mechanisms underlying the altered insulin response remain unclear. The role of the CaN/NFAT pathway in regulating skeletal muscle hypertrophy is better described than its involvement in the pathogenesis of insulin resistance. Thus, there are opportunities for future research in that field. This review presents the role of CaN/NFAT signaling and suggests the relationship with insulin-resistant muscles.
Collapse
Affiliation(s)
- Magdalena Danowska
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marek Strączkowski
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
5
|
Monterrubio-Ledezma F, Navarro-García F, Massieu L, Mondragón-Flores R, Soto-Ponce LA, Magaña JJ, Cisneros B. Rescue of Mitochondrial Function in Hutchinson-Gilford Progeria Syndrome by the Pharmacological Modulation of Exportin CRM1. Cells 2023; 12:275. [PMID: 36672210 PMCID: PMC9856861 DOI: 10.3390/cells12020275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/14/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disorder caused by the expression of progerin, a mutant variant of Lamin A. Recently, HGPS studies have gained relevance because unraveling its underlying mechanism would help to understand physiological aging. We previously reported that the CRM1-mediated nuclear protein export pathway is exacerbated in HGPS cells, provoking the mislocalization of numerous protein targets of CRM1. We showed that normalization of this mechanism by pharmacologically inhibiting CRM1 with LMB (specific CRM1 inhibitor), mitigates the senescent phenotype of HGPS cells. Since mitochondrial dysfunction is a hallmark of HGPS, in this study we analyze the effect of LMB on mitochondrial function. Remarkably, LMB treatment induced the recovery of mitochondrial function in HGPS cells, as shown by the improvement in mitochondrial morphology, mitochondrial membrane potential, and ATP levels, which consequently impeded the accumulation of ROS but not mitochondrial superoxide. We provide evidence that the beneficial effect of LMB is mechanistically based on a combinatory effect on mitochondrial biogenesis via upregulation of PGC-1α expression (master transcription cofactor of mitochondrial genes), and mitophagy through the recovery of lysosomal content. The use of exportin CRM1 inhibitors constitutes a promising strategy to treat HGPS and other diseases characterized by mitochondrial impairment.
Collapse
Affiliation(s)
- Feliciano Monterrubio-Ledezma
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| | - Fernando Navarro-García
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| | - Lourdes Massieu
- Department of Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
| | - Ricardo Mondragón-Flores
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| | - Luz Adriana Soto-Ponce
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| | - Jonathan J. Magaña
- Departament of Bioengineering, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey-Campus Ciudad de México, Ciudad de Mexico 14380, Mexico
- Laboratory of Genomic Medicine, Department of Genetics, National Rehabilitation Institute-Luis Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Bulmaro Cisneros
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City 07360, Mexico
| |
Collapse
|
6
|
Wang XH, Mitch WE, Price SR. Pathophysiological mechanisms leading to muscle loss in chronic kidney disease. Nat Rev Nephrol 2022; 18:138-152. [PMID: 34750550 DOI: 10.1038/s41581-021-00498-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 12/16/2022]
Abstract
Loss of muscle proteins is a deleterious consequence of chronic kidney disease (CKD) that causes a decrease in muscle strength and function, and can lead to a reduction in quality of life and increased risk of morbidity and mortality. The effectiveness of current treatment strategies in preventing or reversing muscle protein losses is limited. The limitations largely stem from the systemic nature of diseases such as CKD, which stimulate skeletal muscle protein degradation pathways while simultaneously activating mechanisms that impair muscle protein synthesis and repair. Stimuli that initiate muscle protein loss include metabolic acidosis, insulin and IGF1 resistance, changes in hormones, cytokines, inflammatory processes and decreased appetite. A growing body of evidence suggests that signalling molecules secreted from muscle can enter the circulation and subsequently interact with recipient organs, including the kidneys, while conversely, pathological events in the kidney can adversely influence protein metabolism in skeletal muscle, demonstrating the existence of crosstalk between kidney and muscle. Together, these signals, whether direct or indirect, induce changes in the levels of regulatory and effector proteins via alterations in mRNAs, microRNAs and chromatin epigenetic responses. Advances in our understanding of the signals and processes that mediate muscle loss in CKD and other muscle wasting conditions will support the future development of therapeutic strategies to reduce muscle loss.
Collapse
Affiliation(s)
- Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, GA, USA
| | - William E Mitch
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - S Russ Price
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA. .,Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
7
|
Shaker M, Khamisipour G, Sadeghipour H, Zar A, Naeimi B, Akbarzadeh S. Effect of resistance training and garlic extract on insulin sensitivity/resistance and biochemical parameters in diabetic rats. COMPARATIVE EXERCISE PHYSIOLOGY 2022. [DOI: 10.3920/cep210031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Diabetes is one of the most important endocrine diseases in the world and obesity is one of the risk factors for this disease. The aim of this study was to evaluate the effect of a resistance exercise and garlic extract on insulin sensitivity/resistance and signal pathway of white adipose tissue to brown factors in diabetic rats. A total of 48 male Wistar rats weighing 180 to 250 g were divided into six groups (n=8): healthy control (C), diabetic control (D), diabetic with garlic extract at a dose of 50 mg/kg body weight (bw) (D+50), diabetic with garlic extract dose of 200 mg/kg bw (D+200), diabetic resistance training (D+Ex), and diabetic resistance training with garlic extract dose of 200 mg/kg bw (D+Ex+200). Plasma irisin levels in the D+200 and D+Ex groups, as well as the D+Ex+200 group showed a significant increase compared to the D group (P<0.001), while in the D+50 group no significant change was observed. Compared with group D, the expression of peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α gene was significantly increased in groups D+200 and D+Ex, as well as group D+Ex+200 (P<0.001). It can be said that resistance exercise with garlic extract is effective in controlling diabetes and reducing its complications. It also increases the expression of PGC-1α and uncoupling protein 1 genes in white adipose tissue and therefore has a positive effect on beta cell function by irisin.
Collapse
Affiliation(s)
- M. Shaker
- Faculty of Science, Agriculture and New Technologies Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - G.R. Khamisipour
- Department of Laboratory Sciences, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - H. Sadeghipour
- Department of Sport Science, School of Literature and Humanities, Persian Gulf University, Bushehr, Iran
| | - A. Zar
- Department of Sport Science, School of Literature and Humanities, Persian Gulf University, Bushehr, Iran
| | - B. Naeimi
- Department of Laboratory Sciences, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - S. Akbarzadeh
- Department of Biochemistry, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
8
|
Peris-Moreno D, Cussonneau L, Combaret L, Polge C, Taillandier D. Ubiquitin Ligases at the Heart of Skeletal Muscle Atrophy Control. Molecules 2021; 26:molecules26020407. [PMID: 33466753 PMCID: PMC7829870 DOI: 10.3390/molecules26020407] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle loss is a detrimental side-effect of numerous chronic diseases that dramatically increases mortality and morbidity. The alteration of protein homeostasis is generally due to increased protein breakdown while, protein synthesis may also be down-regulated. The ubiquitin proteasome system (UPS) is a master regulator of skeletal muscle that impacts muscle contractile properties and metabolism through multiple levers like signaling pathways, contractile apparatus degradation, etc. Among the different actors of the UPS, the E3 ubiquitin ligases specifically target key proteins for either degradation or activity modulation, thus controlling both pro-anabolic or pro-catabolic factors. The atrogenes MuRF1/TRIM63 and MAFbx/Atrogin-1 encode for key E3 ligases that target contractile proteins and key actors of protein synthesis respectively. However, several other E3 ligases are involved upstream in the atrophy program, from signal transduction control to modulation of energy balance. Controlling E3 ligases activity is thus a tempting approach for preserving muscle mass. While indirect modulation of E3 ligases may prove beneficial in some situations of muscle atrophy, some drugs directly inhibiting their activity have started to appear. This review summarizes the main signaling pathways involved in muscle atrophy and the E3 ligases implicated, but also the molecules potentially usable for future therapies.
Collapse
|
9
|
Tai YK, Ng C, Purnamawati K, Yap JLY, Yin JN, Wong C, Patel BK, Soong PL, Pelczar P, Fröhlich J, Beyer C, Fong CHH, Ramanan S, Casarosa M, Cerrato CP, Foo ZL, Pannir Selvan RM, Grishina E, Degirmenci U, Toh SJ, Richards PJ, Mirsaidi A, Wuertz‐Kozak K, Chong SY, Ferguson SJ, Aguzzi A, Monici M, Sun L, Drum CL, Wang J, Franco‐Obregón A. Magnetic fields modulate metabolism and gut microbiome in correlation with
Pgc‐1α
expression: Follow‐up to an in vitro magnetic mitohormetic study. FASEB J 2020; 34:11143-11167. [DOI: 10.1096/fj.201903005rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 06/07/2020] [Accepted: 06/15/2020] [Indexed: 01/07/2023]
Affiliation(s)
- Yee Kit Tai
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Charmaine Ng
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Kristy Purnamawati
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Jasmine Lye Yee Yap
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Jocelyn Naixin Yin
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Craig Wong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Bharati Kadamb Patel
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Poh Loong Soong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Pawel Pelczar
- Centre for Transgenic Models University of Basel Basel Switzerland
- Institute of Laboratory Animal Science University of Zürich Zürich Switzerland
| | | | - Christian Beyer
- Centre Suisse d'électronique et de microtechnique, CSEM SA Neuchatel Switzerland
| | - Charlene Hui Hua Fong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Sharanya Ramanan
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Marco Casarosa
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence Florence Italy
- Institute for Biomechanics ETH Zürich Zürich Switzerland
| | | | - Zi Ling Foo
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Rina Malathi Pannir Selvan
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Elina Grishina
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Ufuk Degirmenci
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
| | - Shi Jie Toh
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Pete J. Richards
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Ali Mirsaidi
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Karin Wuertz‐Kozak
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
- Department of Biomedical Engineering Rochester Institute of Technology (RIT) Rochester NY USA
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Suet Yen Chong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Stephen J. Ferguson
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Adriano Aguzzi
- Institut für Neuropathologie Universitätsspital Zürich Zürich Switzerland
| | - Monica Monici
- ASAcampus JL, ASA Res. Div. ‐ Dept. of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence Florence Italy
| | - Lei Sun
- DUKE‐NUS Graduate Medical School Singapore Singapore Singapore
| | - Chester L. Drum
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Jiong‐Wei Wang
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Alfredo Franco‐Obregón
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Institute for Health Innovation & Technology, iHealthtech National University of Singapore Singapore Singapore
| |
Collapse
|
10
|
Shykholeslami Z, Abdi A, Barari A, Hosseini SA. The effect of aerobic training with Citrus aurantium L. on SIRT1 and PGC-1α gene expression levels in the liver tissue of elderly rats. JORJANI BIOMEDICINE JOURNAL 2019. [DOI: 10.29252/jorjanibiomedj.7.4.57] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
|
11
|
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: 40] [Impact Index Per Article: 6.7] [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
|
12
|
dos Santos JPA, Vizuete AF, Gonçalves CA. Calcineurin-Mediated Hippocampal Inflammatory Alterations in Streptozotocin-Induced Model of Dementia. Mol Neurobiol 2019; 57:502-512. [DOI: 10.1007/s12035-019-01718-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/23/2019] [Indexed: 01/26/2023]
|
13
|
Popov DV, Lysenko EA, Bokov RO, Volodina MA, Kurochkina NS, Makhnovskii PA, Vyssokikh MY, Vinogradova OL. Effect of aerobic training on baseline expression of signaling and respiratory proteins in human skeletal muscle. Physiol Rep 2018; 6:e13868. [PMID: 30198217 PMCID: PMC6129775 DOI: 10.14814/phy2.13868] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/30/2022] Open
Abstract
Most studies examining the molecular mechanisms underlying adaptation of human skeletal muscles to aerobic exercise focused on the response to acute exercise. Here, we examined the effect of a 2-month aerobic training program on baseline parameters in human muscle. Ten untrained males performed a one-legged knee extension exercise for 1 h with the same relative intensity before and after a 2-month aerobic training program. Biopsy samples were taken from vastus lateralis muscle at rest before and after the 2 month training program (baseline samples). Additionally, biopsy samples were taken from the exercised leg 1 and 4 h after the one-legged continuous knee extension exercise. Aerobic training decreases baseline phosphorylation of FOXO1Ser256 , increases that of CaMKIIThr286 , CREB1Ser133 , increases baseline expression of mitochondrial proteins in respiratory complexes I-V, and some regulators of mitochondrial biogenesis (TFAM, NR4A3, and CRTC2). An increase in the baseline content of these proteins was not associated with a change in baseline expression of their genes. The increase in the baseline content of regulators of mitochondrial biogenesis (TFAM and NR4A3) was associated with a transient increase in transcription after acute exercise. Contrariwise, the increase in the baseline content of respiratory proteins does not seem to be regulated at the transcriptional level; rather, it is associated with other mechanisms. Adaptation of human skeletal muscle to regular aerobic exercise is associated not only with transient molecular responses to exercise, but also with changes in baseline phosphorylation and expression of regulatory proteins.
Collapse
Affiliation(s)
- Daniil V. Popov
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
- Faculty of Fundamental MedicineM.V. Lomonosov Moscow State UniversityMoscowRussia
| | - Evgeny A. Lysenko
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
- Faculty of Fundamental MedicineM.V. Lomonosov Moscow State UniversityMoscowRussia
| | - Roman O. Bokov
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| | - Maria A. Volodina
- Laboratory of Mitochondrial MedicineResearch Center for ObstetricsGynecology and PerinatologyMinistry of Healthcare of the Russian FederationMoscowRussia
| | - Nadia S. Kurochkina
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| | - Pavel A. Makhnovskii
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| | - Mikhail Y. Vyssokikh
- Laboratory of Mitochondrial MedicineResearch Center for ObstetricsGynecology and PerinatologyMinistry of Healthcare of the Russian FederationMoscowRussia
| | - Olga L. Vinogradova
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
- Faculty of Fundamental MedicineM.V. Lomonosov Moscow State UniversityMoscowRussia
| |
Collapse
|
14
|
Xie Y, Perry BD, Espinoza D, Zhang P, Price SR. Glucocorticoid-induced CREB activation and myostatin expression in C2C12 myotubes involves phosphodiesterase-3/4 signaling. Biochem Biophys Res Commun 2018; 503:1409-1414. [PMID: 30025893 PMCID: PMC6173943 DOI: 10.1016/j.bbrc.2018.07.056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 11/02/2022]
Abstract
Muscle atrophy in metabolic conditions like chronic kidney disease (CKD) and diabetes are associated with glucocorticoid production, dysfunctional insulin/Akt/FoxO3 signaling and increased myostatin expression. We recently found that CREB, a transcription factor proposed to regulate myostatin expression, is highly phosphorylated in some wasting conditions. Based on a novel Akt-PDE3/4 signaling paradigm, we hypothesized that reduced Akt signaling contributes to CREB activation and myostatin expression. C2C12 myotubes were incubated with dexamethasone (Dex), an atrophy-inducing synthetic glucocorticoid. Akt/CREB signaling and myostatin expression were evaluated by immunoblot and qPCR analyses. Inhibitors of Akt, phosphodiesterase (PDE)-3/4, and protein kinase A (PKA) signaling were used to test our hypothesis. Incubating myotubes with Dex for 3-24 h inhibited Akt phosphorylation and enhanced CREB phosphorylation as well as myostatin mRNA and protein. Inhibition of PI3K/Akt signaling with LY294002 similarly increased CREB phosphorylation. Isobutyl-methylxanthine (IBMX, a pan PDE inhibitor), milrinone (PDE3 inhibitor) and rolipram (PDE4 inhibitor) augmented CREB phosphorylation and myostatin expression. Inhibition of protein kinase A by PKI reverted Dex- or IBMX-induced CREB phosphorylation and myostatin expression. Our study provides evidence supporting a newly identified mechanism by which a glucocorticoid-related reduction in Akt signaling contributes to myostatin expression via CREB activation.
Collapse
Affiliation(s)
- Yang Xie
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA; Department of Nephrology, Xiangya Hospital and Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China; Department of Nephrology, Beijing Hospital, Beijing 100730, PR China
| | - Ben D Perry
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA; Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA; School of Science and Health, Western Sydney University, Campbelltown NSW 2560, Australia
| | - Daniel Espinoza
- Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - Peng Zhang
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA; Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - S Russ Price
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA; Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA; Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
15
|
Brocca L, McPhee JS, Longa E, Canepari M, Seynnes O, De Vito G, Pellegrino MA, Narici M, Bottinelli R. Structure and function of human muscle fibres and muscle proteome in physically active older men. J Physiol 2017; 595:4823-4844. [PMID: 28452077 DOI: 10.1113/jp274148] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Loss of muscle mass and strength in the growing population of elderly people is a major health concern for modern societies. This condition, termed sarcopenia, is a major cause of falls and of the subsequent increase in morbidity and mortality. Despite numerous studies on the impact of ageing on individual muscle fibres, the contribution of single muscle fibre adaptations to ageing-induced atrophy and functional impairment is still unsettled. The level of physical function and disuse is often associated with ageing. We studied relatively healthy older adults in order to understand the effects of ageing per se without the confounding impact of impaired physical function. We found that in healthy ageing, structural and functional alterations of muscle fibres occur. Protein post-translational modifications, oxidation and phosphorylation contribute to such alterations more than loss of myosin and other muscle protein content. ABSTRACT Contradictory results have been reported on the impact of ageing on structure and functions of skeletal muscle fibres, likely to be due to a complex interplay between ageing and other phenomena such as disuse and diseases. Here we recruited healthy, physically and socially active young (YO) and elderly (EL) men in order to study ageing per se without the confounding effects of impaired physical function. In vivo analyses of quadriceps and in vitro analyses of vastus lateralis muscle biopsies were performed. In EL subjects, our results show that (i) quadriceps volume, maximum voluntary contraction isometric torque and patellar tendon force were significantly lower; (ii) muscle fibres went through significant atrophy and impairment of specific force (isometric force/cross-sectional area) and unloaded shortening velocity; (iii) myosin/actin ratio and myosin content in individual muscle fibres were not altered; (iv) the muscle proteome went through quantitative adaptations, namely an up-regulation of the content of several groups of proteins among which were myofibrillar proteins and antioxidant defence systems; (v) the muscle proteome went through qualitative adaptations, namely phosphorylation of several proteins, including myosin light chain-2 slow and troponin T and carbonylation of myosin heavy chains. The present results indicate that impairment of individual muscle fibre structure and function is a major feature of ageing per se and that qualitative adaptations of muscle proteome are likely to be more involved than quantitative adaptations in determining such a phenomenon.
Collapse
Affiliation(s)
- Lorenza Brocca
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Interuniversity Institute of Myology, University of Pavia, Pavia, Italy
| | - Jamie S McPhee
- School of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Emanuela Longa
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Monica Canepari
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Interuniversity Institute of Myology, University of Pavia, Pavia, Italy
| | - Olivier Seynnes
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Giuseppe De Vito
- Institute for Sport and Health, University College Dublin, Ireland
| | - Maria Antonietta Pellegrino
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Interuniversity Institute of Myology, University of Pavia, Pavia, Italy.,Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia, Italy
| | - Marco Narici
- School of Graduate Entry to Medicine and Health, Division of Clinical Physiology, University of Nottingham, Derby, UK
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia, Italy.,Fondazione Salvatore Maugeri (IRCCS), Scientific Institute of Pavia, Pavia, Italy
| |
Collapse
|
16
|
Wang B, Zhang C, Zhang A, Cai H, Price SR, Wang XH. MicroRNA-23a and MicroRNA-27a Mimic Exercise by Ameliorating CKD-Induced Muscle Atrophy. J Am Soc Nephrol 2017; 28:2631-2640. [PMID: 28400445 DOI: 10.1681/asn.2016111213] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/14/2017] [Indexed: 11/03/2022] Open
Abstract
Muscle atrophy is a frequent complication of CKD, and exercise can attenuate the process. This study investigated the role of microRNA-23a (miR-23a) and miR-27a in the regulation of muscle mass in mice with CKD. These miRs are located in a gene cluster that is regulated by the transcription factor NFAT. CKD mice expressed less miR-23a in muscle than controls, and resistance exercise (muscle overload) increased the levels of miR-23a and miR-27a in CKD mice. Injection of an adeno-associated virus encoding the miR-23a/27a/24-2 precursor RNA into the tibialis anterior muscles of normal and CKD mice led to increases in mature miR-23a and miR-27a but not miR-24-2 in the muscles of both cohorts. Overexpression of miR-23a/miR-27a in CKD mice attenuated muscle loss, improved grip strength, increased the phosphorylation of Akt and FoxO1, and decreased the activation of phosphatase and tensin homolog (PTEN) and FoxO1 and the expression of TRIM63/MuRF1 and FBXO32/atrogin-1 proteins. Provision of miR-23a/miR-27a also reduced myostatin expression and downstream SMAD-2/3 signaling, decreased activation of caspase-3 and -7, and increased the expression of markers of muscle regeneration. Lastly, in silico miR target analysis and luciferase reporter assays in primary satellite cells identified PTEN and caspase-7 as targets of miR-23a and FoxO1 as a target of miR-27a in muscle. These findings provide new insights about the roles of the miR-23a/27a-24-2 cluster in CKD-induced muscle atrophy in mice and suggest a mechanism by which exercise helps to maintain muscle mass.
Collapse
Affiliation(s)
- Bin Wang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia.,Institute of Nephrology, Zhong Da Hospital, Southeast University, Nanjing, China
| | - Cong Zhang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia.,Division of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Aiqing Zhang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia.,Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China; and
| | - Hui Cai
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia.,Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - S Russ Price
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia.,Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Xiaonan H Wang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia;
| |
Collapse
|
17
|
Epoxyeicosatrienoic Acid as Therapy for Diabetic and Ischemic Cardiomyopathy. Trends Pharmacol Sci 2016; 37:945-962. [DOI: 10.1016/j.tips.2016.08.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/12/2016] [Accepted: 08/17/2016] [Indexed: 12/19/2022]
|
18
|
Rahnert JA, Zheng B, Hudson MB, Woodworth-Hobbs ME, Price SR. Glucocorticoids Alter CRTC-CREB Signaling in Muscle Cells: Impact on PGC-1α Expression and Atrophy Markers. PLoS One 2016; 11:e0159181. [PMID: 27404111 PMCID: PMC4942104 DOI: 10.1371/journal.pone.0159181] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/28/2016] [Indexed: 01/21/2023] Open
Abstract
Muscle wasting associated with chronic diseases has been linked to decreased expression of PGC-1α and overexpression of PGC-1α counters muscle loss. CREB, in conjunction with the CREB-regulated transcription coactivator (CRTC2), is a positive modulator of PGC-1α transcription. We previously reported that PGC-1α expression is decreased in skeletal muscle of diabetic rats despite a high level of CREB phosphorylation (i.e., activation), suggesting that CRTC2-CREB signaling may be dysregulated. In this study, the relationship between CREB/CRTC signaling and PGC-1α expression was examined in L6 myotubes treated with dexamethasone (Dex, 48h) to induce atrophy. Dex decreased PGC-1α mRNA and protein as well as the levels of CRTC1 and CRTC2 in the nucleus. Dex also altered the nuclear levels of two known regulators of CRTC2 localization; the amount of calcinuerin catalytic A subunit (CnA) was decreased whereas SIK was increased. To assess PGC-1α transcription, muscle cells were transfected with a PGC-1α luciferase reporter plasmid (PGC-1α-Luc). Dex suppressed PGC-1α luciferase activity while both isobutylmethylxanthine (IBMX) and over-expression of CRTC1 or CRTC2 increased PGC-1α-Luc activity. Mutation of the CRE binding site from PGC-1α-Luc reporter attenuated the responses to both IBMX and the CRTC proteins. Consistent with the reporter gene results, overexpression of CRTC2 produced an increase in CRTC2 in the nucleus and in PGC-1α mRNA and PGC-1α protein. Overexpression of CRTC2 was not sufficient to prevent the decrease in PGC-1α mRNA or protein by Dex. In summary, these data suggest that attenuated CREB/CRTC signaling contributes to the decrease in PGC-1α expression during atrophy.
Collapse
Affiliation(s)
- Jill A. Rahnert
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Bin Zheng
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Matthew B. Hudson
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Myra E. Woodworth-Hobbs
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - S. Russ Price
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
- * E-mail:
| |
Collapse
|
19
|
Tessier SN, Storey KB. Lessons from mammalian hibernators: molecular insights into striated muscle plasticity and remodeling. Biomol Concepts 2016; 7:69-92. [DOI: 10.1515/bmc-2015-0031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/21/2016] [Indexed: 12/19/2022] Open
Abstract
AbstractStriated muscle shows an amazing ability to adapt its structural apparatus based on contractile activity, loading conditions, fuel supply, or environmental factors. Studies with mammalian hibernators have identified a variety of molecular pathways which are strategically regulated and allow animals to endure multiple stresses associated with the hibernating season. Of particular interest is the observation that hibernators show little skeletal muscle atrophy despite the profound metabolic rate depression and mechanical unloading that they experience during long weeks of torpor. Additionally, the cardiac muscle of hibernators must adjust to low temperature and reduced perfusion, while the strength of contraction increases in order to pump cold, viscous blood. Consequently, hibernators hold a wealth of knowledge as it pertains to understanding the natural capacity of myocytes to alter structural, contractile and metabolic properties in response to environmental stimuli. The present review outlines the molecular and biochemical mechanisms which play a role in muscular atrophy, hypertrophy, and remodeling. In this capacity, four main networks are highlighted: (1) antioxidant defenses, (2) the regulation of structural, contractile and metabolic proteins, (3) ubiquitin proteosomal machinery, and (4) macroautophagy pathways. Subsequently, we discuss the role of transcription factors nuclear factor (erythroid-derived 2)-like 2 (Nrf2), Myocyte enhancer factor 2 (MEF2), and Forkhead box (FOXO) and their associated posttranslational modifications as it pertains to regulating each of these networks. Finally, we propose that comparing and contrasting these concepts to data collected from model organisms able to withstand dramatic changes in muscular function without injury will allow researchers to delineate physiological versus pathological responses.
Collapse
Affiliation(s)
- Shannon N. Tessier
- 1Department of Surgery and Center for Engineering in Medicine, Massachusetts General Hospital and Harvard Medical School, Building 114 16th Street, Charlestown, MA 02129, USA
| | - Kenneth B. Storey
- 2Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa K1S 5B6, Ontario, Canada
| |
Collapse
|
20
|
Pan LL, Ke JQ, Zhao CC, Huang SY, Shen J, Jiang XX, Wang XT. Electrical Stimulation Improves Rat Muscle Dysfunction Caused by Chronic Intermittent Hypoxia-Hypercapnia via Regulation of miRNA-Related Signaling Pathways. PLoS One 2016; 11:e0152525. [PMID: 27023369 PMCID: PMC4811440 DOI: 10.1371/journal.pone.0152525] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle dysfunction in chronic obstructive pulmonary disease (COPD) patients is common. Neuromuscular Electrical Stimulation (NMES) is a powerful exercise training that may relieve muscle dysfunction in COPD. This study investigated whether electrical stimulation may have atypical adaptations via activation of miRNA related pathways in counteracting COPD muscle dysfunction. Forty-eight male Sprague-Dawley rats were randomly assigned to 3 groups. With the exception of the rats in the control group, the experimental rats were exposed to chronic intermittent hypoxia-hypercapnia (CIHH) (9∼11%O2,5.5∼6.5%CO2) for 2 or 4 weeks. Electrical stimulation was performed immediately after each CIHH session. Following assessment of the running capacity, biopsy samples were obtained from the gastrocnemius of the rats. The miR-1, miR-133a and miR-133b levels were measured, as well as their related proteins: phosphorylation of Akt (p-AKT), PGC-1alpha (PGC-1α), histone deacetylase 4 (HDAC4) and serum response factor (SRF). Myosin heavy chainⅡa (MHCⅡa) and myosin heavy chainⅡb (MHCⅡb) were also measured to assess fiber type changes. After 2 weeks, compared with the controls, only miR-1 and miR-133a were significantly increased (p<0.05) in the exposure group. After 4 weeks, the exposure group exhibited a decreased running distance (p = 0.054) and MHCⅡa-to-MHCⅡb shift (p<0.05). PGC-1α (p = 0.051), nuclear HDAC4 (p = 0.058), HDAC4, p-AKT, PGC-1α and SRF was also significantly decreased (p<0.05). In contrast, miR-1 and miR-133a were significantly increased (p<0.05). Four weeks of electrical stimulation can partly reversed those changes, and miR-133b exhibited a transient increase after 2 weeks electrical stimulation. Our study indicate miRNAs may have roles in the response of CIHH-impaired muscle to changes during electrical stimulation.
Collapse
Affiliation(s)
- Lu-Lu Pan
- Center of Neurology and Rehabilitation, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Rehabilitation, the Traditional Chinese Medical Hospital of Wenzhou City, Wenzhou, China
| | - Jiang-Qiong Ke
- Center of Neurology and Rehabilitation, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | - Shi-Yuan Huang
- Center of Neurology and Rehabilitation, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Shen
- Center of Neurology and Rehabilitation, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xian-Xun Jiang
- Center of Neurology and Rehabilitation, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Tong Wang
- Center of Neurology and Rehabilitation, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- * E-mail:
| |
Collapse
|
21
|
Tóth A, Fodor J, Vincze J, Oláh T, Juhász T, Zákány R, Csernoch L, Zádor E. The Effect of SERCA1b Silencing on the Differentiation and Calcium Homeostasis of C2C12 Skeletal Muscle Cells. PLoS One 2015; 10:e0123583. [PMID: 25893964 PMCID: PMC4404259 DOI: 10.1371/journal.pone.0123583] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/04/2015] [Indexed: 12/12/2022] Open
Abstract
The sarcoplasmic/endoplasmic reticulum Ca2+ATPases (SERCAs) are the main Ca2+ pumps which decrease the intracellular Ca2+ level by reaccumulating Ca2+ into the sarcoplasmic reticulum. The neonatal SERCA1b is the major Ca2+ pump in myotubes and young muscle fibers. To understand its role during skeletal muscle differentiation its synthesis has been interfered with specific shRNA sequence. Stably transfected clones showing significantly decreased SERCA1b expression (cloneC1) were selected for experiments. The expression of the regulatory proteins of skeletal muscle differentiation was examined either by Western-blot at the protein level for MyoD, STIM1, calsequestrin (CSQ), and calcineurin (CaN) or by RT-PCR for myostatin and MCIP1.4. Quantitative analysis revealed significant alterations in CSQ, STIM1, and CaN expression in cloneC1 as compared to control cells. To examine the functional consequences of the decreased expression of SERCA1b, repeated Ca2+-transients were evoked by applications of 120 mM KCl. The significantly higher [Ca2+]i measured at the 20th and 40th seconds after the beginning of KCl application (112±3 and 110±3 nM vs. 150±7 and 135±5 nM, in control and in cloneC1 cells, respectively) indicated a decreased Ca2+-uptake capability which was quantified by extracting the maximal pump rate (454±41 μM/s vs. 144±24 μM/s, in control and in cloneC1 cells). Furthermore, the rate of calcium release from the SR (610±60 vs. 377±64 μM/s) and the amount of calcium released (843±75 μM vs. 576±80 μM) were also significantly suppressed. These changes were also accompanied by a reduced activity of CaN in cells with decreased SERCA1b. In parallel, cloneC1 cells showed inhibited cell proliferation and decreased myotube nuclear numbers. Moreover, while cyclosporineA treatment suppressed the proliferation of parental cultures it had no effect on cloneC1 cells. SERCA1b is thus considered to play an essential role in the regulation of [Ca2+]i and its ab ovo gene silencing results in decreased skeletal muscle differentiation.
Collapse
Affiliation(s)
- Adrienn Tóth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Vincze
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Juhász
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Róza Zákány
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- * E-mail:
| | - Ernő Zádor
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
22
|
In vivo calcium regulation in diabetic skeletal muscle. Cell Calcium 2014; 56:381-9. [PMID: 25224503 DOI: 10.1016/j.ceca.2014.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 07/23/2014] [Accepted: 08/09/2014] [Indexed: 01/30/2023]
Abstract
In skeletal muscle, dysfunctional contractile activity has been linked to impaired intracellular Ca(2+) concentration ([Ca(2+)]i) regulation. Muscle force production is impaired and fatigability and muscle fragility deteriorate with diabetes. Use of a novel in vivo model permits investigation of [Ca(2+)]i homeostasis in diabetic skeletal muscle. Within this in vivo environment we have shown that diabetes perturbs the Ca(2+) regulatory system such that resting [Ca(2+)]i homeostasis following muscle contractions is compromised and elevations of [Ca(2+)]i are exacerbated. This review considers the impact of diabetes on the capacity of skeletal muscle to regulate [Ca(2+)]i, following muscle contractions and, in particular, the relationship between muscle fatigue and elevated [Ca(2+)]i in a highly ecologically relevant circulation-intact environment. Importantly, the role of mitochondria in calcium sequestration and the possibility that diabetes impacts this process is explored. Given the profound microcirculatory dysfunction in diabetes this preparation offers the unique opportunity to study the interrelationships among microvascular function, blood-myocyte oxygen flux and [Ca(2+)]i as they relate to enhanced muscle fatigability and exercise intolerance.
Collapse
|
23
|
Cannavino J, Brocca L, Sandri M, Bottinelli R, Pellegrino MA. PGC1-α over-expression prevents metabolic alterations and soleus muscle atrophy in hindlimb unloaded mice. J Physiol 2014; 592:4575-89. [PMID: 25128574 DOI: 10.1113/jphysiol.2014.275545] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Prolonged skeletal muscle inactivity causes muscle fibre atrophy. Redox imbalance has been considered one of the major triggers of skeletal muscle disuse atrophy, but whether redox imbalance is actually the major cause or simply a consequence of muscle disuse remains of debate. Here we hypothesized that a metabolic stress mediated by PGC-1α down-regulation plays a major role in disuse atrophy. First we studied the adaptations of soleus to mice hindlimb unloading (HU) in the early phase of disuse (3 and 7 days of HU) with and without antioxidant treatment (trolox). HU caused a reduction in cross-sectional area, redox status alteration (NRF2, SOD1 and catalase up-regulation), and induction of the ubiquitin proteasome system (MuRF-1 and atrogin-1 mRNA up-regulation) and autophagy (Beclin1 and p62 mRNA up-regulation). Trolox completely prevented the induction of NRF2, SOD1 and catalase mRNAs, but not atrophy or induction of catabolic systems in unloaded muscles, suggesting that oxidative stress is not a major cause of disuse atrophy. HU mice showed a marked alteration of oxidative metabolism. PGC-1α and mitochondrial complexes were down-regulated and DRP1 was up-regulated. To define the link between mitochondrial dysfunction and disuse muscle atrophy we unloaded mice overexpressing PGC-1α. Transgenic PGC-1α animals did not show metabolic alteration during unloading, preserving muscle size through the reduction of autophagy and proteasome degradation. Our results indicate that mitochondrial dysfunction plays a major role in disuse atrophy and that compounds inducing PGC-1α expression could be useful to treat/prevent muscle atrophy.
Collapse
Affiliation(s)
- Jessica Cannavino
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy
| | - Lorenza Brocca
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy
| | - Marco Sandri
- Venetian Institute of Molecular Medicine and Dulbecco Telethon Institute, 35129, Padova, Italy Interuniversity Institute of Myology, University of Pavia, Pavia, Italy
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy Fondazione Salvatore Maugeri (IRCCS), Scientific Institute of Pavia, Pavia, Italy Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia, Italy
| | - Maria Antonietta Pellegrino
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy Interuniversity Institute of Myology, University of Pavia, Pavia, Italy Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
24
|
Hudson MB, Rahnert JA, Zheng B, Woodworth-Hobbs ME, Franch HA, Price SR. miR-182 attenuates atrophy-related gene expression by targeting FoxO3 in skeletal muscle. Am J Physiol Cell Physiol 2014; 307:C314-9. [PMID: 24871856 DOI: 10.1152/ajpcell.00395.2013] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Skeletal muscle atrophy occurs in response to a variety of conditions including chronic kidney disease, diabetes, cancer, and elevated glucocorticoids. MicroRNAs (miR) may play a role in the wasting process. Activation of the forkhead box O3 (FoxO3) transcription factor causes skeletal muscle atrophy in patients, animals, and cultured cells by increasing the expression of components of the ubiquitin-proteasome and autophagy-lysosome proteolytic systems. To identify microRNAs that potentially modulate the atrophy process, an in silico target analysis was performed and miR-182 was predicted to target FoxO3 mRNA. Using a combination of immunoblot analysis, quantitative real-time RT-PCR, and FoxO3 3'-UTR luciferase reporter genes, miR-182 was confirmed to regulate FoxO3 expression in C2C12 myotubes. Transfection of miR-182 into muscle cells decreased FoxO3 mRNA 30% and FoxO3 protein 67% (P < 0.05) and also prevented a glucocorticoid-induced upregulation of multiple FoxO3 gene targets including MAFbx/atrogin-1, autophagy-related protein 12 (ATG12), cathepsin L, and microtubule-associated protein light chain 3 (LC3). Treatment of C2C12 myotubes with dexamethasone (Dex) (1 μM, 6 h) to induce muscle atrophy decreased miR-182 expression by 63% (P < 0.05). Similarly, miR-182 was decreased 44% (P < 0.05) in the gastrocnemius muscle of rats injected with streptozotocin to induce diabetes compared with controls. Finally, miR-182 was present in exosomes isolated from the media of C2C12 myotubes and Dex increased its abundance. These data identify miR-182 as an important regulator of FoxO3 expression that participates in the control of atrophy-inducing genes during catabolic diseases.
Collapse
Affiliation(s)
- Matthew B Hudson
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia;
| | - Jill A Rahnert
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Bin Zheng
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Myra E Woodworth-Hobbs
- Nutrition and Health Sciences Ph.D. Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, Georgia
| | - Harold A Franch
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - S Russ Price
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia; Biomedical Laboratory Research and Development Service, Atlanta VA Medical Center, Decatur, Georgia
| |
Collapse
|
25
|
Hudson MB, Woodworth-Hobbs ME, Zheng B, Rahnert JA, Blount MA, Gooch JL, Searles CD, Price SR. miR-23a is decreased during muscle atrophy by a mechanism that includes calcineurin signaling and exosome-mediated export. Am J Physiol Cell Physiol 2013; 306:C551-8. [PMID: 24336651 DOI: 10.1152/ajpcell.00266.2013] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Skeletal muscle atrophy is prevalent in chronic diseases, and microRNAs (miRs) may play a key role in the wasting process. miR-23a was previously shown to inhibit the expression of atrogin-1 and muscle RING-finger protein-1 (MuRF1) in muscle. It also was reported to be regulated by cytoplasmic nuclear factor of activated T cells 3 (NFATc3) in cardiomyocytes. The objective of this study was to determine if miR-23a is regulated during muscle atrophy and to evaluate the relationship between calcineurin (Cn)/NFAT signaling and miR-23a expression in skeletal muscle cells during atrophy. miR-23a was decreased in the gastrocnemius of rats with acute streptozotocin-induced diabetes, a condition known to increase atrogin-1 and MuRF1 expression and cause atrophy. Treatment of C2C12 myotubes with dexamethasone (Dex) for 48 h also reduced miR-23a as well as RCAN1.4 mRNA, which is transcriptionally regulated by NFAT. NFATc3 nuclear localization and the amount of miR-23a decreased rapidly within 1 h of Dex administration, suggesting a link between Cn signaling and miR-23a. The level of miR-23a was lower in primary myotubes from mice lacking the α- or β-isoform of the CnA catalytic subunit than wild-type mice. Dex did not further suppress miR-23a in myotubes from Cn-deficient mice. Overexpression of CnAβ in C2C12 myotubes prevented Dex-induced suppression of miR-23a. Finally, miR-23a was present in exosomes isolated from the media of C2C12 myotubes, and Dex increased its exosomal abundance. Dex did not alter the number of exosomes released into the media. We conclude that atrophy-inducing conditions downregulate miR-23a in muscle by mechanisms involving attenuated Cn/NFAT signaling and selective packaging into exosomes.
Collapse
Affiliation(s)
- Matthew B Hudson
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Calcineurin: a poorly understood regulator of muscle mass. Int J Biochem Cell Biol 2013; 45:2173-8. [PMID: 23838168 DOI: 10.1016/j.biocel.2013.06.029] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 06/26/2013] [Accepted: 06/28/2013] [Indexed: 01/14/2023]
Abstract
This review will discuss the existing literature that has examined the role of calcineurin (CnA) in the regulation of skeletal muscle mass in conditions associated with hypertrophic growth or atrophy. Muscle mass is determined by the balance between protein synthesis and degradation which is controlled by a number of intracellular signaling pathways, most notably the insulin/IGF/phosphatidylinositol 3-kinase (PI3K)/Akt system. Despite being activated by IGF-1 and having well-described functions in the determination of muscle fiber phenotypes, calcineurin (CnA), a Ca(2+)-activated serine/threonine phosphatase, and its downstream signaling partners have garnered little attention as a regulator of muscle mass. Compared to other signaling pathways, the relatively few studies that have examined the role of CnA in the regulation of muscle size have produced discordant results. The reasons for these differences is not obvious but may be due to the selective nature of the genetic models studied, fluctuations in the endogenous level of CnA activity in various muscles, and the variable use of CnA inhibitors to inhibit CnA signaling. Despite the inconsistent nature of the outcomes, there is sufficient direct and indirect evidence to conclude that CnA plays a role in the regulation of skeletal muscle mass. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
|
27
|
Insulin resistance and muscle metabolism in chronic kidney disease. ISRN ENDOCRINOLOGY 2013; 2013:329606. [PMID: 23431467 PMCID: PMC3575670 DOI: 10.1155/2013/329606] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 01/21/2013] [Indexed: 12/31/2022]
Abstract
Insulin resistance is a common finding in chronic kidney disease (CKD) and is manifested by mild fasting hyperglycemia and abnormal glucose tolerance testing. Circulating levels of glucocorticoids are high. In muscle, changes in the insulin signaling pathway occur. An increase in the regulatory p85 subunit of Class I phosphatidylinositol 3-Kinase enzyme leads to decreased activation of the downstream effector protein kinase B (Akt). Mechanisms promoting muscle proteolysis and atrophy are unleashed. The link of Akt to the ubiquitin proteasome pathway, a major degradation pathway in muscle, is discussed. Another factor associated with insulin resistance in CKD is angiotensin II (Ang II) which appears to induce its intracellular effects through inflammatory cytokines or reactive oxygen species. Skeletal muscle ATP is depleted and the ability of AMP-activated protein kinase (AMPK) to replenish energy stores is blocked. How this can be reversed is discussed. Interleukin-6 (IL-6) levels are elevated in CKD and impair insulin signaling at the level of IRS-1. With exercise, IL-6 levels are reduced; glucose uptake and utilization are increased. For patients with CKD, exercise may improve insulin signaling and build up muscle. Treatment strategies for preventing muscle atrophy are discussed.
Collapse
|
28
|
Alamdari N, Aversa Z, Castillero E, Hasselgren PO. Acetylation and deacetylation--novel factors in muscle wasting. Metabolism 2013; 62:1-11. [PMID: 22626763 PMCID: PMC3430797 DOI: 10.1016/j.metabol.2012.03.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 03/25/2012] [Accepted: 03/29/2012] [Indexed: 11/24/2022]
Abstract
We review recent evidence that acetylation and deacetylation of cellular proteins, including transcription factors and nuclear cofactors, may be involved in the regulation of muscle mass. The level of protein acetylation is balanced by histone acetyltransferases (HATs) and histone deacetylases (HDACs) and studies suggest that this balance is perturbed in muscle wasting. Hyperacetylation of transcription factors and nuclear cofactors regulating gene transcription in muscle wasting may influence muscle mass. In addition, hyperacetylation may render proteins susceptible to degradation by different mechanisms, including intrinsic ubiquitin ligase activity exerted by HATs and by dissociation of proteins from cellular chaperones. In recent studies, inhibition of p300/HAT expression and activity and stimulation of SIRT1-dependent HDAC activity reduced glucocorticoid-induced catabolic response in skeletal muscle, providing further evidence that hyperacetylation plays a role in muscle wasting. It should be noted, however, that although several studies advocate a role of hyperacetylation in muscle wasting, apparently contradictory results have also been reported. For example, muscle atrophy caused by denervation or immobilization may be associated with reduced, rather than increased, protein acetylation. In addition, whereas hyperacetylation results in increased degradation of certain proteins, other proteins may be stabilized by increased acetylation. Thus, the role of acetylation and deacetylation in the regulation of muscle mass may be both condition- and protein-specific. The influence of HATs and HDACs on the regulation of muscle mass, as well as methods to modulate protein acetylation, is an important area for continued research aimed at preventing and treating muscle wasting.
Collapse
Affiliation(s)
- Nima Alamdari
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
29
|
Liu J, Peng Y, Cui Z, Wu Z, Qian A, Shang P, Qu L, Li Y, Liu J, Long J. Depressed mitochondrial biogenesis and dynamic remodeling in mouse tibialis anterior and gastrocnemius induced by 4-week hindlimb unloading. IUBMB Life 2012; 64:901-10. [PMID: 23024034 DOI: 10.1002/iub.1087] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 08/09/2012] [Indexed: 12/13/2022]
Abstract
Mitochondrial dynamics is highly involved in muscle atrophy, the slow twitch muscle as soleus, preferentially affected by hindlimb unloading (HU), was well characterized by mitochondrial dysfunction in biogenesis. However, the fast twitch muscles like tibialis anterior (TA) and gastrocnemius (GAS), which are the most massive parts of the hindlimb muscles, are less elucidated on mitochondrial adaptations responding to HU. To investigate the mitochondrial dynamic responses and the involved molecules mediating atrophy in TA and GAS, we studied a 4-week HU mouse model. We found GAS was preferentially affected to atrophy by unloading compared with TA. Furthermore, the depressed mitochondrial biogenesis occurred, accounting for mitochondrial loss in GAS by unloading. PGC-1α, as well as its transcriptional/post-translational modification regulators, such as p-CREB, SIRT1, and p-AMPK, were consistently reduced in response to unloading in GAS. Molecules relevant to autophagy, mitochondrial fusion, and fission, were compromised following unloading both in TA and GAS. These results suggested that TA exhibited resistance to unloading induced muscle atrophy while GAS presented significant mitochondrial loss, which might be due to the mitochondrial biogenesis suppressed by the inactivation of PGC-1α. However, both in TA and GAS muscles, a similar sedentary mitochondrial dynamics of mitochondrial fusion and fission was induced by unloading though TA exhibited little muscle atrophy.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University School of Life Science and Technology, Xi'an 710049, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Goodman CA, Mayhew DL, Hornberger TA. Recent progress toward understanding the molecular mechanisms that regulate skeletal muscle mass. Cell Signal 2011; 23:1896-906. [PMID: 21821120 PMCID: PMC3744211 DOI: 10.1016/j.cellsig.2011.07.013] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 07/15/2011] [Indexed: 01/30/2023]
Abstract
The maintenance of muscle mass is critical for health and issues associated with the quality of life. Over the last decade, extensive progress has been made with regard to our understanding of the molecules that regulate skeletal muscle mass. Not surprisingly, many of these molecules are intimately involved in the regulation of protein synthesis and protein degradation [e.g. the mammalian target of rapamycin (mTOR), eukaryotic initiation factor 2B (eIF2B), eukaryotic initiation factor 3f (eIF3f) and the forkhead box O (FoxO) transcription factors]. It is also becoming apparent that molecules which sense, or control, the energetic status of the cell play a key role in the regulation of muscle mass [e.g. AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator-1 α (PGC1α)]. In this review we will attempt to summarize the current knowledge of how these molecules regulate skeletal muscle mass.
Collapse
Affiliation(s)
- Craig A Goodman
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin Madison, Madison, WI 53706, USA.
| | | | | |
Collapse
|
31
|
Aversa Z, Alamdari N, Hasselgren PO. Molecules modulating gene transcription during muscle wasting in cancer, sepsis, and other critical illness. Crit Rev Clin Lab Sci 2011; 48:71-86. [DOI: 10.3109/10408363.2011.591365] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
Vicencio JM, Estrada M, Galvis D, Bravo R, Contreras AE, Rotter D, Szabadkai G, Hill JA, Rothermel BA, Jaimovich E, Lavandero S. Anabolic androgenic steroids and intracellular calcium signaling: a mini review on mechanisms and physiological implications. Mini Rev Med Chem 2011; 11:390-8. [PMID: 21443511 DOI: 10.2174/138955711795445880] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 01/21/2011] [Indexed: 02/05/2023]
Abstract
Increasing evidence suggests that nongenomic effects of testosterone and anabolic androgenic steroids (AAS) operate concertedly with genomic effects. Classically, these responses have been viewed as separate and independent processes, primarily because nongenomic responses are faster and appear to be mediated by membrane androgen receptors, whereas long-term genomic effects are mediated through cytosolic androgen receptors regulating transcriptional activity. Numerous studies have demonstrated increases in intracellular Ca2+ in response to AAS. These Ca2+ mediated responses have been seen in a diversity of cell types, including osteoblasts, platelets, skeletal muscle cells, cardiac myocytes and neurons. The versatility of Ca2+ as a second messenger provides these responses with a vast number of pathophysiological implications. In cardiac cells, testosterone elicits voltage-dependent Ca2+ oscillations and IP3R-mediated Ca2+ release from internal stores, leading to activation of MAPK and mTOR signaling that promotes cardiac hypertrophy. In neurons, depending upon concentration, testosterone can provoke either physiological Ca2+ oscillations, essential for synaptic plasticity, or sustained, pathological Ca2+ transients that lead to neuronal apoptosis. We propose therefore, that Ca2+ acts as an important point of crosstalk between nongenomic and genomic AAS signaling, representing a central regulator that bridges these previously thought to be divergent responses.
Collapse
Affiliation(s)
- J M Vicencio
- Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences/Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen GQ, Mou CY, Yang YQ, Wang S, Zhao ZW. Exercise training has beneficial anti-atrophy effects by inhibiting oxidative stress-induced MuRF1 upregulation in rats with diabetes. Life Sci 2011; 89:44-9. [PMID: 21620866 DOI: 10.1016/j.lfs.2011.04.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 04/25/2011] [Accepted: 04/27/2011] [Indexed: 11/25/2022]
Abstract
AIMS MuRF1 E3 ubiquitin ligase has been identified as a mediator of skeletal muscle wasting in various skeletal muscle atrophy models, and its expression is upregulated by oxidative stress. Exercise training could decrease oxidative stress and restore the atrophied skeletal muscle. Here, our aim was to investigate whether exercise training has any effect on MuRF1 expression in rats with diabetes. MAIN METHODS Rats with streptozotocin-induced diabetes were subjected to exercise training, after which oxidative stress was determined, and MuRF1 expression was analyzed by immunohistochemistry, real-time RT-PCR and Western blot analysis. In addition, we analyzed C2C12 myotubes in an in vitro model to examine the effects of oxidative stress on the protein levels of MuRF1 and myosin heavy chain (MHC). KEY FINDINGS While oxidative stress and MuRF1 expression were increased in rats with diabetes, exercise training diminished the skeletal muscle wasting in diabetic rats by decreasing oxidative stress and inhibiting MuRF1 expression at both the mRNA and protein levels. In addition, oxidative stress-induced MuRF1 upregulation promoted proteasome dependent degradation of the myosin heavy chain (MHC) in C2C12 myotubes. SIGNIFICANCE Our study provides the first evidence that the beneficial anti-atrophy effects of exercise training on diabetes might be mediated by inhibiting oxidative stress-induced MuRF1 upregulation and preventing MuRF1-mediated degradation of MHC.
Collapse
Affiliation(s)
- Guo-Qing Chen
- Department of Health Science, Wuhan Institute of Physical Education, Luoyu Road No. 461, Wuhan City, Hubei Province, China
| | | | | | | | | |
Collapse
|
34
|
Silencing SERCA1b in a few fibers stimulates growth in the entire regenerating soleus muscle. Histochem Cell Biol 2010; 135:11-20. [DOI: 10.1007/s00418-010-0766-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2010] [Indexed: 11/26/2022]
|