1
|
Zhou Y, Zhang X, Baker JS, Davison GW, Yan X. Redox signaling and skeletal muscle adaptation during aerobic exercise. iScience 2024; 27:109643. [PMID: 38650987 PMCID: PMC11033207 DOI: 10.1016/j.isci.2024.109643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Redox regulation is a fundamental physiological phenomenon related to oxygen-dependent metabolism, and skeletal muscle is mainly regarded as a primary site for oxidative phosphorylation. Several studies have revealed the importance of reactive oxygen and nitrogen species (RONS) in the signaling process relating to muscle adaptation during exercise. To date, improving knowledge of redox signaling in modulating exercise adaptation has been the subject of comprehensive work and scientific inquiry. The primary aim of this review is to elucidate the molecular and biochemical pathways aligned to RONS as activators of skeletal muscle adaptation and to further identify the interconnecting mechanisms controlling redox balance. We also discuss the RONS-mediated pathways during the muscle adaptive process, including mitochondrial biogenesis, muscle remodeling, vascular angiogenesis, neuron regeneration, and the role of exogenous antioxidants.
Collapse
Affiliation(s)
- Yingsong Zhou
- Faculty of Sports Science, Ningbo University, Ningbo, China
| | - Xuan Zhang
- School of Wealth Management, Ningbo University of Finance and Economics, Ningbo, China
| | - Julien S. Baker
- Centre for Health and Exercise Science Research, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong
| | - Gareth W. Davison
- Sport and Exercise Sciences Research Institute, Ulster University, Belfast BT15 IED, UK
| | - Xiaojun Yan
- School of Marine Sciences, Ningbo University, Ningbo, China
| |
Collapse
|
2
|
Blottner D, Moriggi M, Trautmann G, Furlan S, Block K, Gutsmann M, Torretta E, Barbacini P, Capitanio D, Rittweger J, Limper U, Volpe P, Gelfi C, Salanova M. Nitrosative Stress in Astronaut Skeletal Muscle in Spaceflight. Antioxidants (Basel) 2024; 13:432. [PMID: 38671880 PMCID: PMC11047620 DOI: 10.3390/antiox13040432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Long-duration mission (LDM) astronauts from the International Space Station (ISS) (>180 ISS days) revealed a close-to-normal sarcolemmal nitric oxide synthase type-1 (NOS1) immunoexpression in myofibers together with biochemical and quantitative qPCR changes in deep calf soleus muscle. Nitro-DIGE analyses identified functional proteins (structural, metabolic, mitochondrial) that were over-nitrosylated post- vs. preflight. In a short-duration mission (SDM) astronaut (9 ISS days), s-nitrosylation of a nodal protein of the glycolytic flux, specific proteins in tricarboxylic acid (TCA) cycle, respiratory chain, and over-nitrosylation of creatine kinase M-types as signs of impaired ATP production and muscle contraction proteins were seen. S-nitrosylation of serotransferrin (TF) or carbonic anhydrase 3 (CA3b and 3c) represented signs of acute response microgravity muscle maladaptation. LDM nitrosoprofiles reflected recovery of mitochondrial activity, contraction proteins, and iron transporter TF as signs of muscle adaptation to microgravity. Nitrosated antioxidant proteins, alcohol dehydrogenase 5/S-nitrosoglutathione reductase (ADH5/GSNOR), and selenoprotein thioredoxin reductase 1 (TXNRD1) levels indicated signs of altered redox homeostasis and reduced protection from nitrosative stress in spaceflight. This work presents a novel spaceflight-generated dataset on s-nitrosylated muscle protein signatures from astronauts that helps both to better understand the structural and molecular networks associated to muscular nitrosative stress and to design countermeasures to dysfunction and impaired performance control in human spaceflight missions.
Collapse
Affiliation(s)
- Dieter Blottner
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
- NeuroMuscular System and Signaling Group, Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (M.M.); (P.B.); (D.C.); (C.G.)
| | - Gabor Trautmann
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
| | - Sandra Furlan
- C.N.R. Neuroscience Institute, I-35121 Padova, Italy;
| | - Katharina Block
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
| | - Martina Gutsmann
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
| | - Enrica Torretta
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy;
| | - Pietro Barbacini
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (M.M.); (P.B.); (D.C.); (C.G.)
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (M.M.); (P.B.); (D.C.); (C.G.)
| | - Joern Rittweger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (J.R.); (U.L.)
| | - Ulrich Limper
- Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (J.R.); (U.L.)
- Anesthesiology and Intensive Care Medicine, Merheim Medical Center, Witten/Herdecke University, 51109 Cologne, Germany
| | - Pompeo Volpe
- Department of Biomedical Sciences, Università di Padova, I-35121 Padova, Italy;
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (M.M.); (P.B.); (D.C.); (C.G.)
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy;
| | - Michele Salanova
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
- NeuroMuscular System and Signaling Group, Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany
| |
Collapse
|
3
|
Gorji AE, Ostaszewski P, Urbańska K, Sadkowski T. Does β-Hydroxy-β-Methylbutyrate Have Any Potential to Support the Treatment of Duchenne Muscular Dystrophy in Humans and Animals? Biomedicines 2023; 11:2329. [PMID: 37626825 PMCID: PMC10452677 DOI: 10.3390/biomedicines11082329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Skeletal muscle is the protein reservoir of our body and an important regulator of glucose and lipid homeostasis. The dystrophin gene is the largest gene and has a key role in skeletal muscle construction and function. Mutations in the dystrophin gene cause Duchenne and Becker muscular dystrophy in humans, mice, dogs, and cats. Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular condition causing progressive muscle weakness and premature death. β-hydroxy β-methylbutyrate (HMB) prevents deleterious muscle responses under pathological conditions, including tumor and chronic steroid therapy-related muscle losses. The use of HMB as a dietary supplement allows for increasing lean weight gain; has a positive immunostimulatory effect; is associated with decreased mortality; and attenuates sarcopenia in elderly animals and individuals. This study aimed to identify some genes, metabolic pathways, and biological processes which are common for DMD and HMB based on existing literature and then discuss the consequences of that interaction.
Collapse
Affiliation(s)
- Abdolvahab Ebrahimpour Gorji
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (A.E.G.); (P.O.)
| | - Piotr Ostaszewski
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (A.E.G.); (P.O.)
| | - Kaja Urbańska
- Department of Morphological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland;
| | - Tomasz Sadkowski
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (A.E.G.); (P.O.)
| |
Collapse
|
4
|
L-Arginine/nitric oxide regulates skeletal muscle development via muscle fibre-specific nitric oxide/mTOR pathway in chickens. ANIMAL NUTRITION 2022; 10:68-85. [PMID: 35647326 PMCID: PMC9125674 DOI: 10.1016/j.aninu.2022.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/29/2021] [Accepted: 04/24/2022] [Indexed: 11/21/2022]
Abstract
L-Arginine (L-Arg), the precursor of nitric oxide (NO), plays an important role in muscle function. Fast-twitch glycolytic fibres are more susceptible to age-related atrophy than slow-twitch oxidative fibres. The effect of L-Arg/NO on protein metabolism of fast- and slow-twitch muscle fibres was evaluated in chickens. In Exp. 1, 48 chicks at 1 day old were divided into 4 groups of 12 birds and subjected to 4 treatments: basal diet without supplementation or supplemented with 1% L-Arg, and water supplemented with or without L-nitro-arginine methyl ester (L-NAME, 18.5 mM). In Exp. 2, 48 chicks were divided into 4 groups of 12 birds fed with the basal diet and subjected to the following treatments: tap water (control), tap water supplemented with L-NAME (18.5 mM), or molsidomine (MS, 0.1 mM), or 18.5 mM L-NAME + 0.1 mM MS (NAMS). The regulatory effect of L-Arg/NO was further investigated in vitro with myoblasts obtained from chicken embryo pectoralis major (PM) and biceps femoris (BF). In vivo, dietary L-Arg supplementation increased breast (+14.94%, P < 0.05) and thigh muscle mass (+23.40%, P < 0.05); whereas, MS treatment had no detectable influence. However, L-NAME treatment blocked the beneficial influence of L-Arg on muscle development. L-Arg decreased (P < 0.05) protein synthesis rate, phosphorylated mTOR and ribosomal protein S6 kinase beta-1 (p70S6K) levels in breast muscle, which was recovered by L-NAME treatment. In vitro, L-Arg or sodium nitroprusside (SNP) reduced protein synthesis rate, suppressed phosphorylated mTOR/p70S6K and decreased atrogin-1 and muscle RING finger 1 (MuRF1) in myoblasts from PM muscle (P < 0.05). L-NAME abolished the inhibitory effect of L-Arg on protein synthesis and the mTOR/p70S6K pathway. However, myoblasts from BF muscle showed the weak influence. Moreover, blocking the mTOR/p70S6K pathway with rapamycin suppressed protein synthesis of the 2 types of myoblasts; whereas, the protein expression of atrogin-1 and MuRF1 levels were restricted only in myoblasts from PM muscle. In conclusion, L-Arg/NO/mTOR/p70S6K pathway enhances protein accumulation and muscle development in fast-twitch glycolytic muscle in chickens. L-Arg/NO regulates protein turnover in a muscle fibre specific way, which highlights the potential clinical application in fast-twitch glycolytic muscle fibres.
Collapse
|
5
|
Sibisi NC, Snyman C, Myburgh KH, Niesler CU. Evaluating the role of nitric oxide in myogenesis in vitro. Biochimie 2021; 196:216-224. [PMID: 34838884 DOI: 10.1016/j.biochi.2021.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/30/2021] [Accepted: 11/21/2021] [Indexed: 12/13/2022]
Abstract
Skeletal muscle injury activates satellite cells to proliferate as myoblasts and migrate, differentiate and fuse with existing fibres at the site of injury. Nitric oxide (NO), a free radical produced by NO synthase, is elevated and supports healing after in vivo injury. NOS-independent elevation of NO levels in vitro is possible via donors such as molsidomine (SIN-1). We hypothesized that alterations in NO levels may directly influence myogenic processes critical for skeletal muscle wound healing. This study aimed to clarify the role of NO in myoblast proliferation, migration and differentiation. Baseline NO levels were established in vitro, whereafter NO levels were manipulated during myogenesis using l-NAME (NOS inhibitor) or SIN-1. Baseline NO levels generated by myoblasts in proliferation media did not change 1 h after stimulation. Addition of a pro-proliferative dose of HGF slightly elevated NO levels 1 h post-stimulation, whereas cell numbers assessed 24 h later increased significantly; l-NAME reduced the HGF-driven increase in NO and proliferation, reducing wound closure over 16 h. In differentiation media, NO levels increased significantly within 24 h, returning to baseline over several days. Regular addition of l-NAME to differentiating cells significantly reduced NO levels and fusion. SIN-1 increased NO levels in a dose-dependent manner, reaching maximal levels 16 h post-treatment. SIN-1, added at 0, 2 and 4 days, significantly increased myofiber area (26 ± 1.8% vs 18.6 ± 3.4% in control at 5 day, p < 0.0001), without affecting proliferation or migration. In conclusion, this study demonstrates that, during skeletal muscle regeneration, increased NO specifically stimulates myoblast differentiation.
Collapse
Affiliation(s)
- N C Sibisi
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| | - C Snyman
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| | - K H Myburgh
- Department Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - C U Niesler
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa.
| |
Collapse
|
6
|
Seppan P, Muhammed I, Mohammad ZIK, Sathyanathan SB. Pathobiology of ischiocavernosus and bulbospongiosus muscles in long-term diabetic male rats and its implication on erectile dysfunction. Aging Male 2020; 23:979-990. [PMID: 31368398 DOI: 10.1080/13685538.2019.1647160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To analyze pathobiology of ischiocavernosus (IC) and bulbospongiosus (BS) muscles in long-term diabetic male rats and its implication on erectile dysfunction (ED). METHODS Male rats were grouped into control and diabetic rats (received single injection of 60 mg/kg bw. of streptozotocin [STZ]). At 120th day, the animals were subjected to various analyses like serum hormone, penile reflex, electromyography of IC and BS muscles, after euthanasia IC and BS muscles were processed for morphological, histology, histometric analysis, immunostaining and immunoblotting synaptophysin, nNOS and NADPH diaphorase histochemistry. RESULTS Significant reduction in serum hormone level, penile reflex, reduced action potential or activity in both these muscles and wide range of histological alterations were observed in STZ rats. Muscles showed significant reduction in the diameter, volume and numerical density of the fiber in both muscles of STZ rats. Synaptophysin, nNOS and NADPH diaphorase were significantly reduced in diabetic animal IC and BS. CONCLUSION Severe neuromuscular circuitry alteration in IC and BS. Study concludes that degenerative changes in IC and BS may play a major role in ED in diabetic condition. Indicating diabetic-induced postsynaptic neuronal degeneration along with impaired motor action of the muscle and severe muscle degeneration affecting ED.
Collapse
Affiliation(s)
- Prakash Seppan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, Tamil Nadu, India
| | - Ibrahim Muhammed
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, Tamil Nadu, India
| | - Zafar Iqbal Khan Mohammad
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, Tamil Nadu, India
| | - Sathya Bharathy Sathyanathan
- Department of Electrical and Electronics Engineering, Loyola-ICAM College of Engineering and Technology, Chennai, Tamil Nadu, India
| |
Collapse
|
7
|
Alami-Durante H, Cluzeaud M, Bazin D, Vachot C, Kaushik S. Variable impacts of L-arginine or L-NAME during early life on molecular and cellular markers of muscle growth mechanisms in rainbow trout. Comp Biochem Physiol A Mol Integr Physiol 2020; 242:110652. [DOI: 10.1016/j.cbpa.2020.110652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/23/2019] [Accepted: 01/06/2020] [Indexed: 10/25/2022]
|
8
|
|
9
|
Guanine-rich RNA binding protein GRSF1 inhibits myoblast differentiation through repressing mitochondrial ROS production. Exp Cell Res 2019; 381:139-149. [DOI: 10.1016/j.yexcr.2019.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 12/23/2022]
|
10
|
Podkalicka P, Mucha O, Dulak J, Loboda A. Targeting angiogenesis in Duchenne muscular dystrophy. Cell Mol Life Sci 2019; 76:1507-1528. [PMID: 30770952 PMCID: PMC6439152 DOI: 10.1007/s00018-019-03006-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 12/28/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) represents one of the most devastating types of muscular dystrophies which affect boys already at early childhood. Despite the fact that the primary cause of the disease, namely the lack of functional dystrophin is known already for more than 30 years, DMD still remains an incurable disease. Thus, an enormous effort has been made during recent years to reveal novel mechanisms that could provide therapeutic targets for DMD, especially because glucocorticoids treatment acts mostly symptomatic and exerts many side effects, whereas the effectiveness of genetic approaches aiming at the restoration of functional dystrophin is under the constant debate. Taking into account that dystrophin expression is not restricted to muscle cells, but is present also in, e.g., endothelial cells, alterations in angiogenesis process have been proposed to have a significant impact on DMD progression. Indeed, already before the discovery of dystrophin, several abnormalities in blood vessels structure and function have been revealed, suggesting that targeting angiogenesis could be beneficial in DMD. In this review, we will summarize current knowledge about the angiogenesis status both in animal models of DMD as well as in DMD patients, focusing on different organs as well as age- and sex-dependent effects. Moreover, we will critically discuss some approaches such as modulation of vascular endothelial growth factor or nitric oxide related pathways, to enhance angiogenesis and attenuate the dystrophic phenotype. Additionally, we will suggest the potential role of other mediators, such as heme oxygenase-1 or statins in those processes.
Collapse
Affiliation(s)
- Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
11
|
A gentle introduction to gasotransmitters with special reference to nitric oxide: biological and chemical implications. REV INORG CHEM 2018. [DOI: 10.1515/revic-2018-0011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AbstractNitric oxide (NO), carbon monoxide (CO) and hydrogen sulfide (H2S) are gaseous molecules of major impact in biology. Despite their toxicity, these molecules have profound effects on mammalian physiology and major implications in therapeutics. At tiny concentrations in human biology, they play key signaling and regulatory functions and hence are now labeled as “gasotransmitters.” In this literature survey, an introduction to gasotransmitters in relevance with NO, CO and H2S has been primarily focused. A special attention has been given to the conjoint physiological, pathophysiological and therapeutic aspects of NO in this work. In addition to the aforementioned elements of the investigation being reported, this report gives a detailed account of some of the recent advancements covering the NO release from both the nitro as well as nitroso compounds. The importance of the metallic center on the eve of producing the reduction center on NO and to develop photolabile properties have been elaborated within the effect of a few examples of metallic centers. Also, theoretical investigations that have been reported in the recent past and some other current theories pertaining to NO chemistry have been enlightened in this review. From the overall study, it is eminent that a number of facts are yet to be explored in context with NO for deeper mechanistic insights, model design for these molecules, other key roles and the search to find the best fit formalism in theoretical chemistry.
Collapse
|
12
|
Lee IH, Lee YJ, Seo H, Kim YS, Nam JO, Jeon BD, Kwon TD. Study of muscle contraction induced by electrical pulse stimulation and nitric oxide in C2C12 myotube cells. J Exerc Nutrition Biochem 2018; 22:22-28. [PMID: 29673242 PMCID: PMC5909072 DOI: 10.20463/jenb.2018.0004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022] Open
Abstract
[Purpose] This study aimed to examine the independent effect of electrical pulse stimulation(EPS) and nitric oxide(NO) on muscle contraction and their synergistic or combined effect on contraction phenomenon using C2C12 mouse skeletal muscle cells. [Methods] Some differentiated C2C12 myotube cells were untreated (control). Other cells did not receive EPS and did receive 0.5, 1.0, or 2.0 mM of the NO donor, S-nitroso-N-acetyl-penicillamine (SNAP; -E/S0.5, -E/S1.0, and -E/S2.0, respectively). For the EPS treatments (0.3 V/mm, 1.0 Hz, and 4.0 ms), differentiated C2C12 myotube cells received only EPS or both EPS and the SNAPtreatments at the same concentrations (+E/-S, +E/S0.5, +E/S1.0, and +E/S2.0, respectively). All samples were then cultured for 4 days. [Results] Differentiated C2C12 cellswere stimulated by the EPS, NO, and EPS+NO treatments. The cell length of the +E/S2.0 Group after the 4-day culture (84.2±13.2㎛) was the shortest of all the groups. The expressions of AMPK, JNK, Akt, eNOS, GLUT4, and PGC1α proteins were noticeably dominant. The results indicated synergistic effect on muscle contraction of simultaneously applied EPS and SNAP. [Conclusion] Motor skills were significantly improved when exercise was accompanied by the intake of NO precursor and/or NO, compared to that upon their independent application or treatment.
Collapse
|
13
|
Le Moal E, Pialoux V, Juban G, Groussard C, Zouhal H, Chazaud B, Mounier R. Redox Control of Skeletal Muscle Regeneration. Antioxid Redox Signal 2017; 27:276-310. [PMID: 28027662 PMCID: PMC5685069 DOI: 10.1089/ars.2016.6782] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/24/2016] [Accepted: 12/27/2016] [Indexed: 12/12/2022]
Abstract
Skeletal muscle shows high plasticity in response to external demand. Moreover, adult skeletal muscle is capable of complete regeneration after injury, due to the properties of muscle stem cells (MuSCs), the satellite cells, which follow a tightly regulated myogenic program to generate both new myofibers and new MuSCs for further needs. Although reactive oxygen species (ROS) and reactive nitrogen species (RNS) have long been associated with skeletal muscle physiology, their implication in the cell and molecular processes at work during muscle regeneration is more recent. This review focuses on redox regulation during skeletal muscle regeneration. An overview of the basics of ROS/RNS and antioxidant chemistry and biology occurring in skeletal muscle is first provided. Then, the comprehensive knowledge on redox regulation of MuSCs and their surrounding cell partners (macrophages, endothelial cells) during skeletal muscle regeneration is presented in normal muscle and in specific physiological (exercise-induced muscle damage, aging) and pathological (muscular dystrophies) contexts. Recent advances in the comprehension of these processes has led to the development of therapeutic assays using antioxidant supplementation, which result in inconsistent efficiency, underlying the need for new tools that are aimed at precisely deciphering and targeting ROS networks. This review should provide an overall insight of the redox regulation of skeletal muscle regeneration while highlighting the limits of the use of nonspecific antioxidants to improve muscle function. Antioxid. Redox Signal. 27, 276-310.
Collapse
Affiliation(s)
- Emmeran Le Moal
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
- Movement, Sport and Health Sciences Laboratory, M2S, EA1274, University of Rennes 2, Bruz, France
| | - Vincent Pialoux
- Laboratoire Interuniversitaire de Biologie de la Motricité, EA7424, Université Claude Bernard Lyon 1, Univ Lyon, Villeurbanne, France
- Institut Universitaire de France, Paris, France
| | - Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| | - Carole Groussard
- Movement, Sport and Health Sciences Laboratory, M2S, EA1274, University of Rennes 2, Bruz, France
| | - Hassane Zouhal
- Movement, Sport and Health Sciences Laboratory, M2S, EA1274, University of Rennes 2, Bruz, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| |
Collapse
|
14
|
Monteiro HP, Ogata FT. S-nitrosylation/denitrosylation regulates myoblast proliferation. Focus on "Balance between S-nitrosylation and denitrosylation modulates myoblast proliferation independently of soluble guanylyl cyclase activation". Am J Physiol Cell Physiol 2017. [PMID: 28637676 DOI: 10.1152/ajpcell.00127.2017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hugo P Monteiro
- Department of Biochemistry - Center for Cellular and Molecular Therapy, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil; and
| | - Fernando T Ogata
- Division of Biochemistry, Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Yamashita AMS, Ancillotti MTC, Rangel LP, Fontenele M, Figueiredo-Freitas C, Possidonio AC, Soares CP, Sorenson MM, Mermelstein C, Nogueira L. Balance between S-nitrosylation and denitrosylation modulates myoblast proliferation independently of soluble guanylyl cyclase activation. Am J Physiol Cell Physiol 2017; 313:C11-C26. [PMID: 28381519 DOI: 10.1152/ajpcell.00140.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 03/13/2017] [Accepted: 03/31/2017] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) contributes to myogenesis by regulating the transition between myoblast proliferation and fusion through cGMP signaling. NO can form S-nitrosothiols (RSNO), which control signaling pathways in many different cell types. However, neither the role of RSNO content nor its regulation by the denitrosylase activity of S-nitrosoglutathione reductase (GSNOR) during myogenesis is understood. Here, we used primary cultures of chick embryonic skeletal muscle cells to investigate whether changes in intracellular RSNO alter proliferation and fusion of myoblasts in the presence and absence of cGMP. Cultures were grown to fuse most of the myoblasts into myotubes, with and without S-nitrosocysteine (CysNO), 8-Br-cGMP, DETA-NO, or inhibitors for NO synthase (NOS), GSNOR, soluble guanylyl cyclase (sGC), or a combination of these, followed by analysis of GSNOR activity, protein expression, RSNO, cGMP, and cell morphology. Although the activity of GSNOR increased progressively over 72 h, inhibiting GSNOR (by GSNOR inhibitor - GSNORi - or by knocking down GSNOR with siRNA) produced an increase in RSNO and in the number of myoblasts and fibroblasts, accompanied by a decrease in myoblast fusion index. This was also detected with CysNO supplementation. Enhanced myoblast number was proportional to GSNOR inhibition. Effects of the GSNORi and GSNOR knockdown were blunted by NOS inhibition, suggesting their dependence on NO synthesis. Interestingly, GSNORi and GSNOR knockdown reversed the attenuated proliferation obtained with sGC inhibition in myoblasts, but not in fibroblasts. Hence myoblast proliferation is enhanced by increasing RSNO, and regulated by GSNOR activity, independently of cGMP production and signaling.
Collapse
Affiliation(s)
- Aline M S Yamashita
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maryana T C Ancillotti
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana P Rangel
- Departamento de Análise Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; and
| | - Marcio Fontenele
- Laboratório de Biologia Molecular do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cicero Figueiredo-Freitas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana C Possidonio
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina P Soares
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martha M Sorenson
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia Mermelstein
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Nogueira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil;
| |
Collapse
|
16
|
McCormick R, Pearson T, Vasilaki A. Manipulation of environmental oxygen modifies reactive oxygen and nitrogen species generation during myogenesis. Redox Biol 2016; 8:243-51. [PMID: 26827127 PMCID: PMC4753392 DOI: 10.1016/j.redox.2016.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/18/2016] [Accepted: 01/20/2016] [Indexed: 01/29/2023] Open
Abstract
Regulated changes in reactive oxygen and nitrogen species (RONS) activities are important in maintaining the normal sequence and development of myogenesis. Both excessive formation and reduction in RONS have been shown to affect muscle differentiation in a negative way. Cultured cells are typically grown in 20% O2 but this is not an appropriate physiological concentration for a number of cell types, including skeletal muscle. The aim was to examine the generation of RONS in cultured skeletal muscle cells under a physiological oxygen concentration condition (6% O2) and determine the effect on muscle myogenesis. Primary mouse satellite cells were grown in 20% or 6% O2 environments and RONS activity was measured at different stages of myogenesis by real-time fluorescent microscopy using fluorescent probes with different specificities i.e. dihydroethidium (DHE), 4-amino-5-methylamino-2′,7′-difluorofluorescein diacetate (DAF-FM DA) and 5-(and-6)-chloromethyl-2′,7′ -dichlorodihydrofluorescein diacetate (CM-DCFH-DA). Data demonstrate that satellite cell proliferation increased when cells were grown in 6% O2 compared with 20% O2. Myoblasts grown in 20% O2 showed an increase in DCF fluorescence and DHE oxidation compared with myoblasts grown at 6% O2. Myotubes grown in 20% O2 also showed an increase in DCF and DAF-FM fluorescence and DHE oxidation compared with myotubes grown in 6% O2. The catalase and MnSOD contents were also increased in myoblasts and myotubes that were maintained in 20% O2 compared with myoblasts and myotubes grown in 6% O2. These data indicate that intracellular RONS activities in myoblasts and myotubes at rest are influenced by changes in environmental oxygen concentration and that the increased ROS may influence myogenesis in a negative manner. Environmental O2 concentrations were found to influence myogenesis in vitro. Proliferation of primary muscle cells was increased when cells were grown in 6% O2. Cells grown in 20% O2 showed increased RONS formation compared with cells in 6% O2. Cells grown in 20% O2 had higher antioxidant defence enzyme contents. These data suggest that RONS generated at 20% O2 may limit myogenesis in vitro.
Collapse
Affiliation(s)
- Rachel McCormick
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Timothy Pearson
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Aphrodite Vasilaki
- MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing, Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK.
| |
Collapse
|
17
|
Titov VY, Kondratov GV, Ivanova AV. Specific role of nitric oxide (NO) in avian embryonic myogenesis. Bull Exp Biol Med 2015; 158:508-12. [PMID: 25708336 DOI: 10.1007/s10517-015-2796-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Indexed: 11/25/2022]
Abstract
NO plays a specific role in avian embryogenesis stimulating the development of muscle tissue. The main consumer of NO synthesized at the initial stage of avian embryogenesis is presumably a factor stimulating myogenesis.
Collapse
Affiliation(s)
- V Yu Titov
- All-Russian Research and Technological Institute of Poultry Farming, The Russian Academy of Agricultural Sciences, Sergiev Posad, Moscow Region, Russia,
| | | | | |
Collapse
|
18
|
Tidball JG, Wehling-Henricks M. Nitric oxide synthase deficiency and the pathophysiology of muscular dystrophy. J Physiol 2014; 592:4627-38. [PMID: 25194047 DOI: 10.1113/jphysiol.2014.274878] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The secondary loss of neuronal nitric oxide synthase (nNOS) that occurs in dystrophic muscle is the basis of numerous, complex and interacting features of the dystrophic pathology that affect not only muscle itself, but also influence the interaction of muscle with other tissues. Many mechanisms through which nNOS deficiency contributes to misregulation of muscle development, blood flow, fatigue, inflammation and fibrosis in dystrophic muscle have been identified, suggesting that normalization in NO production could greatly attenuate diverse aspects of the pathology of muscular dystrophy through multiple regulatory pathways. However, the relative importance of the loss of nNOS from the sarcolemma versus the importance of loss of total nNOS from dystrophic muscle remains unknown. Although most current evidence indicates that nNOS localization at the sarcolemma is not required to achieve NO-mediated reductions of pathology in muscular dystrophy, the question remains open concerning whether membrane localization would provide a more efficient rescue from features of the dystrophic phenotype.
Collapse
Affiliation(s)
- James G Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, USA Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| | | |
Collapse
|
19
|
McDonald FB, Edge D, O'Halloran KD. Chronic nitric oxide synthase inhibition does not impair upper airway muscle adaptation to chronic intermittent hypoxia in the rat. PROGRESS IN BRAIN RESEARCH 2014; 212:237-51. [PMID: 25194201 DOI: 10.1016/b978-0-444-63488-7.00012-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Nitric oxide (NO) is an important modulator of striated muscle function. Nitric oxide synthase (NOS) expression and activity is altered by hypoxia and NO is implicated in respiratory muscle remodeling following chronic sustained hypoxia. We sought to determine if NO is implicated in upper airway dilator muscle adaptation to chronic intermittent hypoxia (CIH). Thirty-two adult male Wistar rats (284±13, mean±SD) were exposed to alternating bouts of hypoxia (90 s; 5% O2 at the nadir) and normoxia (210 s; 21% O2) for 12 cycles per hour, 8h/day for 3 weeks. Sham animals were exposed to normoxia in parallel. Half of the animals in both groups received the nNOS inhibitor-L-NNA (2mM) in the drinking water throughout the study (N=8 for all groups). Sternohyoid (pharyngeal dilator) muscle contractile and endurance properties were determined ex vivo. Sternohyoid muscle myosin heavy chain (MHC) isoform composition and cross-sectional area was determined by fluorescence microscopy. Chronic nNOS blockade did not alter sternohyoid muscle peak force or force-frequency relationship in sham or CIH-treated animals. In contrast, chronic nNOS blockade significantly decreased sternohyoid muscle endurance with equivalent effects in sham and CIH-treated rats. Our results suggest that NO is an important modulator of sternohyoid muscle endurance. However, our data provide no evidence to suggest that NO is implicated in upper airway muscle adaptation to CIH.
Collapse
Affiliation(s)
- Fiona B McDonald
- Health Sciences Centre, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Deirdre Edge
- Health Sciences Centre, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland; Department of Physiology, School of Medicine, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland.
| |
Collapse
|
20
|
Lee EJ, Kamli MR, Pokharel S, Malik A, Tareq KMA, Roouf Bhat A, Park HB, Lee YS, Kim S, Yang B, Young Chung K, Choi I. Expressed sequence tags for bovine muscle satellite cells, myotube formed-cells and adipocyte-like cells. PLoS One 2013; 8:e79780. [PMID: 24224006 PMCID: PMC3818215 DOI: 10.1371/journal.pone.0079780] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 09/25/2013] [Indexed: 12/25/2022] Open
Abstract
Background Muscle satellite cells (MSCs) represent a devoted stem cell population that is responsible for postnatal muscle growth and skeletal muscle regeneration. An important characteristic of MSCs is that they encompass multi potential mesenchymal stem cell activity and are able to differentiate into myocytes and adipocytes. To achieve a global view of the genes differentially expressed in MSCs, myotube formed-cells (MFCs) and adipocyte-like cells (ALCs), we performed large-scale EST sequencing of normalized cDNA libraries developed from bovine MSCs. Results A total of 24,192 clones were assembled into 3,333 clusters, 5,517 singletons and 3,842contigs. Functional annotation of these unigenes revealed that a large portion of the differentially expressed genes are involved in cellular and signaling processes. Database for Annotation, Visualization and Integrated Discovery (DAVID) functional analysis of three subsets of highly expressed gene lists (MSC233, MFC258, and ALC248) highlighted some common and unique biological processes among MSC, MFC and ALC. Additionally, genes that may be specific to MSC, MFC and ALC are reported here, and the role of dimethylargininedimethylaminohydrolase2 (DDAH2) during myogenesis and hemoglobinsubunitalpha2 (HBA2) during transdifferentiation in C2C12 were assayed as a case study. DDAH2 was up-regulated during myognesis and knockdown of DDAH2 by siRNA significantly decreased myogenin (MYOG) expression corresponding with the slight change in cell morphology. In contrast, HBA2 was up-regulated during ALC formation and resulted in decreased intracellular lipid accumulation and CD36 mRNA expression upon knockdown assay. Conclusion In this study, a large number of EST sequences were generated from the MSC, MFC and ALC. Overall, the collection of ESTs generated in this study provides a starting point for the identification of novel genes involved in MFC and ALC formation, which in turn offers a fundamental resource to enable better understanding of the mechanism of muscle differentiation and transdifferentiation.
Collapse
Affiliation(s)
- Eun Ju Lee
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
- Bovine Genome Resources Bank, Yeungnam University, Gyeongsan, Republic of Korea
| | - Majid Rasool Kamli
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Smritee Pokharel
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Adeel Malik
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - K. M. A. Tareq
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Abdul Roouf Bhat
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Hee-Bok Park
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Yong Seok Lee
- Bovine Genome Resources Bank, Yeungnam University, Gyeongsan, Republic of Korea
- Department of Life Science and Biotechnology, College of Natural Sciences, Soonchunhyang University, Asan, Korea
| | - SangHoon Kim
- Department of Biology, Kyung Hee University, Seoul, Republic of Korea
| | - Bohsuk Yang
- Hanwoo Experiment Station, National Institute of Animal Science, RDA, Pyeongchang, Seoul, Republic of Korea
| | - Ki Young Chung
- Hanwoo Experiment Station, National Institute of Animal Science, RDA, Pyeongchang, Seoul, Republic of Korea
- * E-mail: (IC); (KYC)
| | - Inho Choi
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
- Bovine Genome Resources Bank, Yeungnam University, Gyeongsan, Republic of Korea
- * E-mail: (IC); (KYC)
| |
Collapse
|
21
|
Abstract
Myoblast fusion is a critical process that contributes to the growth of muscle during development and to the regeneration of myofibers upon injury. Myoblasts fuse with each other as well as with multinucleated myotubes to enlarge the myofiber. Initial studies demonstrated that myoblast fusion requires extracellular calcium and changes in cell membrane topography and cytoskeletal organization. More recent studies have identified several cell-surface and intracellular proteins that mediate myoblast fusion. Furthermore, emerging evidence suggests that myoblast fusion is also regulated by the activation of specific cell-signaling pathways that lead to the expression of genes whose products are essential for the fusion process and for modulating the activity of molecules that are involved in cytoskeletal rearrangement. Here, we review the roles of the major signaling pathways in mammalian myoblast fusion.
Collapse
Affiliation(s)
- Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | | | | |
Collapse
|
22
|
Scully MA, Pandya S, Moxley RT. Review of Phase II and Phase III clinical trials for Duchenne muscular dystrophy. Expert Opin Orphan Drugs 2012. [DOI: 10.1517/21678707.2013.746939] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Nitric oxide in skeletal muscle: role on mitochondrial biogenesis and function. Int J Mol Sci 2012; 13:17160-84. [PMID: 23242154 PMCID: PMC3546744 DOI: 10.3390/ijms131217160] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 12/04/2012] [Accepted: 12/05/2012] [Indexed: 01/18/2023] Open
Abstract
Nitric oxide (NO) has been implicated in several cellular processes as a signaling molecule and also as a source of reactive nitrogen species (RNS). NO is produced by three isoenzymes called nitric oxide synthases (NOS), all present in skeletal muscle. While neuronal NOS (nNOS) and endothelial NOS (eNOS) are isoforms constitutively expressed, inducible NOS (iNOS) is mainly expressed during inflammatory responses. Recent studies have demonstrated that NO is also involved in the mitochondrial biogenesis pathway, having PGC-1α as the main signaling molecule. Increased NO synthesis has been demonstrated in the sarcolemma of skeletal muscle fiber and NO can also reversibly inhibit cytochrome c oxidase (Complex IV of the respiratory chain). Investigation on cultured skeletal myotubes treated with NO donors, NO precursors or NOS inhibitors have also showed a bimodal effect of NO that depends on the concentration used. The present review will discuss the new insights on NO roles on mitochondrial biogenesis and function in skeletal muscle. We will also focus on potential therapeutic strategies based on NO precursors or analogs to treat patients with myopathies and mitochondrial deficiency.
Collapse
|
24
|
Nitric oxide in myogenesis and therapeutic muscle repair. Mol Neurobiol 2012; 46:682-92. [PMID: 22821188 DOI: 10.1007/s12035-012-8311-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 07/12/2012] [Indexed: 12/20/2022]
Abstract
Nitric oxide is a short-lived intracellular and intercellular messenger. The first realisation that nitric oxide is important in physiology occurred in 1987 when its identity with the endothelium-derived relaxing factor was discovered. Subsequent studies have shown that nitric oxide possesses a number of physiological functions that are essential not only to vascular homeostasis but also to neurotransmission, such as in the processes of learning and memory and endocrine gland regulation, as well as inflammation and immune responses. The discovery in 1995 that a splice variant of the neuronal nitric oxide synthase is localised at the sarcolemma via the dystrophin-glycoprotein complex and of its displacement in Duchenne muscular dystrophy has stimulated a host of studies exploring the role of nitric oxide in skeletal muscle physiology. Recently, nitric oxide has emerged as a relevant messenger also of myogenesis that it regulates at several key steps, especially when the process is stimulated for muscle repair following acute and chronic muscle injuries. Here, we will review briefly the mechanisms and functions of nitric oxide in skeletal muscle and discuss its role in myogenesis, with specific attention to the promising nitric oxide-based approaches now being explored at the pre-clinical and clinical level for the therapy of muscular dystrophy.
Collapse
|
25
|
Ahmad N, Welch I, Grange R, Hadway J, Dhanvantari S, Hill D, Lee TY, Hoffman LM. Use of imaging biomarkers to assess perfusion and glucose metabolism in the skeletal muscle of dystrophic mice. BMC Musculoskelet Disord 2011; 12:127. [PMID: 21639930 PMCID: PMC3141608 DOI: 10.1186/1471-2474-12-127] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Accepted: 06/04/2011] [Indexed: 11/10/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease that affects 1 in 3500 boys. The disease is characterized by progressive muscle degeneration that results from mutations in or loss of the cytoskeletal protein, dystrophin, from the glycoprotein membrane complex, thus increasing the susceptibility of contractile muscle to injury. To date, disease progression is typically assessed using invasive techniques such as muscle biopsies, and while there are recent reports of the use of magnetic resonance, ultrasound and optical imaging technologies to address the issue of disease progression and monitoring therapeutic intervention in dystrophic mice, our study aims to validate the use of imaging biomarkers (muscle perfusion and metabolism) in a longitudinal assessment of skeletal muscle degeneration/regeneration in two murine models of muscular dystrophy. Methods Wild-type (w.t.) and dystrophic mice (weakly-affected mdx mice that are characterized by a point mutation in dystrophin; severely-affected mdx:utrn-/- (udx) mice that lack functional dystrophin and are null for utrophin) were exercised three times a week for 30 minutes. To follow the progression of DMD, accumulation of 18 F-FDG, a measure of glucose metabolism, in both wild-type and affected mice was measured with a small animal PET scanner (GE eXplore Vista). To assess changes in blood flow and blood volume in the hind limb skeletal muscle, mice were injected intravenously with a CT contrast agent, and imaged with a small animal CT scanner (GE eXplore Ultra). Results In hind limb skeletal muscle of both weakly-affected mdx mice and in severely-affected udx mice, we demonstrate an early, transient increase in both 18F-FDG uptake, and in blood flow and blood volume. Histological analysis of H&E-stained tissue collected from parallel littermates demonstrates the presence of both inflammatory infiltrate and centrally-located nuclei, a classic hallmark of myofibrillar regeneration. In both groups of affected mice, the early transient response was succeeded by a progressive decline in muscle perfusion and metabolism; this was also evidenced histologically. Conclusions The present study demonstrates the utility of non-invasive imaging biomarkers in characterizing muscle degeneration/regeneration in murine models of DMD. These techniques may now provide a promising alternative for assessing both disease progression and the efficacy of new therapeutic treatments in patients.
Collapse
Affiliation(s)
- Nabeel Ahmad
- Imaging Program, Lawson Health Research Institute, 268 Grosvenor St., London N6A4V2, Canada
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Molecular mechanisms of myoblast fusion across species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 713:113-35. [PMID: 21432017 DOI: 10.1007/978-94-007-0763-4_8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Skeletal muscle development, growth and regeneration depend on the ability of progenitor myoblasts to fuse to one another in a series of ordered steps. Whereas the cellular steps leading to the formation of a multinucleated myofiber are conserved in several model organisms, the molecular regulatory factors may vary. Understanding the common and divergent mechanisms regulating myoblast fusion in Drosophila melanogaster (fruit fly), Danio rerio (zebrafish) and Mus musculus (mouse) provides a better insight into the process of myoblast fusion than any of these models could provide alone. Deciphering the mechanisms of myoblast fusion from simpler to more complex organisms is of fundamental interest to skeletal muscle biology and may provide therapeutic avenues for various diseases that affect muscle.
Collapse
|
27
|
Normal fibroblasts promote myodifferentiation of myoblasts from sex-linked dwarf chicken via up-regulation of β1 integrin. Cell Biol Int 2010; 34:1119-27. [DOI: 10.1042/cbi20090351] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
28
|
Nitric oxide regulates stretch-induced proliferation in C2C12 myoblasts. J Muscle Res Cell Motil 2010; 31:215-25. [DOI: 10.1007/s10974-010-9227-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 08/05/2010] [Indexed: 10/19/2022]
|
29
|
Dahlman JM, Bakkar N, He W, Guttridge DC. NF-kappaB functions in stromal fibroblasts to regulate early postnatal muscle development. J Biol Chem 2009; 285:5479-87. [PMID: 20018862 DOI: 10.1074/jbc.m109.075606] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Classical NF-kappaB activity functions as an inhibitor of the skeletal muscle myogenic program. Recent findings reveal that even in newborn RelA/p65(-/-) mice, myofiber numbers are increased over that of wild type mice, suggesting that NF-kappaB may be a contributing factor in early postnatal skeletal muscle development. Here we show that in addition to p65 deficiency, repression of NF-kappaB with the IkappaB alpha-SR transdominant inhibitor or with muscle-specific deletion of IKKbeta resulted in similar increases in total fiber numbers as well as an up-regulation of myogenic gene products. Upon further characterization of early postnatal muscle, we observed that NF-kappaB activity progressively declines within the first few weeks of development. At birth, the majority of this activity is compartmentalized to muscle fibers, but by neonatal day 8 NF-kappaB activity from the myofibers diminishes, and instead, stromal fibroblasts become the main cellular compartment within the muscle that contains active NF-kappaB. We find that NF-kappaB functions in these fibroblasts to regulate inducible nitric-oxide synthase expression, which we show is important for myoblast fusion during the growth and maturation process of skeletal muscle. Together, these data broaden our understanding of NF-kappaB during development by showing that in addition to its role as a negative regulator of myogenesis, NF-kappaB also regulates nitric-oxide synthase expression within stromal fibroblasts to stimulate myoblast fusion and muscle hypertrophy.
Collapse
Affiliation(s)
- Jason M Dahlman
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | |
Collapse
|
30
|
Wallace GQ, McNally EM. Mechanisms of muscle degeneration, regeneration, and repair in the muscular dystrophies. Annu Rev Physiol 2009; 71:37-57. [PMID: 18808326 DOI: 10.1146/annurev.physiol.010908.163216] [Citation(s) in RCA: 232] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To withstand the rigors of contraction, muscle fibers have specialized protein complexes that buffer against mechanical stress and a multifaceted repair system that is rapidly activated after injury. Genetic studies first identified the mechanosensory signaling network that connects the structural elements of muscle and, more recently, have identified repair elements of muscle. Defects in the genes encoding the components of these systems lead to muscular dystrophy, a family of genetic disorders characterized by progressive muscle wasting. Although the age of onset, affected muscles, and severity vary considerably, all muscular dystrophies are characterized by muscle necrosis that overtakes the regenerative capacity of muscle. The resulting replacement of muscle by fatty and fibrous tissue leaves muscle increasingly weak and nonfunctional. This review discusses the cellular mechanisms that are primarily and secondarily disrupted in muscular dystrophy, focusing on membrane degeneration, muscle regeneration, and the repair of muscle.
Collapse
Affiliation(s)
- Gregory Q Wallace
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
31
|
Benabdellah F, Yu H, Brunelle A, Laprévote O, De La Porte S. MALDI reveals membrane lipid profile reversion in MDX mice. Neurobiol Dis 2009; 36:252-8. [PMID: 19632329 DOI: 10.1016/j.nbd.2009.07.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 06/25/2009] [Accepted: 07/16/2009] [Indexed: 11/30/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), the most common and severe X-linked myopathy, is characterized by the lack of dystrophin, a sub-sarcolemmal protein necessary for normal muscle functions. In a previous study of the lipid content of skeletal muscles of dystrophic (mdx) mice, the animal model for DMD, by in situ Matrix-Assisted Laser Desorption-Ionization Mass Spectrometry (MALDI-MS), an inversion of the phosphatidylcholine PC34:2/PC34:1 ion peaks intensity ratio was observed between destructured (abnormal fiber morphology) and structured (normal fiber morphology). A possible treatment for this dramatic disease is to introduce an exogenous nitric oxide (NO) donor into the organism, leading to an increase of utrophin and a regression of the dystrophic phenotype. In the present work, after confirmation by tandem mass spectrometry of the structure of these two phospholipids, their intensity ratio inversion was used to evidence a restoration of membrane lipid composition very similar to those of wild-type mice after the treatment of mdx mice with molsidomine, a NO donor. This was associated with the observation by immunohistology of an increase of the regeneration process in the mice.
Collapse
Affiliation(s)
- Farida Benabdellah
- Institut de Chimie des Substances Naturelles, CNRS, UPR 2301, Av. de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | | | | | | | | |
Collapse
|
32
|
Nitric oxide enhances osteoclastogenesis possibly by mediating cell fusion. Nitric Oxide 2009; 21:27-36. [PMID: 19389479 DOI: 10.1016/j.niox.2009.04.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 02/17/2009] [Accepted: 04/11/2009] [Indexed: 12/20/2022]
Abstract
Osteoclasts are multinucleated bone resorbing cells which form by fusion of pre-osteoclasts. Here, we investigate how nitric oxide (NO) affects osteoclastogenesis. Time lapse photomicrography, using the fluorescent NO indicator dye, 4,5-diaminofluorescein diacetate, revealed an intense NO signal in pre-osteoclasts preceding cell fusion. Osteoclastogenesis in RAW264.7 cells increased when exposed to the NO synthase inhibitor, L-NMMA (0.25 microM), for the initial 48 h. In contrast, pre-osteoclast fusion decreased when RAW264.7 cells were exposed to L-NMMA from 48 to 96 h. Both NO synthase inhibitors, L-NMMA and L-NAME, decreased osteoclast formation during this time period. The inhibitory effect of L-NMMA on osteoclast formation was abolished with increasing concentrations (25-200 ng/ml) of sRANKL suggesting signaling cross talk. NO donors increased osteoclast formation in a dose-dependent manner, with greatest stimulation at 15 microM NOC-12 (2.3 fold) and 5 microM NOC-18 (2.4 fold). Measuring nitrite (NO end product) daily from culture media of RAW264.7 cells undergoing osteoclastogenesis revealed that an increase in NO production coincided with the fusion of pre-osteoclasts (day 4). Inhibiting fusion by plating cells on polystyrene dishes pre-coated with poly-(L-lysine) decreased both osteoclast formation and NO production. To address if NO mediates fusion through the actin cytoskeleton, actin free barbed ends were measured. 0.25 microM L-NMMA decreased, while 15 microM NOC-12 and 5 microM NOC-18 increased actin free barbed ends. We hypothesize that while NO initially negatively regulates pre-osteoclast differentiation; it later facilitates the fusion of mononuclear pre-osteoclasts, possibly by up regulating actin remodeling.
Collapse
|
33
|
Teichert AM, Scott JA, Robb GB, Zhou YQ, Zhu SN, Lem M, Keightley A, Steer BM, Schuh AC, Adamson SL, Cybulsky MI, Marsden PA. Endothelial nitric oxide synthase gene expression during murine embryogenesis: commencement of expression in the embryo occurs with the establishment of a unidirectional circulatory system. Circ Res 2008; 103:24-33. [PMID: 18556578 DOI: 10.1161/circresaha.107.168567] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To elucidate the role of endothelial NO synthase (eNOS)-derived NO during mammalian embryogenesis, we assessed the expression of the eNOS gene during development. Using transgenic eNOS promoter/reporter mice (with beta-galactosidase and green fluorescent protein reporters), in situ cRNA hybridization, and immunohistochemistry to assess transcription, steady-state mRNA levels, and protein expression, respectively, we noted that eNOS expression in the developing cardiovascular system was highly restricted to endothelial cells of medium- and large-sized arteries and the endocardium. The onset of transcription of the native eNOS gene and reporters coincided with the establishment of robust, unidirectional blood flow at embryonic day 9.5, as assessed by Doppler ultrasound biomicroscopy. Interestingly, reporter transgene expression and native eNOS mRNA were also observed in discrete regions of the developing skeletal musculature and the apical ectodermal ridge of developing limbs, suggesting a role for eNOS-derived NO in limb development. In vitro studies of promoter/reporter constructs indicated that similar eNOS promoter regions operate in both embryonic skeletal muscle and vascular endothelial cells. In summary, transcriptional activity of the eNOS gene in the murine circulatory system occurred following the establishment of embryonic blood flow. Thus, the eNOS gene is a late-onset gene in endothelial ontogeny.
Collapse
Affiliation(s)
- Anouk-Martine Teichert
- Renal Division and Department of Medicine, St. Michael's Hospital, Department of Medicine, University of Toronto, Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The fusion of postmitotic mononucleated myoblasts to form syncytial myofibers is a critical step in the formation of skeletal muscle. Myoblast fusion occurs both during development and throughout adulthood, as skeletal muscle growth and regeneration require the accumulation of additional nuclei within myofibers. Myoblasts must undergo a complex series of molecular and morphological changes prior to fusing with one another. Although many molecules regulating myoblast fusion have been identified, the precise mechanism by which these molecules act in concert to control fusion remains to be elucidated. A comprehensive understanding of how myo-blast fusion is controlled may contribute to the treatment of various disorders associated with loss of muscle mass. In this chapter, we examine progress made toward elucidating the cellular and molecular pathways involved in mammalian myoblast fusion. Special emphasis is placed on the molecules that regulate myofiber formation without discernibly affecting biochemical differentiation.
Collapse
Affiliation(s)
- Katie M Jansen
- Graduate Program in Biochemistry, Cell and Developmental Biology, Department of Pharmacology, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
35
|
Tidball JG, Wehling-Henricks M. The role of free radicals in the pathophysiology of muscular dystrophy. J Appl Physiol (1985) 2006; 102:1677-86. [PMID: 17095633 DOI: 10.1152/japplphysiol.01145.2006] [Citation(s) in RCA: 172] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Null mutation of any one of several members of the dystrophin protein complex can cause progressive, and possibly fatal, muscle wasting. Although these muscular dystrophies arise from mutation of a single gene that is expressed primarily in muscle, the resulting pathology is complex and multisystemic, which shows a broader disruption of homeostasis than would be predicted by deletion of a single-gene product. Before the identification of the deficient proteins that underlie muscular dystrophies, such as Duchenne muscular dystrophy (DMD), oxidative stress was proposed as a major cause of the disease. Now, current knowledge supports the likelihood that interactions between the primary genetic defect and disruptions in the normal production of free radicals contribute to the pathophysiology of muscular dystrophies. In this review, we focus on the pathophysiology that results from dystrophin deficiency in humans with DMD and the mdx mouse model of DMD. Current evidence indicates three general routes through which free radical production can be disrupted in dystrophin deficiency to contribute to the ensuing pathology. First, constitutive differences in free radical production can disrupt signaling processes in muscle and other tissues and thereby exacerbate pathology. Second, tissue responses to the presence of pathology can cause a shift in free radical production that can promote cellular injury and dysfunction. Finally, behavioral differences in the affected individual can cause further changes in the production and stoichiometry of free radicals and thereby contribute to disease. Unfortunately, the complexity of the free radical-mediated processes that are perturbed in complex pathologies such as DMD will make it difficult to develop therapeutic approaches founded on systemic administration of antioxidants. More mechanistic knowledge of the specific disruptions of free radicals that underlie major features of muscular dystrophy is needed to develop more targeted and successful therapeutic approaches.
Collapse
Affiliation(s)
- James G Tidball
- Department of Physiological Science, University of California, Los Angeles, California 90095, USA.
| | | |
Collapse
|
36
|
Yoon S, Molloy MJ, Wu MP, Cowan DB, Gussoni E. C6ORF32 is upregulated during muscle cell differentiation and induces the formation of cellular filopodia. Dev Biol 2006; 301:70-81. [PMID: 17150207 PMCID: PMC1779902 DOI: 10.1016/j.ydbio.2006.11.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 10/12/2006] [Accepted: 11/01/2006] [Indexed: 02/03/2023]
Abstract
We have identified a gene by microarray analysis that is located on chromosome 6 (c6orf32), whose expression is increased during human fetal myoblast differentiation. The protein encoded by c6orf32 is expressed both in myogenic and non-myogenic primary cells isolated from 18-week old human fetal skeletal muscle. Immunofluorescent staining indicated that C6ORF32 localizes to the cellular cytoskeleton and filopodia, and often displays polarized expression within the cell. mRNA knockdown experiments in the C2C12 murine myoblast cell line demonstrated that cells lacking c6orf32 exhibit a myogenic differentiation defect, characterized by a decrease in the expression of myogenin and myosin heavy chain (MHC) proteins, whereas MyoD1 was unaltered. In contrast, overexpression of c6orf32 in C2C12 or HEK293 cells (a non-muscle cell line) promoted formation of long membrane protrusions (filopodia). Analysis of serial deletion mutants demonstrated that amino acids 55-113 of C6ORF32 are likely involved in filopodia formation. These results indicate that C6ORF32 is a novel protein likely to play multiple functions, including promoting myogenic cell differentiation, cytoskeletal rearrangement and filopodia formation.
Collapse
Affiliation(s)
- Soonsang Yoon
- Division of Genetics and Program in Genomics, Children’s Hospital Boston
| | - Michael J. Molloy
- Division of Genetics and Program in Genomics, Children’s Hospital Boston
| | - Melissa P. Wu
- Division of Genetics and Program in Genomics, Children’s Hospital Boston
| | | | - Emanuela Gussoni
- Division of Genetics and Program in Genomics, Children’s Hospital Boston
- *Corresponding author mailing address: Children’s Hospital Boston, Enders 554 320 Longwood Ave Boston, MA 02115 Telephone (617) 919-2152 FAX (617) 730-0253 e-mail:
| |
Collapse
|
37
|
Long JHD, Lira VA, Soltow QA, Betters JL, Sellman JE, Criswell DS. Arginine supplementation induces myoblast fusion via augmentation of nitric oxide production. J Muscle Res Cell Motil 2006; 27:577-84. [PMID: 17051348 DOI: 10.1007/s10974-006-9078-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Accepted: 06/23/2006] [Indexed: 11/30/2022]
Abstract
The semi-essential amino acid, L-arginine (L-Arg), is the substrate for endogenous synthesis of nitric oxide, a molecule that is involved in myoblast proliferation and fusion. Since L-Arg supply may limit nitric oxide synthase (NOS) activity in endothelial cells, we examined L-Arg supplementation in differentiating mouse myoblasts and tested the hypothesis that L-Arg exerts direct effects on myoblast fusion via augmentation of endogenous nitric oxide production. C(2)C(12) myoblasts in differentiation media received one of the following treatments for 120 h: 1 mM L-Arg, 0.1 mM N-nitro-L-arginine methyl ester (L-NAME), L-Arg + L-NAME, 10 mM L-Lysine, or no supplement (Control). Cultures were fixed and stained with hematoxylin and eosin for microphotometric image analysis of myotube density, nuclear density, and fusion index (% of total nuclei in myotubes). Endogenous production of nitric oxide during the treatment period peaked between 24 and 48 h. L-Arg amplified nitric oxide production between 0 and 24 h and increased myotube density, total nuclei number, and nuclear fusion index. These L-Arg effects were prevented by the NOS inhibitor, L-NAME. Further, L-Lysine, a competitive inhibitor of L-Arg uptake, repressed nitric oxide production and reduced myotube density and fusion index. In summary, L-Arg augments myotube formation and increases nitric oxide production in a process limited by cellular L-Arg uptake.
Collapse
Affiliation(s)
- Jodi H D Long
- Center for Exercise Science, Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | | | | | | | | | | |
Collapse
|
38
|
Dudley RWR, Danialou G, Govindaraju K, Lands L, Eidelman DE, Petrof BJ. Sarcolemmal damage in dystrophin deficiency is modulated by synergistic interactions between mechanical and oxidative/nitrosative stresses. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1276-87; quiz 1404-5. [PMID: 16565501 PMCID: PMC1606574 DOI: 10.2353/ajpath.2006.050683] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dystrophin deficiency is the cause of Duchenne muscular dystrophy, but the precise physiological basis for muscle necrosis remains unclear. To determine whether dystrophin-deficient muscles are abnormally susceptible to oxidative and nitric oxide (NO)-driven tissue stress, a hindlimb ischemia/reperfusion (I/R) model was used. Dystrophic mdx mice exhibited abnormally high levels of lipid peroxidation and protein nitration, which were preceded by exaggerated NO production during ischemia. Visualization of NO with the fluorescent probe 4,5-diaminofluorescein diacetate suggested that excess NO production during ischemia occurred within a subset of mdx fibers. In mdx muscles only, prior exposure to I/R dramatically increased the level of sarcolemmal damage resulting from stretch-mediated mechanical stress, indicating greatly exacerbated hyperfragility of the dystrophic fiber membrane. Treatment with NO synthase inhibitors (l-N(G)-nitroarginine methyl ester hydrochloride or 7-nitroindazol) effectively blocked the synergistic interaction between I/R and mechanical stress-mediated sarcolemmal damage under these conditions. Taken together, our findings provide direct ex-perimental evidence that several prevailing hy-potheses regarding the cause of muscle fiber damage in dystrophin-deficient muscle can be integrated into a common pathophysiological framework involving interactions between oxidative stress, ab-normal NO regulation, and hyperfragility of the sarcolemma.
Collapse
Affiliation(s)
- Roy W R Dudley
- Royal Victoria Hospital, Room L411, 687 Pine Ave. West, Montreal, Quebec H3A 1A1, Canada
| | | | | | | | | | | |
Collapse
|
39
|
Soltow QA, Betters JL, Sellman JE, Lira VA, Long JHD, Criswell DS. Ibuprofen Inhibits Skeletal Muscle Hypertrophy in Rats. Med Sci Sports Exerc 2006; 38:840-6. [PMID: 16672835 DOI: 10.1249/01.mss.0000218142.98704.66] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE We sought to determine whether cyclooxygenase (COX) activity is necessary for overload-induced growth of adult rat skeletal muscle, and whether nitric oxide synthase (NOS) activity is involved in upregulation of COX messenger RNA (mRNA) expression in skeletal muscle. METHODS Unilateral surgical removal of the gastrocnemius and soleus was performed on the right hindlimb of 16 female Sprague-Dawley rats (approximately 230 g) to induce chronic overload (OL) of the plantaris for 14 d, with sham surgeries performed on the contralateral leg as a normally loaded (NL) control. Half of the rats were treated with the nonspecific COX inhibitor, ibuprofen (0.2 mg.mL(-1) in drinking water; approximately 20 mg.kg(-1).d(-1)). In a second experiment, the plantaris was unilaterally overloaded for 5 or 14 d in male rats (approximately 350 g; N = 16 rats per time point) and half of the animals were treated with the NOS inhibitor, L-NAME (0.75 mg.mL(-1) in drinking water; approximately 90 mg.kg(-1).d(-1)). RESULTS Ibuprofen treatment inhibited plantaris hypertrophy by approximately 50% (P < 0.05) following 14 d of OL, as did L-NAME treatment (P < 0.05). COX-1 and COX-2 mRNA did not differ between any groups at 5 d. At 14 d, however, L-NAME caused a 30-fold increase in plantaris COX-1 mRNA expression independent of loading condition. Additionally, OL induced a 20-fold increase in COX-2 mRNA expression compared with NL (P < 0.05) at 14 d, without affecting COX-1 mRNA level. L-NAME treatment significantly inhibited OL-induced expression of COX-2 mRNA. CONCLUSION COX activity is important for in vivo muscle hypertrophy, and plantaris overload is associated with NOS activity-dependent COX-2 expression.
Collapse
Affiliation(s)
- Quinlyn A Soltow
- Center for Exercise Science, University of Florida, Gainesville, 32611, USA
| | | | | | | | | | | |
Collapse
|
40
|
Mulvey C, Harno E, Keenan A, Ohlendieck K. Expression of the skeletal muscle dystrophin-dystroglycan complex and syntrophin-nitric oxide synthase complex is severely affected in the type 2 diabetic Goto-Kakizaki rat. Eur J Cell Biol 2006; 84:867-83. [PMID: 16323284 DOI: 10.1016/j.ejcb.2005.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The inability of insulin to stimulate glucose metabolism in skeletal muscle fibres is a classic characteristic of type 2 diabetes. Using the non-obese Goto-Kakizaki rat as an established animal model of this type of diabetes, sucrose gradient centrifugation studies were performed and confirmed the abnormal subcellular location of the glucose transporter GLUT4. In addition, this analysis revealed an unexpected drastic reduction in the surface membrane marker beta-dystroglycan, a dystrophin-associated glycoprotein. Based on this finding, a comprehensive immunoblotting survey was conducted which showed a dramatic decrease in the Dp427 isoform of dystrophin and the alpha/beta-dystroglycan subcomplex, but not in laminin, sarcoglycans, dystrobrevin, and excitation-contraction-relaxation cycle elements. Thus, the backbone of the trans-sarcolemmal linkage between the extracellular matrix and the actin membrane cytoskeleton might be structurally impaired in diabetic fibres. Immunohistochemical studies revealed that the reduction in the dystrophin-dystroglycan complex does not induce obvious signs of muscle pathology, and is neither universal in all fibres, nor fibre-type specific. Most importantly, the expression of alpha-syntrophin and the syntrophin-associated neuronal isoform of nitric oxide synthase, nNOS, was demonstrated to be severely reduced in diabetic fibres. The loss of the dystrophin-dystroglycan complex and the syntrophin-nNOS complex in selected fibres suggests a weakening of the sarcolemma, abnormal signalling and probably a decreased cytoprotective mechanism in diabetes. Impaired anchoring of the cortical actin cytoskeleton via dystrophin might interfere with the proper recruitment of the glucose transporter to the surface membrane, following stimulation by insulin or muscle contraction. This may, at least partially, be responsible for the insulin resistance in diabetic skeletal muscles.
Collapse
Affiliation(s)
- Claire Mulvey
- Department of Pharmacology, Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Belfield, Ireland
| | | | | | | |
Collapse
|
41
|
Pisconti A, Brunelli S, Di Padova M, De Palma C, Deponti D, Baesso S, Sartorelli V, Cossu G, Clementi E. Follistatin induction by nitric oxide through cyclic GMP: a tightly regulated signaling pathway that controls myoblast fusion. ACTA ACUST UNITED AC 2006; 172:233-44. [PMID: 16401724 PMCID: PMC2063553 DOI: 10.1083/jcb.200507083] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The mechanism of skeletal myoblast fusion is not well understood. We show that endogenous nitric oxide (NO) generation is required for myoblast fusion both in embryonic myoblasts and in satellite cells. The effect of NO is concentration and time dependent, being evident only at the onset of differentiation, and direct on the fusion process itself. The action of NO is mediated through a tightly regulated activation of guanylate cyclase and generation of cyclic guanosine monophosphate (cGMP), so much so that deregulation of cGMP signaling leads to a fusion-induced hypertrophy of satellite-derived myotubes and embryonic muscles, and to the acquisition of fusion competence by myogenic precursors in the presomitic mesoderm. NO and cGMP induce expression of follistatin, and this secreted protein mediates their action in myogenesis. These results establish a hitherto unappreciated role of NO and cGMP in regulating myoblast fusion and elucidate their mechanism of action, providing a direct link with follistatin, which is a key player in myogenesis.
Collapse
|
42
|
Püttmann B, Gerlach EM, Krüger M, Blottner D. Neuromuscular contacts induce nitric oxide signals in skeletal myotubes in vitro. Neurosignals 2005; 14:85-95. [PMID: 16088222 DOI: 10.1159/000086290] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2004] [Accepted: 12/20/2004] [Indexed: 12/16/2022] Open
Abstract
It has previously been shown that skeletal myotubes express nitric oxide synthase (NOS) and produce and release NO signals. NOS is also part of agrin-induced acetylcholine receptor aggregations on myotubes. As nerve-muscle interactions underlie reciprocal signaling mechanisms, we hypothesized that NO signals in target myotubes may be induced by neuromuscular contacts in development. Chimeric neuron-myotube co-cultures were prepared using p75-selected spinal cord neurons from embryonic chicken. Confocal imaging revealed robust 1,2-diaminoanthraquinone red fluorescence indicative of de novo formation of NO only in those myotubes which were contacted by neurites, also verified by pre- and postsynaptic marker costaining (anti-synaptotagmin and alpha-bungarotoxin). Neither soluble agrin nor sensory dorsal root ganglionic neurons showed comparable effects in this model. We concluded that in target skeletal muscle cells the NOS/NO system is controlled by motoneuron contacts by as yet incompletely understood signaling mechanisms. Endogenous NO signaling in myotubes may be essential during synapse formation and plasticity of the neuromuscular system.
Collapse
Affiliation(s)
- Britta Püttmann
- Neurobiology and Neuromuscular Working Group, Institute of Anatomy, Campus Benjamin Franklin, Charité-University Medicine, Berlin, Germany
| | | | | | | |
Collapse
|
43
|
Chen SS, Lin CH, Chen TJ. Lead-induced attenuation in the aggregation of acetylcholine receptors during the neuromuscular junction formation. Toxicol Lett 2005; 159:89-99. [PMID: 15916872 DOI: 10.1016/j.toxlet.2005.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2005] [Revised: 04/27/2005] [Accepted: 04/29/2005] [Indexed: 11/18/2022]
Abstract
Lead (Pb2+) toxicity is more common in children and is associated with cognitive deficits, which may reflect lead-induced changes in central synaptic development and function. Aside from neurotoxicity, lead exposure may also impact mature neuromuscular junction (NMJ) and cause muscle weakness. NMJ is known as a peripheral cholinergic synapse and its signaling cascades responsible for development are similar to those for the central synapses. However, the effect of lead exposure on the formation of NMJ in mammals is unclear. In the present study, a NG108-15/C2C12 coculture model was used to measure the acetylcholine receptor (AChR) aggregates formed on the myotubes which was an early hallmark for the NMJ formation. AChR aggregates were identified by alpha-bungarotoxin under fluorescent microscope. Single dose of lead acetate with final concentrations at 10(-3), 10(-1), or 10 microM was applied to dishes at the beginning of coculturing. Following 3-day exposure, although NG108-15 cells could extend long neurites to nearby myotubes, obvious dose-dependent attenuation in AChR aggregation was shown. The averaged area of an AChR aggregate, the averaged number of AChR aggregates per myotube, and the total area of AChR aggregates per myotube were all significantly decreased. In addition, the distribution percentages of various sizes of AChR aggregates showed that almost half of the AChR aggregates were formed with a size of 2-5 microm2 regardless of lead exposure. After treating 10 microM of lead acetate, significantly more AChR aggregates ranged from 2 to 20 microm2 were formed and significantly less AChR aggregates larger than 20 microm2 were formed. These results indicated that lead exposure reduced the extent of AChR aggregation concerning both the size and number of AChR aggregates and large AChR aggregates could hardly be formed after acute high-level lead exposure. No significant change was found in the total amount of AChRs on the myotubes after lead exposure, which indicated that the attenuation of AChR aggregation was not caused by reducing the synthesis of AChRs but by remaining dispersed pattern of AChRs on the myotubes. These data suggest that lead exposure exerts detrimental effects on the formation of NMJ.
Collapse
Affiliation(s)
- Shun-Sheng Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, Kaohsiung County, Taiwan
| | | | | |
Collapse
|
44
|
Hirasaka K, Nikawa T, Yuge L, Ishihara I, Higashibata A, Ishioka N, Okubo A, Miyashita T, Suzue N, Ogawa T, Oarada M, Kishi K. Clinorotation prevents differentiation of rat myoblastic L6 cells in association with reduced NF-kappa B signaling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1743:130-40. [PMID: 15777848 DOI: 10.1016/j.bbamcr.2004.09.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2004] [Revised: 09/06/2004] [Accepted: 09/08/2004] [Indexed: 11/30/2022]
Abstract
In this study, we examined effects of the three-dimensional (3D)-clinorotation, a simulated-model of microgravity, on proliferation/differentiation of rat myoblastic L6 cells. Differentiation of L6 cells into myotubes was significantly disturbed in the 3D-clinorotation culture system, although the 3D-clinorotation had no effect on the proliferation. The 3D-clinorotation also suppressed the expression of myogenesis marker proteins, such as myogenin and myosin heavy chain (MHC), at the mRNA level. In association with this reduced differentiation, we found that the 3D-clinorotation prevented accumulation of ubiquitinated proteins, compared with non-rotation control cells. Based on these findings, we focused on the ubiquitin-dependent degradation of I kappa B, a myogenesis inhibitory protein, to clarify the mechanism of this impaired differentiation. A decline in the amount of I kappa B protein in L6 cells was significantly prevented by the rotation, while the amount of the protein in the non-rotated cells decreased along with the differentiation. Furthermore, the 3D-clinorotation reduced the NF-kappaB-binding activity in L6 cells and prevented the ubiquitination of I kappa B proteins in the I kappa B- and ubiquitin-expressing Cos7 cells. Other myogenic regulatory factors, such as deubiquitinases, cyclin E and oxygen, were not associated with the differentiation impaired by the clinorotation. Our present results suggest that simulated microgravity such as the 3D-clinorotation may disturb skeletal muscle cell differentiation, at least in part, by inhibiting the NF-kappa B pathway.
Collapse
Affiliation(s)
- Katsuya Hirasaka
- Department of Nutrition, The University of Tokushima School of Medicine, Tokushima, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ségalat L, Grisoni K, Archer J, Vargas C, Bertrand A, Anderson JE. CAPON expression in skeletal muscle is regulated by position, repair, NOS activity, and dystrophy. Exp Cell Res 2005; 302:170-9. [PMID: 15561099 DOI: 10.1016/j.yexcr.2004.09.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2004] [Revised: 09/04/2004] [Indexed: 11/16/2022]
Abstract
In skeletal muscle, the localization of nNOS is destabilized in the absence of dystrophin, which impacts muscle function and satellite cell activation. In neurons, the adaptor protein, carboxy-terminal PDZ ligand of nNOS (CAPON), regulates the distribution of neuronal nitric oxide synthase (nNOS), which produces the key signaling molecule nitric oxide (NO). While a CAPON-like gene is known to compensate functionally for a dystrophic phenotype in muscle of Caenorhabditis elegans, CAPON expression has not been reported for mammalian muscle. Here, CAPON expression was identified in mouse muscle using Northern and Western blotting and in situ hybridization in combination with immunostaining for laminin. CAPON RNA was expressed in developing normal and dystrophic muscles near fiber junctions with tendons, and levels increased from 1 to 3 weeks. In regenerating normal muscle and also in dystrophic muscles in the mdx mouse, CAPON transcripts were prominent in satellite cells and new myotubes. Expression of CAPON RNA increased in diaphragm muscle of normal and mdx mice after treatment with L-arginine, the NOS substrate. Both CAPON and utrophin protein levels increased in dystrophic quadriceps muscle after treatment with the steroid deflazacort plus L-arginine, known to reduce the dystrophic phenotype. The identification of CAPON transcripts and protein in mammalian muscle and responses to L-arginine suggest CAPON may have a functional role in stabilizing neuronal NOS in skeletal muscle in the cytoskeletal complex associated with dystrophin/utrophin, with possible applications to therapy for human muscular dystrophy.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Arginine/pharmacology
- Blotting, Western
- Diaphragm/metabolism
- Dystrophin/deficiency
- Immunohistochemistry
- Immunosuppressive Agents/pharmacology
- In Situ Hybridization
- Laminin/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophies/metabolism
- Nitric Oxide Synthase/antagonists & inhibitors
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase/metabolism
- Pregnenediones/pharmacology
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Satellite Cells, Skeletal Muscle/drug effects
- Time Factors
- Utrophin/metabolism
Collapse
Affiliation(s)
- Laurent Ségalat
- CNRS-UPR5534, Université Lyon-1 and Institut Cochin, INSERM U567, 75014 Paris, France
| | | | | | | | | | | |
Collapse
|
46
|
Lee KH, Park JY, Kim K. NMDA receptor-mediated calcium influx plays an essential role in myoblast fusion. FEBS Lett 2004; 578:47-52. [PMID: 15581614 DOI: 10.1016/j.febslet.2004.10.076] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2004] [Revised: 10/21/2004] [Accepted: 10/22/2004] [Indexed: 11/25/2022]
Abstract
Ca2+ influx is known to be prerequisite for myoblast fusion during skeletal muscle differentiation. Here, we show that the N-methyl-D-aspartate (NMDA) receptor is involved in the Ca2+ influx of C2C12 myoblasts. NMDA receptor (NR) 1 and NR2D were expressed in the myoblasts during muscle differentiation. Using Ca2+ imaging analysis, Ca2+ influx through NRs was directly measured at a single-cell level. l-Glutamate increased myoblast fusion as well as intracellular Ca2+ levels, and both effects were completely blocked by MK801, a selective antagonist of NRs. Furthermore, treatment with the Ca2+ ionophore A23187 recovered MK801-mediated inhibition of myoblast fusion. These results suggest that the NRs may play an important role in myoblast fusion by mediating Ca2+ influx.
Collapse
Affiliation(s)
- Kun Ho Lee
- School of Biological Sciences, Seoul National University, Seoul, 151-742, Republic of Korea
| | | | | |
Collapse
|
47
|
Tidball JG, Wehling-Henricks M. Expression of a NOS transgene in dystrophin-deficient muscle reduces muscle membrane damage without increasing the expression of membrane-associated cytoskeletal proteins. Mol Genet Metab 2004; 82:312-20. [PMID: 15308129 DOI: 10.1016/j.ymgme.2004.06.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2004] [Revised: 06/09/2004] [Accepted: 06/10/2004] [Indexed: 10/26/2022]
Abstract
Muscular dystrophy that is caused by mutation of the membrane-associated, cytoskeletal protein called dystrophin, is accompanied by loss of a dystrophin-associated protein complex (DPC) that includes neuronal nitric oxide synthase (nNOS). Previous work showed that expression of a nNOS transgene in the dystrophin-deficient, mdx mouse greatly reduces muscle membrane damage. In this investigation, we test whether expression of a nNOS transgene in wild-type or mdx muscle increases expression of DPC proteins, or functionally related proteins in the integrin complex that are upregulated in dystrophin-deficiency, or affects expression of the dystrophin homolog, utrophin. Many members of the DPC are enriched in Western blots of cell membranes isolated from NOS transgenic muscle, compared to wild-type. Similarly, alpha7-integrin and the associated cytoskeletal proteins talin and vinculin are increased in NOS transgenic, non-dystrophic muscle. However, utrophin expression is unaffected by elevated NOS expression in healthy muscle. A similar trend in mRNA levels for these proteins was observed by expression profiling. Analysis of membrane preparations from mdx mice and NOS transgenic mdx mice shows that expression of the NOS transgene causes significant reductions in utrophin, talin, and vinculin. Expression profiling of mRNA from mdx and NOS transgenic mdx muscles also shows reduced expression of talin. Immunohistochemistry of mdx and NOS transgenic mdx muscle indicates that reduction in utrophin in NOS transgenic mdx muscle results from a decrease in regenerative fibers that express high levels of utrophin. Together, these findings indicate that the NOS transgene does not reduce dystrophinopathy by increasing the expression of compensatory, structural proteins.
Collapse
Affiliation(s)
- James G Tidball
- Department of Physiological Science, University of California, Los Angeles, CA 90095, USA.
| | | |
Collapse
|
48
|
Zhang JS, Kraus WE, Truskey GA. Stretch-induced nitric oxide modulates mechanical properties of skeletal muscle cells. Am J Physiol Cell Physiol 2004; 287:C292-9. [PMID: 15044149 DOI: 10.1152/ajpcell.00018.2004] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this study, we examined the hypothesis that stretch-induced (nitric oxide) NO modulates the mechanical properties of skeletal muscles by increasing accumulation of protein levels of talin and vinculin and by inhibiting calpain-induced proteolysis, thereby stabilizing the focal contacts and the cytoskeleton. Differentiating C2C12myotubes were subjected to a single 10% step stretch for 0–4 days. The apparent elastic modulus of the cells, Eapp, was subsequently determined by atomic force microscopy. Static stretch led to significant increases ( P < 0.01) in Eappbeginning at 2 days. These increases were correlated with increases in NO activity and neuronal NO synthase (nNOS) protein expression. Expression of talin was upregulated throughout, whereas expression of vinculin was significantly increased only on days 3 and 4. Addition of the NO donor l-arginine onto stretched cells further enhanced Eapp, NOS activity, and nNOS expression, whereas the presence of the NO inhibitor Nω-nitro-l-arginine methyl ester (l-NAME) reversed the effects of mechanical stimulation and of l-arginine. Overall, viscous dissipation, as determined by the value of hysteresis, was not significantly altered. For assessment of the role of vinculin and talin stability, cells treated with l-NAME showed a significant decrease in Eapp, whereas addition of a calpain inhibitor abolished the effect. Thus our results show that NO inhibition of calpain-initiated cleavage of cytoskeleton proteins was correlated with the changes in Eapp. Together, our data suggest that NO modulates the mechanical behavior of skeletal muscle cells through the combined action of increased talin and vinculin levels and a decrease in calpain-mediated talin proteolysis.
Collapse
Affiliation(s)
- Jingying Sarah Zhang
- Dept. of Biomedical Engineering, Box 90281, Duke University, Durham, NC 27708-0281, USA
| | | | | |
Collapse
|
49
|
Kasai T, Abeyama K, Hashiguchi T, Fukunaga H, Osame M, Maruyama I. Decreased total nitric oxide production in patients with duchenne muscular dystrophy. J Biomed Sci 2004; 11:534-7. [PMID: 15153790 DOI: 10.1007/bf02256104] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2004] [Accepted: 02/24/2004] [Indexed: 10/25/2022] Open
Abstract
Plasma nitric oxide (NO) levels in Duchenne muscular dystrophy (DMD) patients were significantly lower than those observed in both healthy controls and in patients with other neuromuscular disorders. The correlation between NO level and ejection fraction was significant (r = -0.384, p = 0.0391) in the DMD group. Disruption of NO systems may contribute to the development of muscular dystrophy and have implications for therapeutic strategies.
Collapse
Affiliation(s)
- Takefumi Kasai
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.
| | | | | | | | | | | |
Collapse
|
50
|
Rudnick J, Püttmann B, Tesch PA, Alkner B, Schoser BGH, Salanova M, Kirsch K, Gunga HC, Schiffl G, Lück G, Blottner D. Differential expression of nitric oxide synthases (NOS 1-3) in human skeletal muscle following exercise countermeasure during 12 weeks of bed rest. FASEB J 2004; 18:1228-30. [PMID: 15180967 DOI: 10.1096/fj.03-0792fje] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Adaptive changes of major body systems in astronauts during spaceflight can be simulated by strict anti-orthostatic head-down tilt (HDT) bed rest (BR), a ground-based microgravity (microG) model that provides a meaningful opportunity to study atrophy mechanisms and possible countermeasures under controlled experimental conditions. As nitric oxide (NO) signaling is linked to muscle activity, we investigated altered expression of the three major isoforms of nitric oxide synthase (NOS 1-3) at cellular compartments during prolonged HDT BR without (control group) and with resistance exercise interventions (exercise group) using a flywheel ergometer (FWE). Atrophy detected in mixed (fast-slow) m. vastus lateralis (VL) and slow-type m. soleus (SOL) myofiber Types I and II (minus 35-40% of myofiber cross-sectional area) was prevented by FWE training. Concomitant to muscle atrophy, reduced NOS 1 protein and immunostaining was found in VL not in SOL biopsies. In trained VL, NOS 1 protein and immunostaining at myofibers II were significantly increased at the end of BR. Exercise altered NOS 2/caveolin 3 co-immunostaining patterns of subsarcolemmal focal accumulations in VL or SOL myofibers, which suggests reorganization of sarcolemmal microdomains. In trained VL, increased capillary-to-fiber (C/F) ratio and NOS 3 protein content were documented. Activity-linked NO signaling may be widespread in skeletal muscle cellular compartments that may be directly or indirectly impacted by adequate exercise countermeasure protocols to offset the negative effects induced by disuse, immobilization, or extended exposure to microgravity.
Collapse
Affiliation(s)
- Jana Rudnick
- Department of Anatomy, Campus Benjamin Franklin, Charité University Medicine Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|