1
|
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
|
2
|
Skeletal muscle redox signaling in rheumatoid arthritis. Clin Sci (Lond) 2021; 134:2835-2850. [PMID: 33146370 PMCID: PMC7642299 DOI: 10.1042/cs20190728] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by synovitis and the presence of serum autoantibodies. In addition, skeletal muscle weakness is a common comorbidity that contributes to inability to work and reduced quality of life. Loss in muscle mass cannot alone account for the muscle weakness induced by RA, but instead intramuscular dysfunction appears as a critical factor underlying the decreased force generating capacity for patients afflicted by arthritis. Oxidative stress and associated oxidative post-translational modifications have been shown to contribute to RA-induced muscle weakness in animal models of arthritis and patients with RA. However, it is still unclear how and which sources of reactive oxygen and nitrogen species (ROS/RNS) that are involved in the oxidative stress that drives the progression toward decreased muscle function in RA. Nevertheless, mitochondria, NADPH oxidases (NOX), nitric oxide synthases (NOS) and phospholipases (PLA) have all been associated with increased ROS/RNS production in RA-induced muscle weakness. In this review, we aim to cover potential ROS sources and underlying mechanisms of oxidative stress and loss of force production in RA. We also addressed the use of antioxidants and exercise as potential tools to counteract oxidative stress and skeletal muscle weakness.
Collapse
|
3
|
Shah N, Zhou L. Regulation of Ion Channel Function by Gas Molecules. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:139-164. [DOI: 10.1007/978-981-16-4254-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
4
|
Ganse B, Bosutti A, Drey M, Degens H. Sixty days of head-down tilt bed rest with or without artificial gravity do not affect the neuromuscular secretome. Exp Cell Res 2020; 399:112463. [PMID: 33385417 DOI: 10.1016/j.yexcr.2020.112463] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 01/13/2023]
Abstract
Artificial gravity is a potential countermeasure to attenuate effects of weightlessness during long-term spaceflight, including losses of muscle mass and function, possibly to some extent attributable to disturbed neuromuscular interaction. The 60-day AGBRESA bed-rest study was conducted with 24 participants (16 men, 8 women; 33 ± 9 years; 175 ± 9 cm; 74 ± 10 kg; 8 control group, 8 continuous (cAG) and 8 intermittent (iAG) centrifugation) to assess the impact of bed rest with or without daily 30-min continuous/intermittent centrifugation with 1G at the centre of mass. Fasting blood samples were collected before and on day 6, 20, 40 and 57 during 6° head-down tilt bed rest. Concentrations of circulating markers of muscle wasting (GDF-8/myostatin; slow skeletal muscle troponin T; prostaglandin E2), neurotrophic factors (BDNF; GDNF) and C-terminal Agrin Fragment (CAF) were determined by ELISAs. Creatine kinase activity was assessed by colorimetric enzyme assay. Repeated-measures ANOVAs were conducted with TIME as within-subject, and INTERVENTION and SEX as between-subject factors. The analyses revealed no significant effect of bed rest or sex on any of the parameters. Continuous or intermittent artificial gravity is a safe intervention that does not have a negative impact of the neuromuscular secretome.
Collapse
Affiliation(s)
- Bergita Ganse
- Manchester Metropolitan University, Research Centre for Musculoskeletal Science & Sports Medicine, Faculty of Science and Engineering, John Dalton Building, Manchester, United Kingdom.
| | | | - Michael Drey
- Department of Medicine IV, Geriatrics, University Hospital of LMU Munich, Munich, Germany
| | - Hans Degens
- Manchester Metropolitan University, Research Centre for Musculoskeletal Science & Sports Medicine, Faculty of Science and Engineering, John Dalton Building, Manchester, United Kingdom; Lithuanian Sports University, Institute of Sport Science and Innovations, Kaunas, Lithuania
| |
Collapse
|
5
|
Zullo A, Frisso G, Carsana A. Influence of physical activity on structure and function of the RyR1 calcium channel: a systematic review. GAZZETTA MEDICA ITALIANA ARCHIVIO PER LE SCIENZE MEDICHE 2020. [DOI: 10.23736/s0393-3660.19.04238-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
6
|
Tobias IS, Galpin AJ. Moving human muscle physiology research forward: an evaluation of fiber type-specific protein research methodologies. Am J Physiol Cell Physiol 2020; 319:C858-C876. [DOI: 10.1152/ajpcell.00107.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Human skeletal muscle is a heterogeneous tissue composed of multiple fiber types that express unique contractile and metabolic properties. While analysis of mixed fiber samples predominates and holds value, increasing attention has been directed toward studying proteins segregated by fiber type, a methodological distinction termed “fiber type-specific.” Fiber type-specific protein studies have the advantage of uncovering key molecular effects that are often missed in mixed fiber homogenate studies but also require greater time and resource-intensive methods, particularly when applied to human muscle. This review summarizes and compares current methods used for fiber type-specific protein analysis, highlighting their advantages and disadvantages for human muscle studies, in addition to recent advances in these techniques. These methods can be grouped into three categories based on the initial processing of the tissue: 1) muscle-specific fiber homogenates, 2) cross sections of fiber bundles, and 3) isolated single fibers, with various subtechniques for performing fiber type identification and protein quantification. The relative implementation for each unique methodological approach is analyzed from 83 fiber type-specific studies of proteins in live human muscle found in the literature to date. These studies have investigated several proteins involved in a wide range of cellular functions that are important to muscle tissue. The second half of this review summarizes key findings from this ensemble of fiber type-specific human protein studies. We highlight examples of where this analytical approach has helped to improve understanding of important physiological topics such as insulin sensitivity, muscle hypertrophy, muscle fatigue, and adaptation to different exercise programs.
Collapse
Affiliation(s)
- Irene S. Tobias
- Biochemistry and Molecular Exercise Physiology Laboratory, Center for Sport Performance, California State University, Fullerton, California
| | - Andrew J. Galpin
- Biochemistry and Molecular Exercise Physiology Laboratory, Center for Sport Performance, California State University, Fullerton, California
| |
Collapse
|
7
|
Pagnotti GM, Styner M, Uzer G, Patel VS, Wright LE, Ness KK, Guise TA, Rubin J, Rubin CT. Combating osteoporosis and obesity with exercise: leveraging cell mechanosensitivity. Nat Rev Endocrinol 2019; 15:339-355. [PMID: 30814687 PMCID: PMC6520125 DOI: 10.1038/s41574-019-0170-1] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Osteoporosis, a condition of skeletal decline that undermines quality of life, is treated with pharmacological interventions that are associated with poor adherence and adverse effects. Complicating efforts to improve clinical outcomes, the incidence of obesity is increasing, predisposing the population to a range of musculoskeletal complications and metabolic disorders. Pharmacological management of obesity has yet to deliver notable reductions in weight and debilitating complications are rarely avoided. By contrast, exercise shows promise as a non-invasive and non-pharmacological method of regulating both osteoporosis and obesity. The principal components of exercise - mechanical signals - promote bone and muscle anabolism while limiting formation and expansion of fat mass. Mechanical regulation of bone and marrow fat might be achieved by regulating functions of differentiated cells in the skeletal tissue while biasing lineage selection of their common progenitors - mesenchymal stem cells. An inverse relationship between adipocyte versus osteoblast fate selection from stem cells is implicated in clinical conditions such as childhood obesity and increased marrow adiposity in type 2 diabetes mellitus, as well as contributing to skeletal frailty. Understanding how exercise-induced mechanical signals can be used to improve bone quality while decreasing fat mass and metabolic dysfunction should lead to new strategies to treat chronic diseases such as osteoporosis and obesity.
Collapse
Affiliation(s)
- Gabriel M Pagnotti
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Maya Styner
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Gunes Uzer
- College of Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Vihitaben S Patel
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Laura E Wright
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Theresa A Guise
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Janet Rubin
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Clinton T Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
8
|
Balke JE, Zhang L, Percival JM. Neuronal nitric oxide synthase (nNOS) splice variant function: Insights into nitric oxide signaling from skeletal muscle. Nitric Oxide 2018; 82:35-47. [PMID: 30503614 DOI: 10.1016/j.niox.2018.11.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023]
Abstract
Defects in neuronal nitric oxide synthase (nNOS) splice variant localization and signaling in skeletal muscle are a firmly established pathogenic characteristic of many neuromuscular diseases, including Duchenne and Becker muscular dystrophy (DMD and BMD, respectively). Therefore, substantial efforts have been made to understand and therapeutically target skeletal muscle nNOS isoform signaling. The purpose of this review is to summarize recent salient advances in understanding of the regulation, targeting, and function of nNOSμ and nNOSβ splice variants in normal and dystrophic skeletal muscle, primarily using findings from mouse models. The first focus of this review is how the differential targeting of nNOS splice variants creates spatially and functionally distinct nitric oxide (NO) signaling compartments at the sarcolemma, Golgi complex, and cytoplasm. Particular attention is given to the functions of sarcolemmal nNOSμ and limitations of current nNOS knockout models. The second major focus is to review current understanding of cGMP-mediated nNOS signaling in skeletal muscle and its emergence as a therapeutic target in DMD and BMD. Accordingly, we address the preclinical and clinical successes and setbacks with the testing of phosphodiesterase 5 inhibitors to redress nNOS signaling defects in DMD and BMD. In summary, this review of nNOS function in normal and dystrophic muscle aims to advance understanding how the messenger NO is harnessed for cellular signaling from a skeletal muscle perspective.
Collapse
Affiliation(s)
- Jordan E Balke
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine Miami, Florida, 33101, USA
| | - Ling Zhang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine Miami, Florida, 33101, USA
| | - Justin M Percival
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine Miami, Florida, 33101, USA.
| |
Collapse
|
9
|
Yamada T, Steinz MM, Kenne E, Lanner JT. Muscle Weakness in Rheumatoid Arthritis: The Role of Ca 2+ and Free Radical Signaling. EBioMedicine 2017; 23:12-19. [PMID: 28781131 PMCID: PMC5605300 DOI: 10.1016/j.ebiom.2017.07.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023] Open
Abstract
In addition to the primary symptoms arising from inflammatory processes in the joints, muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Muscle weakness not only reduces the quality of life for the affected patients, but also dramatically increases the burden on society since patients' work ability decreases. A 25–70% reduction in muscular strength has been observed in pateints with RA when compared with age-matched healthy controls. The reduction in muscle strength is often larger than what could be explained by the reduction in muscle size in patients with RA, which indicates that intracellular (intrinsic) muscle dysfunction plays an important role in the underlying mechanism of muscle weakness associated with RA. In this review, we highlight the present understanding of RA-associated muscle weakness with special focus on how enhanced Ca2 + release from the ryanodine receptor and free radicals (reactive oxygen/nitrogen species) contributes to muscle weakness, and recent developments of novel therapeutic interventions. Muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Intrinsic muscle weakness is important in the underlying mechanisms of muscle weakness associated with rheumatoid arthritis. Enhanced Ca2 + release and peroxynitrite-induced stress contributes to RA-induced muscle weakness.
Collapse
Affiliation(s)
- Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Maarten M Steinz
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
10
|
|
11
|
Gentil C, Le Guiner C, Falcone S, Hogrel JY, Peccate C, Lorain S, Benkhelifa-Ziyyat S, Guigand L, Montus M, Servais L, Voit T, Piétri-Rouxel F. Dystrophin Threshold Level Necessary for Normalization of Neuronal Nitric Oxide Synthase, Inducible Nitric Oxide Synthase, and Ryanodine Receptor-Calcium Release Channel Type 1 Nitrosylation in Golden Retriever Muscular Dystrophy Dystrophinopathy. Hum Gene Ther 2016; 27:712-26. [PMID: 27279388 DOI: 10.1089/hum.2016.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
At present, the clinically most advanced strategy to treat Duchenne muscular dystrophy (DMD) is the exon-skipping strategy. Whereas antisense oligonucleotide-based clinical trials are underway for DMD, it is essential to determine the dystrophin restoration threshold needed to ensure improvement of muscle physiology at the molecular level. A preclinical trial has been conducted in golden retriever muscular dystrophy (GRMD) dogs treated in a forelimb by locoregional delivery of rAAV8-U7snRNA to promote exon skipping on the canine dystrophin messenger. Here, we exploited rAAV8-U7snRNA-transduced GRMD muscle samples, well characterized for their percentage of dystrophin-positive fibers, with the aim of defining the threshold of dystrophin rescue necessary for normalization of the status of neuronal nitric oxide synthase mu (nNOSμ), inducible nitric oxide synthase (iNOS), and ryanodine receptor-calcium release channel type 1 (RyR1), crucial actors for efficient contractile function. Results showed that restoration of dystrophin in 40% of muscle fibers is needed to decrease abnormal cytosolic nNOSμ expression and to reduce overexpression of iNOS, these two parameters leading to a reduction in the NO level in the muscle fibers. Furthermore, the same percentage of dystrophin-positive fibers of 40% was associated with the normalization of RyR1 nitrosylation status and with stabilization of the RyR1-calstabin1 complex that is required to facilitate coupled gating. We concluded that a minimal threshold of 40% of dystrophin-positive fibers is necessary for the reinstatement of central proteins needed for proper muscle contractile function, and thus identified a rate of dystrophin expression significantly improving, at the molecular level, the dystrophic muscle physiology.
Collapse
Affiliation(s)
- Christel Gentil
- 1 Sorbonne Universités , UPMC Univ Paris 06/INSERM/CNRS/Institut de Myologie/Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, Paris, France
| | - Caroline Le Guiner
- 2 Atlantic Gene Therapies/INSERM UMR 1089 Université de Nantes , CHU de Nantes, IRT1, Nantes, France.,3 Généthon , Evry, France
| | - Sestina Falcone
- 1 Sorbonne Universités , UPMC Univ Paris 06/INSERM/CNRS/Institut de Myologie/Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, Paris, France
| | | | - Cécile Peccate
- 1 Sorbonne Universités , UPMC Univ Paris 06/INSERM/CNRS/Institut de Myologie/Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, Paris, France
| | - Stéphanie Lorain
- 1 Sorbonne Universités , UPMC Univ Paris 06/INSERM/CNRS/Institut de Myologie/Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, Paris, France
| | - Sofia Benkhelifa-Ziyyat
- 1 Sorbonne Universités , UPMC Univ Paris 06/INSERM/CNRS/Institut de Myologie/Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, Paris, France
| | - Lydie Guigand
- 5 Atlantic Gene Therapies /INRA UMR 703, ONIRIS, Nantes-Atlantic National College of Veterinary Medicine , Food Science, and Engineering, Nantes, France
| | | | - Laurent Servais
- 4 Institut de Myologie , GH Pitié-Salpêtrière, Paris, France
| | - Thomas Voit
- 1 Sorbonne Universités , UPMC Univ Paris 06/INSERM/CNRS/Institut de Myologie/Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, Paris, France
| | - France Piétri-Rouxel
- 1 Sorbonne Universités , UPMC Univ Paris 06/INSERM/CNRS/Institut de Myologie/Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, Paris, France
| |
Collapse
|
12
|
Cheng AJ, Yamada T, Rassier DE, Andersson DC, Westerblad H, Lanner JT. Reactive oxygen/nitrogen species and contractile function in skeletal muscle during fatigue and recovery. J Physiol 2016; 594:5149-60. [PMID: 26857536 DOI: 10.1113/jp270650] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/23/2015] [Indexed: 01/17/2023] Open
Abstract
The production of reactive oxygen/nitrogen species (ROS/RNS) is generally considered to increase during physical exercise. Nevertheless, direct measurements of ROS/RNS often show modest increases in ROS/RNS in muscle fibres even during intensive fatiguing stimulation, and the major source(s) of ROS/RNS during exercise is still being debated. In rested muscle fibres, mild and acute exposure to exogenous ROS/RNS generally increases myofibrillar submaximal force, whereas stronger or prolonged exposure has the opposite effect. Endogenous production of ROS/RNS seems to preferentially decrease submaximal force and positive effects of antioxidants are mainly observed during fatigue induced by submaximal contractions. Fatigued muscle fibres frequently enter a prolonged state of reduced submaximal force, which is caused by a ROS/RNS-dependent decrease in sarcoplasmic reticulum Ca(2+) release and/or myofibrillar Ca(2+) sensitivity. Increased ROS/RNS production during exercise can also be beneficial and recent human and animal studies show that antioxidant supplementation can hamper the beneficial effects of endurance training. In conclusion, increased ROS/RNS production have both beneficial and detrimental effects on skeletal muscle function and the outcome depends on a combination of factors: the type of ROS/RNS; the magnitude, duration and location of ROS/RNS production; and the defence systems, including both endogenous and exogenous antioxidants.
Collapse
Affiliation(s)
| | | | - Dilson E Rassier
- McGill University, 475 Pine Avenue West, Montreal, QC, Canada, H2W1S4
| | | | | | | |
Collapse
|
13
|
Allen DG, Whitehead NP, Froehner SC. Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiol Rev 2016; 96:253-305. [PMID: 26676145 DOI: 10.1152/physrev.00007.2015] [Citation(s) in RCA: 294] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dystrophin is a long rod-shaped protein that connects the subsarcolemmal cytoskeleton to a complex of proteins in the surface membrane (dystrophin protein complex, DPC), with further connections via laminin to other extracellular matrix proteins. Initially considered a structural complex that protected the sarcolemma from mechanical damage, the DPC is now known to serve as a scaffold for numerous signaling proteins. Absence or reduced expression of dystrophin or many of the DPC components cause the muscular dystrophies, a group of inherited diseases in which repeated bouts of muscle damage lead to atrophy and fibrosis, and eventually muscle degeneration. The normal function of dystrophin is poorly defined. In its absence a complex series of changes occur with multiple muscle proteins showing reduced or increased expression or being modified in various ways. In this review, we will consider the various proteins whose expression and function is changed in muscular dystrophies, focusing on Ca(2+)-permeable channels, nitric oxide synthase, NADPH oxidase, and caveolins. Excessive Ca(2+) entry, increased membrane permeability, disordered caveolar function, and increased levels of reactive oxygen species are early changes in the disease, and the hypotheses for these phenomena will be critically considered. The aim of the review is to define the early damage pathways in muscular dystrophy which might be appropriate targets for therapy designed to minimize the muscle degeneration and slow the progression of the disease.
Collapse
Affiliation(s)
- David G Allen
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Nicholas P Whitehead
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Stanley C Froehner
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
14
|
Vibration mechanosignals superimposed to resistive exercise result in baseline skeletal muscle transcriptome profiles following chronic disuse in bed rest. Sci Rep 2015; 5:17027. [PMID: 26596638 PMCID: PMC4657004 DOI: 10.1038/srep17027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/21/2015] [Indexed: 01/08/2023] Open
Abstract
Disuse-induced muscle atrophy is a major concern in aging, in neuromuscular diseases, post-traumatic injury and in microgravity life sciences affecting health and fitness also of crew members in spaceflight. By using a laboratory analogue to body unloading we perform for the first time global gene expression profiling joined to specific proteomic analysis to map molecular adaptations in disused (60 days of bed rest) human soleus muscle (CTR) and in response to a resistive exercise (RE) countermeasure protocol without and with superimposed vibration mechanosignals (RVE). Adopting Affymetrix GeneChip technology we identified 235 differently transcribed genes in the CTR group (end- vs. pre-bed rest). RE comprised 206 differentially expressed genes, whereas only 51 changed gene transcripts were found in RVE. Most gene transcription and proteomic changes were linked to various key metabolic pathways (glycolysis, oxidative phosphorylation, tricarboxylic acid (TCA) cycle, lipid metabolism) and to functional contractile structures. Gene expression profiling in bed rest identified a novel set of genes explicitly responsive to vibration mechanosignals in human soleus. This new finding highlights the efficacy of RVE protocol in reducing key signs of disuse maladaptation and atrophy, and to maintain a close-to-normal skeletal muscle quality outcome following chronic disuse in bed rest.
Collapse
|
15
|
Shenkman BS, Nemirovskaya TL, Lomonosova YN. No-dependent signaling pathways in unloaded skeletal muscle. Front Physiol 2015; 6:298. [PMID: 26582991 PMCID: PMC4628111 DOI: 10.3389/fphys.2015.00298] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/09/2015] [Indexed: 01/22/2023] Open
Abstract
The main focus of the current review is the nitric oxide (NO)-mediated signaling mechanism in unloaded skeletal. Review of the published data describing muscles during physical activity and inactivity demonstrates that NO is an essential trigger of signaling processes, which leads to structural and metabolic changes of the muscle fibers. The experiments with modulation of NO-synthase (NOS) activity during muscle unloading demonstrate the ability of an activated enzyme to stabilize degradation processes and prevent development of muscle atrophy. Various forms of muscle mechanical activity, i.e., plantar afferent stimulation, resistive exercise and passive chronic stretch increase the content of neural NOS (nNOS) and thus may facilitate an increase in NO production. Recent studies demonstrate that NO-synthase participates in the regulation of protein and energy metabolism in skeletal muscle by fine-tuning and stabilizing complex signaling systems which regulate protein synthesis and degradation in the fibers of inactive muscle.
Collapse
Affiliation(s)
- Boris S Shenkman
- Institute of Biomedical Problems, Russian Academy of Sciences Moscow, Russia
| | - Tatiana L Nemirovskaya
- Institute of Biomedical Problems, Russian Academy of Sciences Moscow, Russia ; Faculty of Fundamental Medicine, Lomonosov Moscow State University Moscow, Russia
| | - Yulia N Lomonosova
- Institute of Biomedical Problems, Russian Academy of Sciences Moscow, Russia
| |
Collapse
|
16
|
Salanova M, Gelfi C, Moriggi M, Vasso M, Viganò A, Minafra L, Bonifacio G, Schiffl G, Gutsmann M, Felsenberg D, Cerretelli P, Blottner D. Disuse deterioration of human skeletal muscle challenged by resistive exercise superimposed with vibration: evidence from structural and proteomic analysis. FASEB J 2014; 28:4748-63. [PMID: 25122557 DOI: 10.1096/fj.14-252825] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In the present bed rest (BR) study, 23 volunteers were randomized into 3 subgroups: 60 d BR control (Ctr); BR with resistive exercise (RE; lower-limb load); and resistive vibration exercise (RVE; RE with superimposed vibration). The aim was to analyze by confocal and electron microscopy the effects of vibration on myofibril and filament integrity in soleus (Sol) and vastus lateralis (VL) muscle; differential proteomics of contractile, cytoskeletal, and costameric proteins (TN-C, ROCK1, and FAK); and expression of PGC1α and atrophy-related master genes MuRF1 and MuRF2. RVE (but not RE) preserved myofiber size and phenotype in Sol and VL by overexpressing MYBPC1 (42%, P ≤ 0.01), WDR1 (39%, P ≤ 0.01), sarcosin (84%, P ≤ 0.01), and CKM (20%, P ≤ 0.01) and prevented myofibrillar ultrastructural damage as detectable by MuRF1 expression. In Sol, cytoskeletal and contractile proteins were normalized by RVE, and TN-C increased (59%, P ≤ 0.01); the latter also with RE (108%, P ≤ 0.01). In VL, the outcomes of both RVE (acting on sarcosin and desmin) and RE (by way of troponinT-slow and MYL2) were similar. RVE appears to be a highly efficient countermeasure protocol against muscle atrophy and ultrastructural and molecular dysregulation induced by chronic disuse.
Collapse
Affiliation(s)
- Michele Salanova
- Center of Space Medicine Berlin, Neuromuscular Group, Institute of Anatomy, and
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; Consiglio Nazionale delle Ricerche (CNR), Institute of Bioimaging and Molecular Physiology, Segrate, Italy; Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, San Donato Milanese, Italy;
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Michele Vasso
- Consiglio Nazionale delle Ricerche (CNR), Institute of Bioimaging and Molecular Physiology, Segrate, Italy; Institute of Bioimaging and Molecular Physiology, CNR-Laboratorio di Tecnologie Oncologiche (LATO), Cefalù, Italy; and
| | - Agnese Viganò
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Luigi Minafra
- Institute of Bioimaging and Molecular Physiology, CNR-Laboratorio di Tecnologie Oncologiche (LATO), Cefalù, Italy; and
| | - Gaetano Bonifacio
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Gudrun Schiffl
- Center of Space Medicine Berlin, Neuromuscular Group, Institute of Anatomy, and
| | - Martina Gutsmann
- Center of Space Medicine Berlin, Neuromuscular Group, Institute of Anatomy, and
| | - Dieter Felsenberg
- Center for Muscle and Bone Research (ZMK), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Paolo Cerretelli
- Consiglio Nazionale delle Ricerche (CNR), Institute of Bioimaging and Molecular Physiology, Segrate, Italy
| | - Dieter Blottner
- Center of Space Medicine Berlin, Neuromuscular Group, Institute of Anatomy, and
| |
Collapse
|
17
|
Vitadello M, Gherardini J, Gorza L. The stress protein/chaperone Grp94 counteracts muscle disuse atrophy by stabilizing subsarcolemmal neuronal nitric oxide synthase. Antioxid Redox Signal 2014; 20:2479-96. [PMID: 24093939 PMCID: PMC4025603 DOI: 10.1089/ars.2012.4794] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS Redox and growth-factor imbalance fosters muscle disuse atrophy. Since the endoplasmic-reticulum chaperone Grp94 is required for folding insulin-like growth factors (IGFs) and for antioxidant cytoprotection, we investigated its involvement in muscle mass loss due to inactivity. RESULTS Rat soleus muscles were transfected in vivo and analyzed after 7 days of hindlimb unloading, an experimental model of muscle disuse atrophy, or standard caging. Increased muscle protein carbonylation and decreased Grp94 protein levels (p<0.05) characterized atrophic unloaded solei. Recombinant Grp94 expression significantly reduced atrophy of transfected myofibers, compared with untransfected and empty-vector transfected ones (p<0.01), and decreased the percentage of carbonylated myofibers (p=0.001). Conversely, expression of two different N-terminal deleted Grp94 species did not attenuate myofiber atrophy. No change in myofiber trophism was detected in transfected ambulatory solei. The absence of effects on atrophic untransfected myofibers excluded a major role for IGFs folded by recombinant Grp94. Immunoprecipitation and confocal microscopy assays to investigate chaperone interaction with muscle atrophy regulators identified 160 kDa neuronal nitric oxide synthase (nNOS) as a new Grp94 partner. Unloading was demonstrated to untether nNOS from myofiber subsarcolemma; here, we show that such nNOS localization, revealed by means of NADPH-diaphorase histochemistry, appeared preserved in unloaded myofibers expressing recombinant Grp94, compared to those transfected with the empty vector or deleted Grp94 cDNA (p<0.02). INNOVATION Grp94 interacts with nNOS and prevents its untethering from sarcolemma in unloaded myofibers. CONCLUSION Maintenance of Grp94 expression is sufficient to counter unloading atrophy and oxidative stress by mechanistically stabilizing nNOS-multiprotein complex at the myofiber sarcolemma.
Collapse
|
18
|
Yamada T, Fedotovskaya O, Cheng AJ, Cornachione AS, Minozzo FC, Aulin C, Fridén C, Turesson C, Andersson DC, Glenmark B, Lundberg IE, Rassier DE, Westerblad H, Lanner JT. Nitrosative modifications of the Ca2+ release complex and actin underlie arthritis-induced muscle weakness. Ann Rheum Dis 2014; 74:1907-14. [PMID: 24854355 PMCID: PMC4602262 DOI: 10.1136/annrheumdis-2013-205007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 05/01/2014] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Skeletal muscle weakness is a prominent clinical feature in patients with rheumatoid arthritis (RA), but the underlying mechanism(s) is unknown. Here we investigate the mechanisms behind arthritis-induced skeletal muscle weakness with special focus on the role of nitrosative stress on intracellular Ca(2+) handling and specific force production. METHODS Nitric oxide synthase (NOS) expression, degree of nitrosative stress and composition of the major intracellular Ca(2+) release channel (ryanodine receptor 1, RyR1) complex were measured in muscle. Changes in cytosolic free Ca(2+) concentration ([Ca(2+)]i) and force production were assessed in single-muscle fibres and isolated myofibrils using atomic force cantilevers. RESULTS The total neuronal NOS (nNOS) levels were increased in muscles both from collagen-induced arthritis (CIA) mice and patients with RA. The nNOS associated with RyR1 was increased and accompanied by increased [Ca(2+)]i during contractions of muscles from CIA mice. A marker of peroxynitrite-derived nitrosative stress (3-nitrotyrosine, 3-NT) was increased on the RyR1 complex and on actin of muscles from CIA mice. Despite increased [Ca(2+)]i, individual CIA muscle fibres were weaker than in healthy controls, that is, force per cross-sectional area was decreased. Furthermore, force and kinetics were impaired in CIA myofibrils, hence actin and myosin showed decreased ability to interact, which could be a result of increased 3-NT content on actin. CONCLUSIONS Arthritis-induced muscle weakness is linked to nitrosative modifications of the RyR1 protein complex and actin, which are driven by increased nNOS associated with RyR1 and progressively increasing Ca(2+) activation.
Collapse
Affiliation(s)
- Takashi Yamada
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Olga Fedotovskaya
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Arthur J Cheng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anabelle S Cornachione
- Department of Kinesiology and Physical Education and Department of Physics and Physiology, McGill University, Montreal, Canada
| | - Fabio C Minozzo
- Department of Kinesiology and Physical Education and Department of Physics and Physiology, McGill University, Montreal, Canada
| | - Cecilia Aulin
- Department of Medicine, Rheumatology Unit, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Fridén
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Carl Turesson
- Department of Clinical Sciences, Section of Rheumatology, Lund University, Malmö, Sweden
| | - Daniel C Andersson
- Department of Medicine, Cardiology Unit, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Glenmark
- Department of Clinical Science and Education, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid E Lundberg
- Department of Medicine, Rheumatology Unit, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Dilson E Rassier
- Department of Kinesiology and Physical Education and Department of Physics and Physiology, McGill University, Montreal, Canada
| | - Håkan Westerblad
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
Vitadello M, Germinario E, Ravara B, Libera LD, Danieli-Betto D, Gorza L. Curcumin counteracts loss of force and atrophy of hindlimb unloaded rat soleus by hampering neuronal nitric oxide synthase untethering from sarcolemma. J Physiol 2014; 592:2637-52. [PMID: 24710058 DOI: 10.1113/jphysiol.2013.268672] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Antioxidant administration aimed to antagonize the development and progression of disuse muscle atrophy provided controversial results. Here we investigated the effects of curcumin, a vegetal polyphenol with pleiotropic biological activity, because of its ability to upregulate glucose-regulated protein 94 kDa (Grp94) expression in myogenic cells. Grp94 is a sarco-endoplasmic reticulum chaperone, the levels of which decrease significantly in unloaded muscle. Rats were injected intraperitoneally with curcumin and soleus muscle was analysed after 7 days of hindlimb unloading or standard caging. Curcumin administration increased Grp94 protein levels about twofold in muscles of ambulatory rats (P < 0.05) and antagonized its decrease in unloaded ones. Treatment countered loss of soleus mass and myofibre cross-sectional area by approximately 30% (P ≤ 0.02) and maintained a force-frequency relationship closer to ambulatory levels. Indexes of muscle protein and lipid oxidation, such as protein carbonylation, revealed by Oxyblot, and malondialdehyde, measured with HPLC, were significantly blunted in unloaded treated rats compared to untreated ones (P = 0.01). Mechanistic involvement of Grp94 was suggested by the disruption of curcumin-induced attenuation of myofibre atrophy after transfection with antisense grp94 cDNA and by the drug-positive effect on the maintenance of the subsarcolemmal localization of active neuronal nitric oxide synthase molecules, which were displaced to the sarcoplasm by unloading. The absence of additive effects after combined administration of a neuronal nitric oxide synthase inhibitor further supported curcumin interference with this pro-atrophic pathway. In conclusion, curcumin represents an effective and safe tool to upregulate Grp94 muscle levels and to maintain muscle function during unweighting.
Collapse
Affiliation(s)
| | - Elena Germinario
- Department of Biomedical Sciences, University of Padova, Padova, Italy Interuniversity Institute of Myology, Italy
| | - Barbara Ravara
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Daniela Danieli-Betto
- Department of Biomedical Sciences, University of Padova, Padova, Italy Interuniversity Institute of Myology, Italy
| | - Luisa Gorza
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
20
|
McKeehen JN, Novotny SA, Baltgalvis KA, Call JA, Nuckley DJ, Lowe DA. Adaptations of mouse skeletal muscle to low-intensity vibration training. Med Sci Sports Exerc 2014; 45:1051-9. [PMID: 23274599 DOI: 10.1249/mss.0b013e3182811947] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE We tested the hypothesis that low-intensity vibration training in mice improves contractile function of hindlimb skeletal muscles and promotes exercise-related cellular adaptations. METHODS We subjected C57BL/6J mice to 6 wk, 5 d·wk, 15 min·d of sham or low-intensity vibration (45 Hz, 1.0g) while housed in traditional cages (Sham-Active, n = 8; Vibrated-Active, n = 10) or in small cages to restrict physical activity (Sham-Restricted, n = 8; Vibrated-Restricted, n = 8). Contractile function and resistance to fatigue were tested in vivo (anterior and posterior crural muscles) and ex vivo on the soleus muscle. Tibialis anterior and soleus muscles were evaluated histologically for alterations in oxidative metabolism, capillarity, and fiber types. Epididymal fat pad and hindlimb muscle masses were measured. Two-way ANOVAs were used to determine the effects of vibration and physical inactivity. RESULTS Vibration training resulted in a 10% increase in maximal isometric torque (P = 0.038) and 16% faster maximal rate of relaxation (P = 0.030) of the anterior crural muscles. Posterior crural muscles were unaffected by vibration, except greater rates of contraction in Vibrated-Restricted mice compared with Vibrated-Active and Sham-Restricted mice (P = 0.022). Soleus muscle maximal isometric tetanic force tended to be greater (P = 0.057), and maximal relaxation was 20% faster (P = 0.005) in vibrated compared with sham mice. The restriction of physical activity induced muscle weakness but was not required for vibration to be effective in improving strength or relaxation. Vibration training did not affect muscle fatigability or any indicator of cellular adaptation investigated (P ≥ 0.431). Fat pad but not hindlimb muscle masses were affected by vibration training. CONCLUSION Vibration training in mice improved muscle contractility, specifically strength and relaxation rates, with no indication of adverse effects to muscle function or cellular adaptations.
Collapse
Affiliation(s)
- James N McKeehen
- Rehabilitation Science and Program in Physical Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
21
|
Salanova M, Schiffl G, Gutsmann M, Felsenberg D, Furlan S, Volpe P, Clarke A, Blottner D. Nitrosative stress in human skeletal muscle attenuated by exercise countermeasure after chronic disuse. Redox Biol 2013; 1:514-26. [PMID: 24251120 PMCID: PMC3830069 DOI: 10.1016/j.redox.2013.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 10/22/2013] [Accepted: 10/23/2013] [Indexed: 11/19/2022] Open
Abstract
Activity-induced nitric oxide (NO) imbalance and "nitrosative stress" are proposed mechanisms of disrupted Ca(2+) homeostasis in atrophic skeletal muscle. We thus mapped S-nitrosylated (SNO) functional muscle proteins in healthy male subjects in a long-term bed rest study (BBR2-2 Study) without and with exercise as countermeasure in order to assess (i) the negative effects of chronic muscle disuse by nitrosative stress, (ii) to test for possible attenuation by exercise countermeasure in bed rest and (iii) to identify new NO target proteins. Muscle biopsies from calf soleus and hip vastus lateralis were harvested at start (Pre) and at end (End) from a bed rest disuse control group (CTR, n=9) and two bed rest resistive exercise groups either without (RE, n=7) or with superimposed vibration stimuli (RVE, n=7). At subcellular compartments, strong anti-SNO-Cys immunofluorescence patterns in control muscle fibers after bed rest returned to baseline following vibration exercise. Total SNO-protein levels, Nrf-2 gene expression and nucleocytoplasmic shuttling were changed to varying degrees in all groups. Excess SNO-protein levels of specific calcium release/uptake proteins (SNO-RyR1, -SERCA1 and -PMCA) and of contractile myosin heavy chains seen in biopsy samples of chronically disused skeletal muscle were largely reduced by vibration exercise. We also identified NOS1 as a novel NO target in human skeletal muscle controlled by activity driven auto-nitrosylation mechanisms. Our findings suggest that aberrant levels of functional SNO-proteins represent signatures of uncontrolled nitrosative stress management in disused human skeletal muscle that can be offset by exercise as countermeasure.
Collapse
Affiliation(s)
- Michele Salanova
- Charité - Universitätsmedizin Berlin, Center for Space Medicine Berlin (ZWMB) Germany
- Charité - Universitätsmedizin Berlin, Department of Anatomy, Neuromuscular Group, Berlin, Germany
- Correspondence to: Department of Vegetative Anatomy, Charité Universitätsmedizin Berlin, Philippstrasse 12, 10115, Berlin, Germany. Tel.: +49 30 450528 354; fax: +49 30 450528 954.
| | - Gudrun Schiffl
- Charité - Universitätsmedizin Berlin, Center for Space Medicine Berlin (ZWMB) Germany
- Charité - Universitätsmedizin Berlin, Department of Anatomy, Neuromuscular Group, Berlin, Germany
| | - Martina Gutsmann
- Charité - Universitätsmedizin Berlin, Center for Space Medicine Berlin (ZWMB) Germany
- Charité - Universitätsmedizin Berlin, Department of Anatomy, Neuromuscular Group, Berlin, Germany
| | - Dieter Felsenberg
- Charité - Universitätsmedizin Berlin, Center of Muscle and Bone Research (ZMK) Berlin, Germany
| | - Sandra Furlan
- Dipartimento di Scienze Biomediche, Università di Padova, Italy; C.N.R. Institute of Neuroscience, Padova, Italy
| | - Pompeo Volpe
- Dipartimento di Scienze Biomediche, Università di Padova, Italy; C.N.R. Institute of Neuroscience, Padova, Italy
| | - Andrew Clarke
- Charité - Universitätsmedizin Berlin, Center for Space Medicine Berlin (ZWMB) Germany
| | - Dieter Blottner
- Charité - Universitätsmedizin Berlin, Center for Space Medicine Berlin (ZWMB) Germany
- Charité - Universitätsmedizin Berlin, Department of Anatomy, Neuromuscular Group, Berlin, Germany
| |
Collapse
|
22
|
Rehn TA, Munkvik M, Lunde PK, Sjaastad I, Sejersted OM. Intrinsic skeletal muscle alterations in chronic heart failure patients: a disease-specific myopathy or a result of deconditioning? Heart Fail Rev 2013; 17:421-36. [PMID: 21996779 DOI: 10.1007/s10741-011-9289-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Chronic heart failure (CHF) patients frequently experience impaired exercise tolerance due to skeletal muscle fatigue. Studies suggest that this in part is due to intrinsic alterations in skeletal muscle of CHF patients, often interpreted as a disease-specific myopathy. Knowledge about the mechanisms underlying these skeletal muscle alterations is of importance for the pathophysiological understanding of CHF, therapeutic approach and rehabilitation strategies. We here critically review the evidence for skeletal muscle alterations in CHF, the underlying mechanisms of such alterations and how skeletal muscle responds to training in this patient group. Skeletal muscle characteristics in CHF patients are very similar to what is reported in response to chronic obstructive pulmonary disease (COPD), detraining and deconditioning. Furthermore, skeletal muscle alterations observed in CHF patients are reversible by training, and skeletal muscle of CHF patients seems to be at least as trainable as that of matched controls. We argue that deconditioning is a major contributor to the skeletal muscle dysfunction in CHF patients and that further research is needed to determine whether, and to what extent, the intrinsic skeletal muscle alterations in CHF represent an integral part of the pathophysiology in this disease.
Collapse
Affiliation(s)
- T A Rehn
- Institute for Experimental Medical Research, Oslo University Hospital, Ullevaal, Oslo, Norway.
| | | | | | | | | |
Collapse
|
23
|
Intense resistance exercise induces early and transient increases in ryanodine receptor 1 phosphorylation in human skeletal muscle. PLoS One 2012; 7:e49326. [PMID: 23173055 PMCID: PMC3500289 DOI: 10.1371/journal.pone.0049326] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Accepted: 10/10/2012] [Indexed: 12/22/2022] Open
Abstract
Background While ryanodine receptor 1 (RyR1) critically contributes to skeletal muscle contraction abilities by mediating Ca2+ion oscillation between sarcoplasmatic and myofibrillar compartments, AMP-activated protein kinase (AMPK) senses contraction-induced energetic stress by phosphorylation at Thr172. Phosphorylation of RyR1 at serine2843 (pRyR1Ser2843) results in leaky RyR1 channels and impaired Ca2+homeostasis. Because acute resistance exercise exerts decreased contraction performance in skeletal muscle, preceded by high rates of Ca2+-oscillation and energetic stress, intense myofiber contractions may induce increased RyR1 and AMPK phosphorylation. However, no data are available regarding the time-course and magnitude of early RyR1 and AMPK phosphorylation in human myofibers in response to acute resistance exercise. Purpose Determine the effects and early time-course of resistance exercise on pRyR1Ser2843 and pAMPKThr172 in type I and II myofibers. Methods 7 male subjects (age 23±2 years, height: 185±7 cm, weight: 82±5 kg) performed 3 sets of 8 repetitions of maximum eccentric knee extensions. Muscle biopsies were taken at rest, 15, 30 and 60 min post exercise. pRyR1Ser2843 and pAMPKThr172 levels were determined by western blot and semi-quantitative immunohistochemistry techniques. Results While total RyR1 and total AMPK levels remained unchanged, RyR1 was significantly more abundant in type II than type I myofibers. pRyR1Ser2843 increased 15 min and peaked 30 min (p<0.01) post exercise in both myofiber types. Type I fibers showed relatively higher increases in pRyR1Ser2843 levels than type II myofibers and remained elevated up to 60 min post resistance exercise (p<0.05). pAMPKThr172 also increased 15 to 30 min post exercise (p<0.01) in type I and II myofibers and in whole skeletal muscle. Conclusion Resistance exercise induces acutely increased pRyR1Ser2843 and concomitantly pAMPKThr172 levels for up to 30 min in resistance exercised myofibers. This provides a time-course by which pRyR1Ser2843 can mechanistically impact Ca2+handling properties and consequently induce reduced myofiber contractility beyond immediate fatiguing mechanisms.
Collapse
|
24
|
Wang Y, Winters J, Subramaniam S. Functional classification of skeletal muscle networks. II. Applications to pathophysiology. J Appl Physiol (1985) 2012; 113:1902-20. [PMID: 23085957 DOI: 10.1152/japplphysiol.01515.2011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In our preceding companion paper (Wang Y, Winters J, Subramaniam S. J Appl Physiol. doi: 10.1152/japplphysiol.01514.2011), we used extensive expression profile data on normal human subjects, in combination with legacy knowledge to classify skeletal muscle function into four models, namely excitation-activation, mechanical, metabolic, and signaling-production model families. In this paper, we demonstrate how this classification can be applied to study two well-characterized myopathies: amyotrophic lateral sclerosis (ALS) and Duchenne muscular dystrophy (DMD). Using skeletal muscle profile data from ALS and DMD patients compared with that from normal subjects, normal young in the case of DMD, we delineate molecular mechanisms that are causative and consequential to skeletal muscle dysfunction. In ALS, our analysis establishes the metabolic role and specifically identifies the mechanisms of calcium dysregulation and defects in mitochondrial transport of materials as important for muscle dysfunction. In DMD, we illustrate how impaired mechanical function is strongly coordinated with other three functional networks, resulting in transformation of the skeletal muscle into hybrid forms as a compensatory mechanism. Our functional models also provide, in exquisite detail, the mechanistic role of myriad proteins in these four families in normal and disease function.
Collapse
Affiliation(s)
- Yu Wang
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093-0412, USA
| | | | | |
Collapse
|
25
|
Xu PT, Li Q, Sheng JJ, Chang H, Song Z, Yu ZB. Passive stretch reduces calpain activity through nitric oxide pathway in unloaded soleus muscles. Mol Cell Biochem 2012; 367:113-24. [PMID: 22547201 DOI: 10.1007/s11010-012-1325-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 04/18/2012] [Indexed: 11/25/2022]
Abstract
Unloading in spaceflight or long-term bed rest induces to pronounced atrophy of anti-gravity skeletal muscles. Passive stretch partially resists unloading-induced atrophy of skeletal muscle, but the mechanism remains elusive. The aims of this study were to investigate the hypotheses that stretch tension might increase protein level of neuronal nitric oxide synthase (nNOS) in unloaded skeletal muscle, and then nNOS-derived NO alleviated atrophy of skeletal muscle by inhibiting calpain activity. The tail-suspended rats were used to unload rat hindlimbs for 2 weeks, at the same time, left soleus muscle was stretched by applying a plaster cast to fix the ankle at 35° dorsiflexion. Stretch partially resisted atrophy and inhibited the decreased protein level and activity of nNOS in unloaded soleus muscles. Unloading increased frequency of calcium sparks and elevated intracellular resting and caffeine-induced Ca(2+) concentration ([Ca(2+)]i) in unloaded soleus muscle fibers. Stretch reduced frequency of calcium sparks and restored intracellular resting and caffeine-induced Ca(2+) concentration to control levels in unloaded soleus muscle fibers. The increased protein level and activity of calpain as well as the higher degradation of desmin induced by unloading were inhibited by stretch in soleus muscles. In conclusion, these results suggest that stretch can preserve the stability of sarcoplasmic reticulum Ca(2+) release channels which prevents the elevated [Ca(2+)]i by means of keeping nNOS activity, and then the enhanced protein level and activity of calpain return to control levels in unloaded soleus muscles. Therefore, stretch can resist in part atrophy of unloaded soleus muscles.
Collapse
Affiliation(s)
- Peng-Tao Xu
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | |
Collapse
|
26
|
nNOS regulation of skeletal muscle fatigue and exercise performance. Biophys Rev 2011; 3:209-217. [PMID: 28510048 DOI: 10.1007/s12551-011-0060-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 10/17/2011] [Indexed: 10/15/2022] Open
Abstract
Neuronal nitric oxide synthases (nNOS) are Ca2+/calmodulin-activated enzymes that synthesize the gaseous messenger nitric oxide (NO). nNOSμ and the recently described nNOSβ, both spliced nNOS isoforms, are important enzymatic sources of NO in skeletal muscle, a tissue long considered to be a paradigmatic system for studying NO-dependent redox signaling. nNOS is indispensable for skeletal muscle integrity and contractile performance, and deregulation of nNOSμ signaling is a common pathogenic feature of many neuromuscular diseases. Recent evidence suggests that both nNOSμ and nNOSβ regulate skeletal muscle size, strength, and fatigue resistance, making them important players in exercise performance. nNOSμ acts as an activity sensor and appears to assist skeletal muscle adaptation to new functional demands, particularly those of endurance exercise. Prolonged inactivity leads to nNOS-mediated muscle atrophy through a FoxO-dependent pathway. nNOS also plays a role in modulating exercise performance in neuromuscular disease. In the mdx mouse model of Duchenne muscular dystrophy, defective nNOS signaling is thought to restrict contractile capacity of working muscle in two ways: loss of sarcolemmal nNOSμ causes excessive ischemic damage while residual cytosolic nNOSμ contributes to hypernitrosylation of the ryanodine receptor, causing pathogenic Ca2+ leak. This defect in Ca2+ handling promotes muscle damage, weakness, and fatigue. This review addresses these recent advances in the understanding of nNOS-dependent redox regulation of skeletal muscle function and exercise performance under physiological and neuromuscular disease conditions.
Collapse
|
27
|
Salanova M, Bortoloso E, Schiffl G, Gutsmann M, Belavy DL, Felsenberg D, Furlan S, Volpe P, Blottner D. Expression and regulation of Homer in human skeletal muscle during neuromuscular junction adaptation to disuse and exercise. FASEB J 2011; 25:4312-25. [PMID: 21885651 DOI: 10.1096/fj.11-186049] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Protein calcium sensors of the Homer family have been proposed to modulate the activity of various ion channels and nuclear factor of activated T cells (NFAT), the transcription factor modulating skeletal muscle differentiation. We monitored Homer expression and subcellular localization in human skeletal muscle biopsies following 60 d of bedrest [Second Berlin Bedrest Study (BBR2-2)]. Soleus (SOL) and vastus lateralis (VL) biopsies were taken at start (pre) and at end (end) of bedrest from healthy male volunteers of a control group without exercise (CTR; n=9), a resistive-only exercise group (RE; n=7), and a combined resistive/vibration exercise group (RVE; n=7). Confocal analysis showed Homer immunoreactivity at the postsynaptic microdomain of the neuromuscular junction (NMJ) at bedrest start. After bedrest, Homer immunoreactivity decreased (CTR), remained unchanged (RE), or increased (RVE) at the NMJ. Homer2 mRNA and protein were differently regulated in a muscle-specific way. Activated NFATc1 translocates from cytoplasm to nucleus; increased amounts of NFATc1-immunopositive slow-type myonuclei were found in RVE myofibers of both muscles. Pulldown assays identified NFATc1 and Homer as molecular partners in skeletal muscle. A direct motor nerve control of Homer2 was confirmed in rat NMJs by in vivo denervation. Homer2 is localized at the NMJ and is part of the calcineurin-NFATc1 signaling pathway. RVE has additional benefit over RE as countermeasure preventing disuse-induced neuromuscular maladaptation during bedrest.
Collapse
Affiliation(s)
- Michele Salanova
- Center of Space Medicine Berlin, Neuromuscular Group, Institute of Anatomy, Charité Universitätsmedizin Berlin, Campus Charité Mitte, Philippstrasse 12, 10115, Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Marozkina NV, Gaston B. S-Nitrosylation signaling regulates cellular protein interactions. Biochim Biophys Acta Gen Subj 2011; 1820:722-9. [PMID: 21745537 DOI: 10.1016/j.bbagen.2011.06.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 06/13/2011] [Accepted: 06/16/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND S-Nitrosothiols are made by nitric oxide synthases and other metalloproteins. Unlike nitric oxide, S-nitrosothiols are involved in localized, covalent signaling reactions in specific cellular compartments. These reactions are enzymatically regulated. SCOPE S-Nitrosylation affects interactions involved in virtually every aspect of normal cell biology. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation. MAJOR CONCLUSIONS AND SIGNIFICANCE S-Nitrosylation is a regulated signaling reaction.
Collapse
Affiliation(s)
- Nadzeya V Marozkina
- University of Virginia School of Medicine, Division of Pediatric Respiratory Medicine, PO Box 800386, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
29
|
Ryan T, Sharma P, Ignatchenko A, MacLennan DH, Kislinger T, Gramolini AO. Identification of novel ryanodine receptor 1 (RyR1) protein interaction with calcium homeostasis endoplasmic reticulum protein (CHERP). J Biol Chem 2011; 286:17060-8. [PMID: 21454501 DOI: 10.1074/jbc.m110.197186] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ryanodine receptor type 1 (RyR1) is a homotetrameric Ca(2+) release channel located in the sarcoplasmic reticulum of skeletal muscle where it plays a role in the initiation of skeletal muscle contraction. A soluble, 6×-histidine affinity-tagged cytosolic fragment of RyR1 (amino acids 1-4243) was expressed in HEK-293 cells, and metal affinity chromatography under native conditions was used to purify the peptide together with interacting proteins. When analyzed by gel-free liquid chromatography mass spectrometry (LC-MS), 703 proteins were identified under all conditions. This group of proteins was filtered to identify putative RyR interacting proteins by removing those proteins found in only 1 RyR purification and proteins for which average spectral counts were enriched by less than 4-fold over control values. This resulted in 49 potential RyR1 interacting proteins, and 4 were selected for additional interaction studies: calcium homeostasis endoplasmic reticulum protein (CHERP), endoplasmic reticulum-Golgi intermediate compartment 53-kDa protein (LMAN1), T-complex protein, and phosphorylase kinase. Western blotting showed that only CHERP co-purified with affinity-tagged RyR1 and was eluted with imidazole. Immunofluorescence showed that endogenous CHERP co-localizes with endogenous RyR1 in the sarcoplasmic reticulum of rat soleus muscle. A combination of overexpression of RyR1 in HEK-293 cells with siRNA-mediated suppression of CHERP showed that CHERP affects Ca(2+) release from the ER via RyR1. Thus, we propose that CHERP is an RyR1 interacting protein that may be involved in the regulation of excitation-contraction coupling.
Collapse
Affiliation(s)
- Timothy Ryan
- Department of Physiology, University of Toronto, Ontario M5G 1L7, Canada
| | | | | | | | | | | |
Collapse
|
30
|
Cárdenas C, Juretić N, Bevilacqua JA, García IE, Figueroa R, Hartley R, Taratuto AL, Gejman R, Riveros N, Molgó J, Jaimovich E. Abnormal distribution of inositol 1,4,5‐trisphosphate receptors in human muscle can be related to altered calcium signals and gene expression in Duchenne dystrophy‐derived cells. FASEB J 2010; 24:3210-21. [DOI: 10.1096/fj.09-152017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- César Cárdenas
- Centro de Estudios Moleculares de la CélulaInstituto de Ciencias BiomédicasFacultad de MedicinaUniversidad de Chile Santiago Chile
- Department of PhysiologyUniversity of Pennsylvania Philadelphia Pennsylvania USA
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Nevenka Juretić
- Centro de Estudios Moleculares de la CélulaInstituto de Ciencias BiomédicasFacultad de MedicinaUniversidad de Chile Santiago Chile
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Jorge A. Bevilacqua
- Centro de Estudios Moleculares de la CélulaInstituto de Ciencias BiomédicasFacultad de MedicinaUniversidad de Chile Santiago Chile
- Programa de Anatomía y Biología del DesarrolloInstituto de Ciencias BiomédicasFacultad de MedicinaUniversidad de Chile Santiago Chile
- Departamento de Neurología y NeurocirugíaHospital Clínico Universidad de Chile Independencia Chile
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Isaac E. García
- Centro de Estudios Moleculares de la CélulaInstituto de Ciencias BiomédicasFacultad de MedicinaUniversidad de Chile Santiago Chile
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Reinaldo Figueroa
- Centro de Estudios Moleculares de la CélulaInstituto de Ciencias BiomédicasFacultad de MedicinaUniversidad de Chile Santiago Chile
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Ricardo Hartley
- Centro de Estudios Moleculares de la CélulaInstituto de Ciencias BiomédicasFacultad de MedicinaUniversidad de Chile Santiago Chile
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Ana L. Taratuto
- Departamento de NeuropatologíaInstituto de Investigaciones NeurológicasFLENI Buenos Aires Argentina
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Roger Gejman
- Departamento de Anatomía PatológicaFacultad de MedicinaPontificia Universidad Católica de Chile Santiago Chile
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Nora Riveros
- Centro de Estudios Moleculares de la CélulaInstituto de Ciencias BiomédicasFacultad de MedicinaUniversidad de Chile Santiago Chile
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Jordi Molgó
- Department of PhysiologyUniversity of Pennsylvania Philadelphia Pennsylvania USA
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| | - Enrique Jaimovich
- Centro de Estudios Moleculares de la CélulaInstituto de Ciencias BiomédicasFacultad de MedicinaUniversidad de Chile Santiago Chile
- Centre National de la Recherche ScientifiqueInstitut de Neurobiologie Alfred FessardFRC2118Laboratoire de Neurobiologie Cellulaire et Moléculaire UPR9040 Gif sur Yvette France
| |
Collapse
|
31
|
Vibration as an exercise modality: how it may work, and what its potential might be. Eur J Appl Physiol 2009; 108:877-904. [PMID: 20012646 DOI: 10.1007/s00421-009-1303-3] [Citation(s) in RCA: 437] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2009] [Indexed: 12/23/2022]
Abstract
Whilst exposure to vibration is traditionally regarded as perilous, recent research has focussed on potential benefits. Here, the physical principles of forced oscillations are discussed in relation to vibration as an exercise modality. Acute physiological responses to isolated tendon and muscle vibration and to whole body vibration exercise are reviewed, as well as the training effects upon the musculature, bone mineral density and posture. Possible applications in sports and medicine are discussed. Evidence suggests that acute vibration exercise seems to elicit a specific warm-up effect, and that vibration training seems to improve muscle power, although the potential benefits over traditional forms of resistive exercise are still unclear. Vibration training also seems to improve balance in sub-populations prone to fall, such as frail elderly people. Moreover, literature suggests that vibration is beneficial to reduce chronic lower back pain and other types of pain. Other future indications are perceivable.
Collapse
|
32
|
Extending the knowledge in histochemistry and cell biology. Histochem Cell Biol 2009; 133:1-40. [PMID: 19946696 DOI: 10.1007/s00418-009-0665-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2009] [Indexed: 01/21/2023]
Abstract
Central to modern Histochemistry and Cell Biology stands the need for visualization of cellular and molecular processes. In the past several years, a variety of techniques has been achieved bridging traditional light microscopy, fluorescence microscopy and electron microscopy with powerful software-based post-processing and computer modeling. Researchers now have various tools available to investigate problems of interest from bird's- up to worm's-eye of view, focusing on tissues, cells, proteins or finally single molecules. Applications of new approaches in combination with well-established traditional techniques of mRNA, DNA or protein analysis have led to enlightening and prudent studies which have paved the way toward a better understanding of not only physiological but also pathological processes in the field of cell biology. This review is intended to summarize articles standing for the progress made in "histo-biochemical" techniques and their manifold applications.
Collapse
|
33
|
Salanova M, Schiffl G, Blottner D. Atypical fast SERCA1a protein expression in slow myofibers and differential S-nitrosylation prevented by exercise during long term bed rest. Histochem Cell Biol 2009; 132:383-94. [DOI: 10.1007/s00418-009-0624-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2009] [Indexed: 10/20/2022]
|
34
|
Chopard A, Hillock S, Jasmin BJ. Molecular events and signalling pathways involved in skeletal muscle disuse-induced atrophy and the impact of countermeasures. J Cell Mol Med 2009; 13:3032-50. [PMID: 19656243 PMCID: PMC4516463 DOI: 10.1111/j.1582-4934.2009.00864.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Disuse-induced skeletal muscle atrophy occurs following chronic periods of inactivity such as those involving prolonged bed rest, trauma and microgravity environments. Deconditioning of skeletal muscle is mainly characterized by a loss of muscle mass, decreased fibre cross-sectional area, reduced force, increased fatigability, increased insulin resistance and transitions in fibre types. A description of the role of specific transcriptional mechanisms contributing to muscle atrophy by altering gene expression during muscle disuse has recently emerged and focused primarily on short period of inactivity. A better understanding of the transduction pathways involved in activation of proteolytic and apoptotic pathways continues to represent a major objective, together with the study of potential cross-talks in these cellular events. In parallel, evaluation of the impact of countermeasures at the cellular and molecular levels in short- and long-term disuse experimentations or microgravity environments should undoubtedly and synergistically increase our basic knowledge in attempts to identify new physical, pharmacological and nutritional targets to counteract muscle atrophy. These investigations are important as skeletal muscle atrophy remains an important neuromuscular challenge with impact in clinical and social settings affecting a variety of conditions such as those seen in aging, cancer cachexia, muscle pathologies and long-term space exploration.
Collapse
Affiliation(s)
- Angèle Chopard
- Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | | | | |
Collapse
|
35
|
Dalla Libera L, Ravara B, Gobbo V, Tarricone E, Vitadello M, Biolo G, Vescovo G, Gorza L. A transient antioxidant stress response accompanies the onset of disuse atrophy in human skeletal muscle. J Appl Physiol (1985) 2009; 107:549-57. [PMID: 19478193 DOI: 10.1152/japplphysiol.00280.2009] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
It is presently unknown whether oxidative stress increases in disused skeletal muscle in humans. Markers of oxidative stress were investigated in biopsies from the vastus lateralis muscle, collected from healthy subjects before [time 0 (T0)], after 1 wk (T8), and after 5 wk (T35) of bed rest. An 18% decrease in fiber cross-sectional area was detected in T35 biopsies (P<0.05). Carbonylation of muscle proteins significantly increased about twofold at T35 (P<0.02) and correlated positively with the decrease in fiber cross-sectional area (P=0.04). Conversely, T8 biopsies showed a significant increase in protein levels of heme oxygenase-1 and glucose-regulated protein-75 (Grp75)/mitochondrial heat shock protein-70, two stress proteins involved in the antioxidant defense (P<0.05). Heme oxygenase-1 increase, which involved a larger proportion of slow fibers compared with T0, appeared blunted in T35 biopsies. Grp75 protein level increased threefold in T8 biopsies and localized especially in slow fibers (P<0.025), to decrease significantly in T35 biopsies (P<0.05). Percent change in Grp75 levels positively correlated with fiber cross-sectional area (P=0.01). Parallel investigations on rat soleus muscles, performed after 1-15 days of hindlimb suspension, showed that Grp75 protein levels significantly increased after 24 h of unloading (P = 0.02), i.e., before statistically significant evidence of muscle atrophy, to decrease thereafter in relation to the degree of muscle atrophy (P=0.03). Therefore, in humans as in rodents, disuse muscle atrophy is characterized by increased protein carbonylation and by the blunting of the antioxidant stress response evoked by disuse.
Collapse
Affiliation(s)
- Luciano Dalla Libera
- Consiglio Nazionale delle Ricerche-Institute for Neuroscience, Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Sun J, Yamaguchi N, Xu L, Eu JP, Stamler JS, Meissner G. Regulation of the cardiac muscle ryanodine receptor by O(2) tension and S-nitrosoglutathione. Biochemistry 2009; 47:13985-90. [PMID: 19053230 DOI: 10.1021/bi8012627] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The cardiac and skeletal muscle sarcoplasmic reticulum ryanodine receptor Ca(2+) release channels contain thiols that are potential targets of endogenously produced reactive oxygen and nitrogen intermediates. Previously, we showed that the skeletal muscle ryanodine receptor (RyR1) has O(2)-sensitive thiols; only when these thiols are in the reduced state (pO(2) approximately 10 mmHg) can physiological concentrations of NO (nanomolar) activate RyR1. Here, we report that cardiac muscle ryanodine receptor (RyR2) activity also depends on pO(2), but unlike RyR1, RyR2 was not activated or S-nitrosylated directly by NO. Rather, activation and S-nitrosylation of RyR2 required S-nitrosoglutathione. The effects of peroxynitrite were indiscriminate on RyR1 and RyR2. Our results indicate that both RyR1 and RyR2 are pO(2)-responsive yet point to different mechanisms by which NO and S-nitrosoglutathione influence cardiac and skeletal muscle sarcoplasmic reticulum Ca(2+) release.
Collapse
Affiliation(s)
- Junhui Sun
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | |
Collapse
|
37
|
State-of-the-art technologies, current opinions and developments, and novel findings: news from the field of histochemistry and cell biology. Histochem Cell Biol 2008; 130:1205-51. [PMID: 18985372 DOI: 10.1007/s00418-008-0535-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2008] [Indexed: 10/25/2022]
Abstract
Investigations of cell and tissue structure and function using innovative methods and approaches have again yielded numerous exciting findings in recent months and have added important data to current knowledge, inspiring new ideas and hypotheses in various fields of modern life sciences. Topics and contents of comprehensive expert reviews covering different aspects in methodological advances, cell biology, tissue function and morphology, and novel findings reported in original papers are summarized in the present review.
Collapse
|
38
|
Wang Y, Liu T, Wu C, Li H. A strategy for direct identification of protein S-nitrosylation sites by quadrupole time-of-flight mass spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2008; 19:1353-60. [PMID: 18635375 PMCID: PMC2577058 DOI: 10.1016/j.jasms.2008.06.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 06/03/2008] [Accepted: 06/05/2008] [Indexed: 05/22/2023]
Abstract
S-nitrosylation of proteins serves an important role in regulating diverse cellular processes including signal transduction, DNA repair, and neurotransmission. Identification of S-nitrosylation sites is crucial for understanding the significance of this post-translational modification (PTM) in modulating the function of a protein. However, it is challenging to identify S-nitrosylation sites directly by mass spectrometric (MS) methods due to the labile nature of the S-NO bond. Here we describe a strategy for direct identification of protein S-nitrosylation sites in an electrospray ionization (ESI) quadrupole time-of-flight (QTOF) mass spectrometer without prior chemical derivatization of S-nitrosylated peptides. Both sample buffer composition and MS hardware parameters were carefully adjusted to ensure that S-nitrosylated peptide ions could be analyzed by the QTOF MS with optimal signal/noise ratios. It was crucial that the proteins were preserved in a sample solution containing 1 mM EDTA and 0.1 mM neocuproine at neutral pH. Proteins dissolved in this solution are amenable to in-solution tryptic digestion, which is important for the analysis of biological samples. S-nitrosylated peptides were effectively analyzed by LC/MS/MS on QTOF MS, with an optimized cone voltage of 20 V and collision energy of 4 V. We have successfully applied this method to thioredoxin, a key antioxidant protein, and identified within it an S-nitrosylation site at Cys73.
Collapse
Affiliation(s)
- Yan Wang
- Center for Advanced Proteomics Research and Department of Biochemistry and Molecular Biology, UMDNJ -New Jersey Medical School Cancer Center, Newark, NJ 07103
- School of Pharmacy, Shanghai Jiaotong University, Shanghai, 200240, People's Republic of China
| | - Tong Liu
- Center for Advanced Proteomics Research and Department of Biochemistry and Molecular Biology, UMDNJ -New Jersey Medical School Cancer Center, Newark, NJ 07103
| | - Changgong Wu
- Center for Advanced Proteomics Research and Department of Biochemistry and Molecular Biology, UMDNJ -New Jersey Medical School Cancer Center, Newark, NJ 07103
| | - Hong Li
- Center for Advanced Proteomics Research and Department of Biochemistry and Molecular Biology, UMDNJ -New Jersey Medical School Cancer Center, Newark, NJ 07103
| |
Collapse
|