1
|
Miyazaki D, Sato M, Shiba N, Yoshizawa T, Nakamura A. Becker muscular dystrophy mice showed site-specific decay of type IIa fibers with capillary change in skeletal muscle. eLife 2025; 13:RP100665. [PMID: 40094282 PMCID: PMC11913446 DOI: 10.7554/elife.100665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
Becker muscular dystrophy (BMD), an X-linked muscular dystrophy, is mostly caused by an in-frame deletion of Duchenne muscular dystrophy (DMD). BMD severity varies from asymptomatic to severe, associated with the genotype of DMD. However, the underlying mechanisms remain unclear. We established BMD mice carrying three representative exon deletions: ex45-48 del., ex45-47 del., and ex45-49 del. (d45-48, d45-47, and d45-49), with high frequencies and different severities in the human BMD hotspot. All three BMD mice showed muscle weakness, muscle degeneration, and fibrosis, but these changes appeared at different times for each exon deletion, consistent with the severities obtained by the natural history study of BMD. BMD mice showed site-specific muscle changes, unlike mdx mice, which showed diffuse muscle changes, and we demonstrated selective type IIa fiber reduction in BMD mice. Furthermore, BMD mice showed sarcolemmal neuronal nitric oxide synthase (nNOS) reduction and morphological capillary changes around type IIa fibers. These results suggest that capillary changes caused by nNOS reduction may be associated with the mechanism of skeletal muscle degeneration and type IIa fiber reduction in BMD mice. BMD mice may be useful in elucidating the pathomechanisms and developing vascular targeted therapies for human BMD.
Collapse
Affiliation(s)
- Daigo Miyazaki
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of MedicineMatsumotoJapan
- Intractable Disease Care Center, Shinshu University HospitalMatsumotoJapan
| | - Mitsuto Sato
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of MedicineMatsumotoJapan
- Department of Brain Disease Research, Shinshu University School of MedicineMatsumotoJapan
| | - Naoko Shiba
- Department of Regenerative Science and Medicine, Shinshu UniversityMatsumotoJapan
| | - Takahiro Yoshizawa
- Research Center for Advanced Science and Technology, Shinshu UniversityMatsumotoJapan
| | - Akinori Nakamura
- Department of Clinical Research, NHO Matsumoto Medical CenterMatsumotoJapan
- Third Department of Medicine, Shinshu University School of MedicineMatsumotoJapan
| |
Collapse
|
2
|
Bahadoran Z, Mirmiran P, Ghasemi A. Type 2 diabetes-related sarcopenia: role of nitric oxide. Nutr Metab (Lond) 2024; 21:107. [PMID: 39695784 DOI: 10.1186/s12986-024-00883-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Sarcopenia, characterized by progressive and generalized loss of skeletal muscle (SkM) mass, strength, and physical performance, is a prevalent complication in type 2 diabetes (T2D). Nitric oxide (NO), a multifunctional gasotransmitter involved in whole-body glucose and insulin homeostasis, plays key roles in normal SkM physiology and function. Here, we highlight the role of NO in SkM mass maintenance and its potential contribution to the development of T2D-related sarcopenia. Physiologic NO level, primarily produced by sarcolemmal neuronal nitric oxide synthase (nNOSμ isoform), is involved in protein synthesis in muscle fibers and maintenance of SkM mass. The observed effect of nNOSμ on SkM mass is muscle-type specific and sex-dependent. Impaired NO homeostasis [due to a diminished nNOSμ-NO availability and excessive NO production through inducible NOS (iNOS) in response to atrophic stimuli, e.g., inflammatory cytokines] in SkM occurred during the development and progression of T2D, may cause sarcopenia. Theoretically, restoration of NO through nNOS overexpression, supplying NOS substrates (e.g., L-arginine and L-citrulline), phosphodiesterase (PDE) inhibition, and supplementation with NO donors (e.g., inorganic nitrate) may be potential therapeutic approaches to preserve SkM mass and prevents sarcopenia in T2D.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Micronutrient Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Sahid-Erabi St, Yemen St, Chamran Exp, P.O. Box 19395-4763, Tehran, Iran.
| |
Collapse
|
3
|
Nguyen MN, Hooper C, Stefanini M, Vrellaku B, Carnicer R, Wood MJ, Simon JN, Casadei B. Why is early-onset atrial fibrillation uncommon in patients with Duchenne muscular dystrophy? Insights from the mdx mouse. Cardiovasc Res 2024; 120:519-530. [PMID: 38270932 PMCID: PMC11060487 DOI: 10.1093/cvr/cvae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024] Open
Abstract
AIMS A reduction in both dystrophin and neuronal nitric oxide synthase (NOS1) secondary to microRNA-31 (miR-31) up-regulation contributes to the atrial electrical remodelling that underpins human and experimental atrial fibrillation (AF). In contrast, patients with Duchenne muscular dystrophy (DMD), who lack dystrophin and NOS1 and, at least in the skeletal muscle, have raised miR-31 expression, do not have increase susceptibility to AF in the absence of left ventricular (LV) dysfunction. Here, we investigated whether dystrophin deficiency is also associated with atrial up-regulation of miR-31, loss of NOS1 protein, and increased AF susceptibility in young mdx mice. METHODS AND RESULTS Echocardiography showed normal cardiac structure and function in 12-13 weeks mdx mice, with no indication by assay of hydroxyproline that atrial fibrosis had developed. The absence of dystrophin in mdx mice was accompanied by an overall reduction in syntrophin and a lower NOS1 protein content in the skeletal muscle and in the left atrial and ventricular myocardium, with the latter occurring alongside reduced Nos1 transcript levels (exons 1-2 by quantitative polymerase chain reaction) and an increase in NOS1 polyubiquitination [assessed using tandem polyubiquitination pulldowns; P < 0.05 vs. wild type (WT)]. Neither the up-regulation of miR-31 nor the substantial reduction in NOS activity observed in the skeletal muscle was present in the atrial tissue of mdx mice. At difference with the skeletal muscle, the mdx atrial myocardium showed a reduction in the constitutive NOS inhibitor, caveolin-1, coupled with an increase in NOS3 serine1177 phosphorylation, in the absence of differences in the protein content of other NOS isoforms or in the relative expression NOS1 splice variants. In line with these findings, transoesophageal atrial burst pacing revealed no difference in AF susceptibility between mdx mice and their WT littermates. CONCLUSION Dystrophin depletion is not associated with atrial miR-31 up-regulation, reduced NOS activity, or increased AF susceptibility in the mdx mouse. Compared with the skeletal muscle, the milder atrial biochemical phenotype may explain why patients with DMD do not exhibit a higher prevalence of atrial arrhythmias despite a reduction in NOS1 content.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, L6 West Wing, Oxford OX3 9DU, UK
| | - Charlotte Hooper
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, L6 West Wing, Oxford OX3 9DU, UK
| | - Matilde Stefanini
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, L6 West Wing, Oxford OX3 9DU, UK
| | - Besarte Vrellaku
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, L6 West Wing, Oxford OX3 9DU, UK
| | - Ricardo Carnicer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, L6 West Wing, Oxford OX3 9DU, UK
| | - Matthew J Wood
- Department of Paediatrics and Muscular Dystrophy UK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Jillian N Simon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, L6 West Wing, Oxford OX3 9DU, UK
| | - Barbara Casadei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, L6 West Wing, Oxford OX3 9DU, UK
| |
Collapse
|
4
|
Supruniuk E, Górski J, Chabowski A. Endogenous and Exogenous Antioxidants in Skeletal Muscle Fatigue Development during Exercise. Antioxidants (Basel) 2023; 12:antiox12020501. [PMID: 36830059 PMCID: PMC9952836 DOI: 10.3390/antiox12020501] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Muscle fatigue is defined as a decrease in maximal force or power generated in response to contractile activity, and it is a risk factor for the development of musculoskeletal injuries. One of the many stressors imposed on skeletal muscle through exercise is the increased production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), which intensifies as a function of exercise intensity and duration. Exposure to ROS/RNS can affect Na+/K+-ATPase activity, intramyofibrillar calcium turnover and sensitivity, and actin-myosin kinetics to reduce muscle force production. On the other hand, low ROS/RNS concentrations can likely upregulate an array of cellular adaptative responses related to mitochondrial biogenesis, glucose transport and muscle hypertrophy. Consequently, growing evidence suggests that exogenous antioxidant supplementation might hamper exercise-engendering upregulation in the signaling pathways of mitogen-activated protein kinases (MAPKs), peroxisome-proliferator activated co-activator 1α (PGC-1α), or mammalian target of rapamycin (mTOR). Ultimately, both high (exercise-induced) and low (antioxidant intervention) ROS concentrations can trigger beneficial responses as long as they do not override the threshold range for redox balance. The mechanisms underlying the two faces of ROS/RNS in exercise, as well as the role of antioxidants in muscle fatigue, are presented in detail in this review.
Collapse
Affiliation(s)
- Elżbieta Supruniuk
- Department of Physiology, Medical University of Białystok, 15-222 Białystok, Poland
- Correspondence: ; Tel.: +48-(85)-748-55-85
| | - Jan Górski
- Department of Medical Sciences, Academy of Applied Sciences, 18-400 Łomża, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Białystok, 15-222 Białystok, Poland
| |
Collapse
|
5
|
Basile L, Marino M, La Vignera S. Is sildenafil a doping drug in hypoxic conditions? Aging Male 2022; 25:156-158. [PMID: 35612871 DOI: 10.1080/13685538.2022.2079628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Livia Basile
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - Marta Marino
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
6
|
Boyd A, Montandon M, Wood AJ, Currie PD. FKRP directed fibronectin glycosylation: A novel mechanism giving insights into muscular dystrophies? Bioessays 2022; 44:e2100270. [PMID: 35229908 DOI: 10.1002/bies.202100270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 12/15/2022]
Abstract
The recently uncovered role of Fukutin-related protein (FKRP) in fibronectin glycosylation has challenged our understanding of the basis of disease pathogenesis in the muscular dystrophies. FKRP is a Golgi-resident glycosyltransferase implicated in a broad spectrum of muscular dystrophy (MD) pathologies that are not fully attributable to the well-described α-Dystroglycan hypoglycosylation. By revealing a new role for FKRP in the glycosylation of fibronectin, a modification critical for the development of the muscle basement membrane (MBM) and its associated muscle linkages, new possibilities for understanding clinical phenotype arise. This modification involves an interaction between FKRP and myosin-10, a protein involved in the Golgi organization and function. These observations suggest a FKRP nexus exists that controls two critical aspects to muscle fibre integrity, both fibre stability at the MBM and its elastic properties. This review explores the new potential disease axis in the context of our current knowledge of muscular dystrophies.
Collapse
Affiliation(s)
- Andrew Boyd
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Margo Montandon
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
7
|
Kasamatsu S, Tsutsuki H, Ida T, Sawa T, Watanabe Y, Akaike T, Ihara H. Regulation of nitric oxide/reactive oxygen species redox signaling by nNOS splicing variants. Nitric Oxide 2022; 120:44-52. [PMID: 35033681 DOI: 10.1016/j.niox.2022.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 10/19/2022]
Abstract
We previously demonstrated different expression patterns of the neuronal nitric oxide synthase (nNOS) splicing variants, nNOS-μ and nNOS-α, in the rat brain; however, their exact functions have not been fully elucidated. In this study, we compared the enzymatic activities of nNOS-μ and nNOS-α and investigated intracellular redox signaling in nNOS-expressing PC12 cells, stimulated with a neurotoxicant, 1-methyl-4-phenylpyridinium ion (MPP+), to enhance the nNOS uncoupling reaction. Using in vitro studies, we show that nNOS-μ produced nitric oxide (NO), as did nNOS-α, in the presence of tetrahydrobiopterin (BH4), an important cofactor for the enzymatic activity. However, nNOS-μ generated more NO and less superoxide than nNOS-α in the absence of BH4. MPP + treatment induced more reactive oxygen species (ROS) production in nNOS-α-expressing PC12 cells than in those expressing nNOS-μ, which correlated with the intracellular production of 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), a downstream messenger of nNOS redox signaling, and apoptosis in these cells. Furthermore, post-treatment with 8-nitro-cGMP aggravated MPP+-induced cytotoxicity via activation of the H-Ras/extracellular signal-regulated kinase signaling pathway. In conclusion, our results provide strong evidence that nNOS-μ exhibits distinctive enzymatic properties of NO/ROS production, contributing to the regulation of intracellular redox signaling, including the downstream production of 8-nitro-cGMP.
Collapse
Affiliation(s)
- Shingo Kasamatsu
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Osaka, 599-8531, Japan
| | - Hiroyasu Tsutsuki
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yasuo Watanabe
- Department of Pharmacology, Showa Pharmaceutical University, Tokyo, 194-8543, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Hideshi Ihara
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Osaka, 599-8531, Japan.
| |
Collapse
|
8
|
Pavitt MJ, Lewis A, Buttery SC, Fernandez BO, Mikus-Lelinska M, Banya WAS, Feelisch M, Polkey MI, Hopkinson NS. Dietary nitrate supplementation to enhance exercise capacity in hypoxic COPD: EDEN-OX, a double-blind, placebo-controlled, randomised cross-over study. Thorax 2021; 77:968-975. [PMID: 34853156 DOI: 10.1136/thoraxjnl-2021-217147] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/04/2021] [Indexed: 11/03/2022]
Abstract
RATIONALE Dietary nitrate supplementation improves skeletal muscle oxygen utilisation and vascular endothelial function. We hypothesised that these effects might be sufficient to improve exercise performance in patients with COPD and hypoxia severe enough to require supplemental oxygen. METHODS We conducted a single-centre, double-blind, placebo-controlled, cross-over study, enrolling adults with COPD who were established users of long-term oxygen therapy. Participants performed an endurance shuttle walk test, using their prescribed oxygen, 3 hours after consuming either 140 mL of nitrate-rich beetroot juice (BRJ) (12.9 mmol nitrate) or placebo (nitrate-depleted BRJ). Treatment order was allocated (1:1) by computer-generated block randomisation. MEASUREMENTS The primary outcome was endurance shuttle walk test time. The secondary outcomes included area under the curve to isotime for fingertip oxygen saturation and heart rate parameters during the test, blood pressure, and endothelial function assessed using flow-mediated dilatation. Plasma nitrate and nitrite levels as well as FENO were also measured. MAIN RESULTS 20 participants were recruited and all completed the study. Nitrate-rich BRJ supplementation prolonged exercise endurance time in all participants as compared with placebo: median (IQR) 194.6 (147.5-411.7) s vs 159.1 (121.9-298.5) s, estimated treatment effect 62 (33-106) s (p<0.0001). Supplementation also improved endothelial function: NR-BRJ group +4.1% (-1.1% to 14.8%) vs placebo BRJ group -5.0% (-10.6% to -0.6%) (p=0.0003). CONCLUSION Acute dietary nitrate supplementation increases exercise endurance in patients with COPD who require supplemental oxygen.Trial registration number ISRCTN14888729.
Collapse
Affiliation(s)
- Matthew J Pavitt
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Adam Lewis
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sara C Buttery
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | - Winston A S Banya
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin Feelisch
- Faculty of Medicine, Clinical and Experimental Sciences, University of Southampton, Southampton, UK.,Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, UK
| | - Michael I Polkey
- National Heart and Lung Institute, Imperial College London, London, UK.,Respiratory Medicine, Royal Brompton Hospital, London, UK
| | | |
Collapse
|
9
|
Weissman D, Maack C. Redox signaling in heart failure and therapeutic implications. Free Radic Biol Med 2021; 171:345-364. [PMID: 34019933 DOI: 10.1016/j.freeradbiomed.2021.05.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/17/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Heart failure is a growing health burden worldwide characterized by alterations in excitation-contraction coupling, cardiac energetic deficit and oxidative stress. While current treatments are mostly limited to antagonization of neuroendocrine activation, more recent data suggest that also targeting metabolism may provide substantial prognostic benefit. However, although in a broad spectrum of preclinical models, oxidative stress plays a causal role for the development and progression of heart failure, no treatment that targets reactive oxygen species (ROS) directly has entered the clinical arena yet. In the heart, ROS derive from various sources, such as NADPH oxidases, xanthine oxidase, uncoupled nitric oxide synthase and mitochondria. While mitochondria are the primary source of ROS in the heart, communication between different ROS sources may be relevant for physiological signalling events as well as pathologically elevated ROS that deteriorate excitation-contraction coupling, induce hypertrophy and/or trigger cell death. Here, we review the sources of ROS in the heart, the modes of pathological activation of ROS formation as well as therapeutic approaches that may target ROS specifically in mitochondria.
Collapse
Affiliation(s)
- David Weissman
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany; Department of Internal Medicine 1, University Clinic Würzburg, Würzburg, Germany.
| |
Collapse
|
10
|
Aguiar AF, Casonatto J. Effects of Citrulline Malate Supplementation on Muscle Strength in Resistance-Trained Adults: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J Diet Suppl 2021; 19:772-790. [PMID: 34176406 DOI: 10.1080/19390211.2021.1939473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Although the ergogenic mechanisms of supplementation with citrulline malate are well known, unclear findings regarding variables of muscle strength have been recorded. Such misleading findings in the literature illustrate the need for well-conducted meta-analysis research to elucidate the possible ergogenic impact, which could have major practical consequences for athletes and recreational practitioners seeking to optimize gains in muscle strength. The objective of this systematic review was to summarize the existing literature that evaluated the effects of citrulline malate supplementation on muscle strength outcomes from resistance exercise in resistance-trained individuals. A systematic electronic search in Medline and Scientific Electronic Library Online (SciELO) was completed in August 2020 identifying randomized controlled trials investigating the effect of citrulline malate supplementation on muscle strength in resistance-trained adults. A subsequent meta-analysis was performed. The meta-analysis involved four studies and 138 assessments (69 in citrulline-malate and 69 in placebo groups). We did not observe an overall effect favoring citrulline-malate supplementation (SMD95% = 0.13 [-0.21; 0.46]). Considering the lower (SMD95% = 0.06 [-0.47; 0.60]) and upper (SMD95% = 0.17 [-0.26; 0.60]) limbs, a non-significant overall effect was identified. The mean effects were similar for "limbs" (upper vs lower) [p = 0.763]. Accordingly, our findings suggest that citrulline malate supplementation does not improve muscle strength in healthy and resistance-trained individuals (PROSPERO registration number: CRD42020159338).
Collapse
Affiliation(s)
- Andreo F Aguiar
- Research Laboratory in Muscular System and Physical Exercise, University of Northern Paraná, Londrina, Brazil
| | - Juliano Casonatto
- Research Group in Physiology and Physical Activity, University of Northern Paraná, Londrina, Brazil
| |
Collapse
|
11
|
Senefeld JW, Wiggins CC, Regimbal RJ, Dominelli PB, Baker SE, Joyner MJ. Ergogenic Effect of Nitrate Supplementation: A Systematic Review and Meta-analysis. Med Sci Sports Exerc 2021; 52:2250-2261. [PMID: 32936597 PMCID: PMC7494956 DOI: 10.1249/mss.0000000000002363] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Supplemental digital content is available in the text. Although over 100 studies and reviews have examined the ergogenic effects of dietary nitrate (NO3−) supplementation in young, healthy men and women, it is unclear if participant and environmental factors modulate the well-described ergogenic effects—particularly relevant factors include biological sex, aerobic fitness, and fraction of inspired oxygen (FiO2) during exercise. To address this limitation, the literature was systematically reviewed for randomized, crossover, placebo-controlled studies reporting exercise performance outcome metrics with NO3− supplementation in young, healthy adults. Of the 2033 articles identified, 80 were eligible for inclusion in the meta-analysis. Random-effects meta-analysis demonstrated that exercise performance improved with NO3− supplementation compared with placebo (d = 0.174; 95% confidence interval (CI), 0.120–0.229; P < 0.001). Subgroup analyses conducted on biological sex, aerobic fitness, and FiO2 demonstrated that the ergogenic effect of NO3− supplementation was as follows: 1) not observed in studies with only women (n = 6; d = 0.116; 95% CI, −0.126 to 0.358; P = 0.347), 2) not observed in well-trained endurance athletes (≥65 mL·kg−1·min−1; n = 26; d = 0.021; 95% CI, −0.103 to 0.144; P = 0.745), and 3) not modulated by FiO2 (hypoxia vs normoxia). Together, the meta-analyses demonstrated a clear ergogenic effect of NO3− supplementation in recreationally active, young, healthy men across different exercise paradigms and NO3− supplementation parameters; however, the effect size of NO3− supplementation was objectively small (d = 0.174). NO3− supplementation has more limited utility as an ergogenic aid in participants with excellent aerobic fitness that have optimized other training parameters. Mechanistic research and studies incorporating a wide variety of subjects (e.g., women) are needed to advance the study of NO3− supplementation; however, additional descriptive studies of young, healthy men may have limited utility.
Collapse
Affiliation(s)
- Jonathon W Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Chad C Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Riley J Regimbal
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | | | - Sarah E Baker
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
12
|
Potter RA, Griffin DA, Heller KN, Peterson EL, Clark EK, Mendell JR, Rodino-Klapac LR. Dose-Escalation Study of Systemically Delivered rAAVrh74.MHCK7.micro-dystrophin in the mdx Mouse Model of Duchenne Muscular Dystrophy. Hum Gene Ther 2021; 32:375-389. [PMID: 33397205 PMCID: PMC8063270 DOI: 10.1089/hum.2019.255] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare, X-linked, fatal, degenerative neuromuscular disease caused by mutations in the DMD gene. More than 2,000 mutations of the DMD gene are responsible for progressive loss of muscle strength, loss of ambulation, and generally respiratory and cardiac failure by age 30. Recently, gene transfer therapy has received widespread interest as a disease-modifying treatment for all patients with DMD. We designed an adeno-associated virus vector (rAAVrh74) containing a codon-optimized human micro-dystrophin transgene driven by a skeletal and cardiac muscle-specific promoter, MHCK7. To test the efficacy of rAAVrh74.MHCK7.micro-dystrophin, we evaluated systemic injections in mdx (dystrophin-null) mice at low (2 × 1012 vector genome [vg] total dose, 8 × 1013 vg/kg), intermediate (6 × 1012 vg total dose, 2 × 1014 vg/kg), and high doses (1.2 × 1013 vg total dose, 6 × 1014 vg/kg). Three months posttreatment, specific force increased in the diaphragm (DIA) and tibialis anterior muscle, with intermediate and high doses eliciting force outputs at wild-type (WT) levels. Histological improvement included reductions in fibrosis and normalization of myofiber size, specifically in the DIA, where results for low and intermediate doses were not significantly different from the WT. Significant reduction in central nucleation was also observed, although complete normalization to WT was not seen. No vector-associated toxicity was reported either by clinical or organ-specific laboratory assessments or following formal histopathology. The findings in this preclinical study provided proof of principle for safety and efficacy of systemic delivery of rAAVrh74.MHCK7.micro-dystrophin at high vector titers, supporting initiation of a Phase I/II safety study in boys with DMD.
Collapse
Affiliation(s)
- Rachael A Potter
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA.,Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Danielle A Griffin
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA.,Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Kristin N Heller
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Ellyn L Peterson
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA.,Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Emma K Clark
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jerry R Mendell
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio, USA
| | - Louise R Rodino-Klapac
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA.,Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
13
|
Boycott HE, Nguyen MN, Vrellaku B, Gehmlich K, Robinson P. Nitric Oxide and Mechano-Electrical Transduction in Cardiomyocytes. Front Physiol 2020; 11:606740. [PMID: 33384614 PMCID: PMC7770138 DOI: 10.3389/fphys.2020.606740] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/23/2020] [Indexed: 12/22/2022] Open
Abstract
The ability§ of the heart to adapt to changes in the mechanical environment is critical for normal cardiac physiology. The role of nitric oxide is increasingly recognized as a mediator of mechanical signaling. Produced in the heart by nitric oxide synthases, nitric oxide affects almost all mechano-transduction pathways within the cardiomyocyte, with roles mediating mechano-sensing, mechano-electric feedback (via modulation of ion channel activity), and calcium handling. As more precise experimental techniques for applying mechanical stresses to cells are developed, the role of these forces in cardiomyocyte function can be further understood. Furthermore, specific inhibitors of different nitric oxide synthase isoforms are now available to elucidate the role of these enzymes in mediating mechano-electrical signaling. Understanding of the links between nitric oxide production and mechano-electrical signaling is incomplete, particularly whether mechanically sensitive ion channels are regulated by nitric oxide, and how this affects the cardiac action potential. This is of particular relevance to conditions such as atrial fibrillation and heart failure, in which nitric oxide production is reduced. Dysfunction of the nitric oxide/mechano-electrical signaling pathways are likely to be a feature of cardiac pathology (e.g., atrial fibrillation, cardiomyopathy, and heart failure) and a better understanding of the importance of nitric oxide signaling and its links to mechanical regulation of heart function may advance our understanding of these conditions.
Collapse
Affiliation(s)
- Hannah E. Boycott
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - My-Nhan Nguyen
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Besarte Vrellaku
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Péladeau C, Jasmin BJ. Targeting IRES-dependent translation as a novel approach for treating Duchenne muscular dystrophy. RNA Biol 2020; 18:1238-1251. [PMID: 33164678 DOI: 10.1080/15476286.2020.1847894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Internal-ribosomal entry sites (IRES) are translational elements that allow the initiation machinery to start protein synthesis via internal initiation. IRESs promote tissue-specific translation in stress conditions when conventional cap-dependent translation is inhibited. Since many IRES-containing mRNAs are relevant to diseases, this cellular mechanism is emerging as an attractive therapeutic target for pharmacological and genetic modulations. Indeed, there has been growing interest over the past years in determining the therapeutic potential of IRESs for several disease conditions such as cancer, neurodegeneration and neuromuscular diseases including Duchenne muscular dystrophy (DMD). IRESs relevant for DMD have been identified in several transcripts whose protein product results in functional improvements in dystrophic muscles. Together, these converging lines of evidence indicate that activation of IRES-mediated translation of relevant transcripts in DMD muscle represents a novel and appropriate therapeutic strategy for DMD that warrants further investigation, particularly to identify agents that can modulate their activity.
Collapse
Affiliation(s)
- Christine Péladeau
- Department of Cellular and Molecular Medicine, and the Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, and the Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
15
|
Wang WL, Ge TY, Chen X, Mao Y, Zhu YZ. Advances in the Protective Mechanism of NO, H 2S, and H 2 in Myocardial Ischemic Injury. Front Cardiovasc Med 2020; 7:588206. [PMID: 33195476 PMCID: PMC7661694 DOI: 10.3389/fcvm.2020.588206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/28/2020] [Indexed: 12/30/2022] Open
Abstract
Myocardial ischemic injury is among the top 10 leading causes of death from cardiovascular diseases worldwide. Myocardial ischemia is caused mainly by coronary artery occlusion or obstruction. It usually occurs when the heart is insufficiently perfused, oxygen supply to the myocardium is reduced, and energy metabolism in the myocardium is abnormal. Pathologically, myocardial ischemic injury generates a large number of inflammatory cells, thus inducing a state of oxidative stress. This sharp reduction in the number of normal cells as a result of apoptosis leads to organ and tissue damage, which can be life-threatening. Therefore, effective methods for the treatment of myocardial ischemic injury and clarification of the underlying mechanisms are urgently required. Gaseous signaling molecules, such as NO, H2S, H2, and combined gas donors, have gradually become a focus of research. Gaseous signaling molecules have shown anti-apoptotic, anti-oxidative and anti-inflammatory effects as potential therapeutic agents for myocardial ischemic injury in a large number of studies. In this review, we summarize and discuss the mechanism underlying the protective effect of gaseous signaling molecules on myocardial ischemic injury.
Collapse
Affiliation(s)
| | | | - Xu Chen
- Guilin Medical College, Guilin, China
| | - Yicheng Mao
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yi-Zhun Zhu
- Guilin Medical College, Guilin, China.,Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
16
|
Garbincius JF, Merz LE, Cuttitta AJ, Bayne KV, Schrade S, Armstead EA, Converso-Baran KL, Whitesall SE, D'Alecy LG, Michele DE. Enhanced dimethylarginine degradation improves coronary flow reserve and exercise tolerance in Duchenne muscular dystrophy carrier mice. Am J Physiol Heart Circ Physiol 2020; 319:H582-H603. [PMID: 32762558 DOI: 10.1152/ajpheart.00333.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked disease caused by null mutations in dystrophin and characterized by muscle degeneration. Cardiomyopathy is common and often prevalent at similar frequency in female DMD carriers irrespective of whether they manifest skeletal muscle disease. Impaired muscle nitric oxide (NO) production in DMD disrupts muscle blood flow regulation and exaggerates postexercise fatigue. We show that circulating levels of endogenous methylated arginines including asymmetric dimethylarginine (ADMA), which act as NO synthase inhibitors, are elevated by acute necrotic muscle damage and in chronically necrotic dystrophin-deficient mice. We therefore hypothesized that excessive ADMA impairs muscle NO production and diminishes exercise tolerance in DMD. We used transgenic expression of dimethylarginine dimethylaminohydrolase 1 (DDAH), which degrades methylated arginines, to investigate their contribution to exercise-induced fatigue in DMD. Although infusion of exogenous ADMA was sufficient to impair exercise performance in wild-type mice, transgenic DDAH expression did not rescue exercise-induced fatigue in dystrophin-deficient male mdx mice. Surprisingly, DDAH transgene expression did attenuate exercise-induced fatigue in dystrophin-heterozygous female mdx carrier mice. Improved exercise tolerance was associated with reduced heart weight and improved cardiac β-adrenergic responsiveness in DDAH-transgenic mdx carriers. We conclude that DDAH overexpression increases exercise tolerance in female DMD carriers, possibly by limiting cardiac pathology and preserving the heart's responses to changes in physiological demand. Methylated arginine metabolism may be a new target to improve exercise tolerance and cardiac function in DMD carriers or act as an adjuvant to promote NO signaling alongside therapies that partially restore dystrophin expression in patients with DMD.NEW & NOTEWORTHY Duchenne muscular dystrophy (DMD) carriers are at risk for cardiomyopathy. The nitric oxide synthase inhibitor asymmetric dimethylarginine (ADMA) is released from damaged muscle in DMD and impairs exercise performance. Transgenic expression of dimethylarginine dimethylaminohydrolase to degrade ADMA prevents cardiac hypertrophy, improves cardiac function, and improves exercise tolerance in DMD carrier mice. These findings highlight the relevance of ADMA to muscular dystrophy and have important implications for therapies targeting nitric oxide in patients with DMD and DMD carriers.
Collapse
Affiliation(s)
- Joanne F Garbincius
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Lauren E Merz
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Ashley J Cuttitta
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Kaitlynn V Bayne
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Sara Schrade
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Emily A Armstead
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | | | - Steven E Whitesall
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.,Physiology Phenotyping Core, University of Michigan, Ann Arbor, Michigan
| | - Louis G D'Alecy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.,Physiology Phenotyping Core, University of Michigan, Ann Arbor, Michigan
| | - Daniel E Michele
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.,Physiology Phenotyping Core, University of Michigan, Ann Arbor, Michigan.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
17
|
Cavallaro M, Guerrera MC, Abbate F, Levanti MB, Laurà R, Ammendolia G, Malara D, Stipa MG, Battaglia P. Morphological, ultrastructural and immunohistochemical study on the skin ventral photophores of
Diaphus holti
Tåning, 1918 (Family: Myctophidae). ACTA ZOOL-STOCKHOLM 2020. [DOI: 10.1111/azo.12348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mauro Cavallaro
- Department of Veterinary Sciences University of Messina Messina Italy
| | | | - Francesco Abbate
- Department of Veterinary Sciences University of Messina Messina Italy
| | | | - Rosaria Laurà
- Scienze veterinarie Facoltà degli studi di Messina Messina Italy
| | | | | | | | | |
Collapse
|
18
|
Abstract
The skeletal muscle is the largest organ in the body, by mass. It is also the regulator of glucose homeostasis, responsible for 80% of postprandial glucose uptake from the circulation. Skeletal muscle is essential for metabolism, both for its role in glucose uptake and its importance in exercise and metabolic disease. In this article, we give an overview of the importance of skeletal muscle in metabolism, describing its role in glucose uptake and the diseases that are associated with skeletal muscle metabolic dysregulation. We focus on the role of skeletal muscle in peripheral insulin resistance and the potential for skeletal muscle-targeted therapeutics to combat insulin resistance and diabetes, as well as other metabolic diseases like aging and obesity. In particular, we outline the possibilities and pitfalls of the quest for exercise mimetics, which are intended to target the molecular mechanisms underlying the beneficial effects of exercise on metabolic disease. We also provide a description of the molecular mechanisms that regulate skeletal muscle glucose uptake, including a focus on the SNARE proteins, which are essential regulators of glucose transport into the skeletal muscle. © 2020 American Physiological Society. Compr Physiol 10:785-809, 2020.
Collapse
Affiliation(s)
- Karla E. Merz
- Department of Molecular and Cellular Endocrinology, City of Hope Beckman Research Institute, Duarte, California, USA
- The Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, California, USA
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, City of Hope Beckman Research Institute, Duarte, California, USA
| |
Collapse
|
19
|
Gantner BN, LaFond KM, Bonini MG. Nitric oxide in cellular adaptation and disease. Redox Biol 2020; 34:101550. [PMID: 32438317 PMCID: PMC7235643 DOI: 10.1016/j.redox.2020.101550] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide synthases are the major sources of nitric oxide, a critical signaling molecule involved in a wide range of cellular and physiological processes. These enzymes comprise a family of genes that are highly conserved across all eukaryotes. The three family members found in mammals are important for inter- and intra-cellular signaling in tissues that include the nervous system, the vasculature, the gut, skeletal muscle, and the immune system, among others. We summarize major advances in the understanding of biochemical and tissue-specific roles of nitric oxide synthases, with a focus on how these mechanisms enable tissue adaptation and health or dysfunction and disease. We highlight the unique mechanisms and processes of neuronal nitric oxide synthase, or NOS1. This was the first of these enzymes discovered in mammals, and yet much remains to be understood about this highly conserved and complex gene. We provide examples of two areas that will likely be of increasing importance in nitric oxide biology. These include the mechanisms by which these critical enzymes promote adaptation or disease by 1) coordinating communication by diverse cell types within a tissue and 2) directing cellular differentiation/activation decisions processes.
Collapse
Affiliation(s)
- Benjamin N Gantner
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, USA.
| | - Katy M LaFond
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, USA
| | - Marcelo G Bonini
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, USA; Feinberg School of Medicine, Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, USA
| |
Collapse
|
20
|
Cocksedge SP, Breese BC, Morgan PT, Nogueira L, Thompson C, Wylie LJ, Jones AM, Bailey SJ. Influence of muscle oxygenation and nitrate-rich beetroot juice supplementation on O 2 uptake kinetics and exercise tolerance. Nitric Oxide 2020; 99:25-33. [PMID: 32272260 DOI: 10.1016/j.niox.2020.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 11/25/2022]
Abstract
We tested the hypothesis that acute supplementation with nitrate (NO3-)-rich beetroot juice (BR) would improve quadriceps muscle oxygenation, pulmonary oxygen uptake (V˙O2) kinetics and exercise tolerance (Tlim) in normoxia and that these improvements would be augmented in hypoxia and attenuated in hyperoxia. In a randomised, double-blind, cross-over study, ten healthy males completed two-step cycle tests to Tlim following acute consumption of 210 mL BR (18.6 mmol NO3-) or NO3--depleted beetroot juice placebo (PL; 0.12 mmol NO3-). These tests were completed in normobaric normoxia [fraction of inspired oxygen (FIO2): 21%], hypoxia (FIO2: 15%) and hyperoxia (FIO2: 40%). Pulmonary V˙O2 and quadriceps tissue oxygenation index (TOI), derived from multi-channel near-infrared spectroscopy, were measured during all trials. Plasma [nitrite] was higher in all BR compared to all PL trials (P < 0.05). Quadriceps TOI was higher in normoxia compared to hypoxia (P < 0.05) and higher in hyperoxia compared to hypoxia and normoxia (P < 0.05). Tlim was improved after BR compared to PL ingestion in the hypoxic trials (250 ± 44 vs. 231 ± 41 s; P = 0.006; d = 1.13), with the magnitude of improvement being negatively correlated with quadriceps TOI at Tlim (r = -0.78; P < 0.05). Tlim was not improved following BR ingestion in normoxia (BR: 364 ± 98 vs. PL: 344 ± 78 s; P = 0.087, d = 0.61) or hyperoxia (BR: 492 ± 212 vs. PL: 472 ± 196 s; P = 0.273, d = 0.37). BR ingestion increased peak V˙O2 in hypoxia (P < 0.05), but not normoxia or hyperoxia (P > 0.05). These findings indicate that BR supplementation is more likely to improve Tlim and peak V˙O2 in situations when skeletal muscle is more hypoxic.
Collapse
Affiliation(s)
- Stuart P Cocksedge
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK; School of Sport, Exercise and Health Sciences, Loughborough University, Epinal Way, Loughborough, UK
| | - Brynmor C Breese
- School of Biological and Biomedical Sciences, Portland Square Building, Plymouth University, Drake Circus, Plymouth, UK
| | - Paul T Morgan
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Leonardo Nogueira
- Section of Physiology, Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of California, San Diego, CA, USA; Instituto de Bioquímica Médica Leopoldo de Meis (Medical Biochemistry Institute Leopoldo de Meis), Federal University of Rio de Janeiro, RJ, Brazil
| | - Christopher Thompson
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Lee J Wylie
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Andrew M Jones
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Stephen J Bailey
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK; School of Sport, Exercise and Health Sciences, Loughborough University, Epinal Way, Loughborough, UK.
| |
Collapse
|
21
|
Blottner D, Hastermann M, Weber R, Lenz R, Gambara G, Limper U, Rittweger J, Bosutti A, Degens H, Salanova M. Reactive Jumps Preserve Skeletal Muscle Structure, Phenotype, and Myofiber Oxidative Capacity in Bed Rest. Front Physiol 2020; 10:1527. [PMID: 32009969 PMCID: PMC6974579 DOI: 10.3389/fphys.2019.01527] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/04/2019] [Indexed: 12/18/2022] Open
Abstract
Identification of countermeasures able to prevent disuse-induced muscle wasting is crucial to increase performance of crew members during space flight as well as ameliorate patient’s clinical outcome after long immobilization periods. We report on the outcome of short but high-impact reactive jumps (JUMP) as countermeasure during 60 days of 6° head-down tilt (HDT) bed rest on myofiber size, type composition, capillarization, and oxidative capacity in tissue biopsies (pre/post/recovery) from the knee extensor vastus lateralis (VL) and deep calf soleus (SOL) muscle of 22 healthy male participants (Reactive jumps in a sledge, RSL-study 2015–2016, DLR:envihab, Cologne). Bed rest induced a slow-to-fast myofiber shift (type I –>II) with an increased prevalence of hybrid fibers in SOL after bed rest without jumps (control, CTRL, p = 0.016). In SOL, JUMP countermeasure in bed rest prevented both fast and slow myofiber cross-sectional area (CSA) decrements (p = 0.005) in CTRL group. In VL, bed rest only induced capillary rarefaction, as reflected by the decrease in local capillary-to-fiber ratio (LCFR) for both type II (pre vs. post/R + 10, p = 0.028/0.028) and type I myofibers (pre vs. R + 10, p = 0.012), which was not seen in the JUMP group. VO2maxFiber (pL × mm–1 × min–1) calculated from succinate dehydrogenase (SDH)-stained cryosections (OD660 nm) showed no significant differences between groups. High-impact jump training in bed rest did not prevent disuse-induced myofiber atrophy in VL, mitigated phenotype transition (type I – >II) in SOL, and attenuated capillary rarefaction in the prime knee extensor VL however with little impact on oxidative capacity changes.
Collapse
Affiliation(s)
- Dieter Blottner
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institute of Integrative Neuroanatomy, Berlin, Germany.,NeuroMuscular Group, Center of Space Medicine and Extreme Environments, Berlin, Germany
| | - Maria Hastermann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institute of Integrative Neuroanatomy, Berlin, Germany.,NeuroMuscular Group, Center of Space Medicine and Extreme Environments, Berlin, Germany
| | - Robert Weber
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institute of Integrative Neuroanatomy, Berlin, Germany
| | - Regina Lenz
- Department of Movement and Neurosciences, German Sports University, Cologne, Germany
| | - Guido Gambara
- Charité Comprehensive Cancer Center, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrich Limper
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany.,Department of Anesthesiology and Intensive Care Medicine, Merheim Medical Center, Hospitals of Cologne, University of Witten/Herdecke, Cologne, Germany
| | - Jörn Rittweger
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Cologne, Germany
| | | | - Hans Degens
- Research Centre for Musculoskeletal Science & Sports Medicine, Manchester Metropolitan University, Manchester, United Kingdom.,Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Michele Salanova
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institute of Integrative Neuroanatomy, Berlin, Germany.,NeuroMuscular Group, Center of Space Medicine and Extreme Environments, Berlin, Germany
| |
Collapse
|
22
|
Kim TW, Park SS, Kim BK, Sim YJ, Shin MS. Effects of sildenafil citrate on peripheral fatigue and exercise performance after exhaustive swimming exercise in rats. J Exerc Rehabil 2019; 15:751-756. [PMID: 31938694 PMCID: PMC6944887 DOI: 10.12965/jer.1938712.356] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/20/2019] [Indexed: 12/24/2022] Open
Abstract
Sildenafil citrate is a potent and selective inhibitor of phosphodiesterase type-5 used to treat erectile dysfunction. We investigated the effects of sildenafil citrate treatment on peripheral fatigue and exercise performance after exhaustive swimming exercise in rats. The rats in the sildenafil citrate-treated groups received sildenafil citrate orally once a day for 14 consecutive days at respective dosage. On the 14 days after starting experiment, each animal was submitted to swimming test with intensity equivalent to overload. The exhaustion was defined as a state in which coordinated movements did not return to the water surface for breathing within 10 sec. Western blot for monocarboxylate transporter (MCT)1, MCT4, and neuronal nitric oxide synthase (nNOS) were performed. Exhaustive swimming exercise decreased time of exhaustion and increased lactate concentration, however, sildenafil citrate enhanced time of exhaustion and decreased lactate concentration. Exhaustive swimming exercise increased MCT1 and MCT4 expressions in the gastrocnemius muscles and sildenafil citrate further enhanced MCT1 and MCT4 expressions in the exhaustive swimming exercise rats. Exhaustive swimming exercise decreased nNOS expression in the gastrocnemius muscles and sildenafil citrate enhanced nNOS expression in the exhaustive swimming exercise rats. The most potent effect appeared in the 20-mg/kg sildenafil citrate. Sildenafil citrate might be proposed as a potential ergogenic aid through antiperipheral fatigue.
Collapse
Affiliation(s)
- Tae-Woon Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Seo Park
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Bo-Kyun Kim
- Department of Emergency Technology, College of Health Science, Gachon University, Incheon, Korea
| | - Young-Je Sim
- Department of Physical Education, Kunsan National University, Gunsan, Korea
| | - Mal-Soon Shin
- College of Culture and Sports, Division of Global Sport Studies, Korea University, Sejong, Korea
| |
Collapse
|
23
|
Tomiga Y, Ito A, Sudo M, Ando S, Eshima H, Sakai K, Nakashima S, Uehara Y, Tanaka H, Soejima H, Higaki Y. One week, but not 12 hours, of cast immobilization alters promotor DNA methylation patterns in the nNOS gene in mouse skeletal muscle. J Physiol 2019; 597:5145-5159. [PMID: 31490543 DOI: 10.1113/jp277019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 08/27/2019] [Indexed: 12/31/2022] Open
Abstract
KEY POINTS DNA methylation may play an important role in regulating gene expression in skeletal muscle to adapt to physical activity and inactivity. Neuronal nitric oxide synthase (nNOS) in skeletal muscle is a key regulator of skeletal muscle mass; however, it is unclear whether nNOS expression is regulated by DNA methylation. We found that 1 week of cast immobilization increased nNOS DNA methylation levels and downregulated nNOS gene expression in atrophic slow-twitch soleus muscle from the mouse leg. These changes were not detected in non-atrophic fast-twitch extensor digitorum longus muscle. Twelve hours of cast immobilization decreased nNOS gene expression, whereas nNOS DNA methylation levels were unchanged, suggesting that downregulation of nNOS gene expression by short-term muscle inactivity is independent of the DNA methylation pattern. These findings contribute to a better understanding of the maintenance of skeletal muscle mass and prevention of muscle atrophy by epigenetic mechanisms via the nNOS/NO pathway. ABSTRACT DNA methylation is a mechanism that controls gene expression in skeletal muscle under various environmental stimuli, such as physical activity and inactivity. Neuronal nitric oxide synthase (nNOS) regulates muscle atrophy in skeletal muscle. However, the mechanisms regulating nNOS expression in atrophic muscle remain unclear. We hypothesized that nNOS expression in atrophic muscle is regulated by DNA methylation of the nNOS promotor in soleus (Sol; slow-twitch fibre dominant) and extensor digitorum longus (EDL; fast-twitch fibre dominant) muscles. One week of cast immobilization induced significant muscle atrophy in Sol but not in EDL. We showed that 1 week of cast immobilization increased nNOS DNA methylation levels in Sol, although only a minor change was detected in EDL. Consistent with the increased DNA methylation levels in atrophic Sol, the gene expression levels of total nNOS and nNOSµ (i.e. the major splicing variant of nNOS in skeletal muscle) decreased. The abundance of the nNOS protein and cell membrane (especially type IIa fibre) immunoreactivity also decreased in atrophic Sol. These changes were not observed in EDL after 1 week of cast immobilization. Furthermore, despite the lack of significant atrophy, 12 h of cast immobilization decreased gene expression levels of total nNOS and nNOSµ in Sol. However, no association was detected between nNOS DNA methylation and gene expression. The expression of the nNOSβ gene, another splicing variant of nNOS, in EDL was unchanged by cast immobilization, whereas its expression was not detected in Sol. We concluded that chronic adaptation of nNOS gene expression in cast immobilized muscle may involve nNOS DNA methylation.
Collapse
Affiliation(s)
- Yuki Tomiga
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan.,The Fukuoka University Institute for Physical Activity, Fukuoka, Japan
| | - Ai Ito
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Mizuki Sudo
- Physical Fitness Research Institute Meiji Yasuda Life Foundation of Health and Welfare, Tokyo, Japan
| | - Soichi Ando
- Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan
| | - Hiroaki Eshima
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan.,Diabetes and Metabolism Research Centre, Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
| | - Kazuya Sakai
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Shihoko Nakashima
- The Fukuoka University Institute for Physical Activity, Fukuoka, Japan.,Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Yoshinari Uehara
- The Fukuoka University Institute for Physical Activity, Fukuoka, Japan.,Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Hiroaki Tanaka
- The Fukuoka University Institute for Physical Activity, Fukuoka, Japan.,Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Hidenobu Soejima
- Division of Molecular Genetics and Epigenetics, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, Japan
| | - Yasuki Higaki
- The Fukuoka University Institute for Physical Activity, Fukuoka, Japan.,Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
24
|
Oddoux S, Randazzo D, Kenea A, Alonso B, Zaal KJM, Ralston E. Misplaced Golgi Elements Produce Randomly Oriented Microtubules and Aberrant Cortical Arrays of Microtubules in Dystrophic Skeletal Muscle Fibers. Front Cell Dev Biol 2019; 7:176. [PMID: 31620435 PMCID: PMC6759837 DOI: 10.3389/fcell.2019.00176] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022] Open
Abstract
Differentiated mammalian cells and tissues, such as skeletal muscle fibers, acquire an organization of Golgi complex and microtubules profoundly different from that in proliferating cells and still poorly understood. In adult rodent skeletal muscle, the multinucleated muscle fibers have hundreds of Golgi elements (GE), small stacks of cisternae that serve as microtubule-organizing centers. We are interested in the role of the GE in organizing a peculiar grid of microtubules located in the fiber cortex, against the sarcolemma. Modifications of this grid in the mdx mouse model of Duchenne muscular dystrophy have led to identifying dystrophin, the protein missing in both human disease and mouse model, as a microtubule guide. Compared to wild-type (WT), mdx microtubules are disordered and more dense and they have been linked to the dystrophic pathology. GE themselves are disordered in mdx. Here, to identify the causes of GE and microtubule alterations in the mdx muscle, we follow GFP-tagged microtubule markers in live mdx fibers and investigate the recovery of GE and microtubules after treatment with nocodazole. We find that mdx microtubules grow 10% faster but in 30% shorter bouts and that they begin to form a tangled network, rather than an orthogonal grid, right after nucleation from GE. Strikingly, a large fraction of microtubules in mdx muscle fibers seem to dissociate from GE after nucleation. Moreover, we report that mdx GE are mispositioned and increased in number and size. These results were replicated in WT fibers overexpressing the beta-tubulin tubb6, which is elevated in Duchenne muscular dystrophy, in mdx and in regenerating muscle. Finally, we examine the association of GE with ER exit sites and ER-to-Golgi intermediate compartment, which starts during muscle differentiation, and find it persisting in mdx and tubb6 overexpressing fibers. We conclude that GE are full, small, Golgi complexes anchored, and positioned through ER Exit Sites. We propose a model in which GE mispositioning, together with the absence of microtubule guidance due to the lack of dystrophin, determines the differences in GE and microtubule organization between WT and mdx muscle fibers.
Collapse
Affiliation(s)
- Sarah Oddoux
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Davide Randazzo
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Aster Kenea
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Bruno Alonso
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kristien J M Zaal
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Evelyn Ralston
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
25
|
Kim MJ, Whitehead NP, Bible KL, Adams ME, Froehner SC. Mice lacking α-, β1- and β2-syntrophins exhibit diminished function and reduced dystrophin expression in both cardiac and skeletal muscle. Hum Mol Genet 2019; 28:386-395. [PMID: 30256963 DOI: 10.1093/hmg/ddy341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/21/2018] [Indexed: 01/16/2023] Open
Abstract
Syntrophins are a family of modular adaptor proteins that are part of the dystrophin protein complex, where they recruit and anchor a variety of signaling proteins. Previously we generated mice lacking α- and/or β2-syntrophin but showed that in the absence of one isoform, other syntrophin isoforms can partially compensate. Therefore, in the current study, we generated mice that lacked α, β1 and β2-syntrophins [triple syntrophin knockout (tKO) mice] and assessed skeletal and cardiac muscle function. The tKO mice showed a profound reduction in voluntary wheel running activity at both 6 and 12 months of age. Function of the tibialis anterior was assessed in situ and we found that the specific force of tKO muscle was decreased by 20-25% compared with wild-type mice. This decrease was accompanied by a shift in fiber-type composition from fast 2B to more oxidative fast 2A fibers. Using echocardiography to measure cardiac function, it was revealed that tKO hearts had left ventricular cardiac dysfunction and were hypertrophic, with a thicker left ventricular posterior wall. Interestingly, we also found that membrane-localized dystrophin expression was lower in both skeletal and cardiac muscles of tKO mice. Since dystrophin mRNA levels were not different in tKO, this finding suggests that syntrophins may regulate dystrophin trafficking to, or stabilization at, the sarcolemma. These results show that the loss of all three major muscle syntrophins has a profound effect on exercise performance, and skeletal and cardiac muscle dysfunction contributes to this deficiency.
Collapse
Affiliation(s)
- Min Jeong Kim
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Nicholas P Whitehead
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Kenneth L Bible
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Marvin E Adams
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Stanley C Froehner
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Zhao J, Yang HT, Wasala L, Zhang K, Yue Y, Duan D, Lai Y. Dystrophin R16/17 protein therapy restores sarcolemmal nNOS in trans and improves muscle perfusion and function. Mol Med 2019; 25:31. [PMID: 31266455 PMCID: PMC6607532 DOI: 10.1186/s10020-019-0101-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/20/2019] [Indexed: 01/08/2023] Open
Abstract
Background Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma leads to functional muscle ischemia. This contributes to the pathogenesis in cachexia, aging and muscular dystrophy. Mutations in the gene encoding dystrophin result in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). In many BMD patients and DMD patients that have been converted to BMD by gene therapy, sarcolemmal nNOS is missing due to the lack of dystrophin nNOS-binding domain. Methods Dystrophin spectrin-like repeats 16 and 17 (R16/17) is the sarcolemmal nNOS localization domain. Here we explored whether R16/17 protein therapy can restore nNOS to the sarcolemma and prevent functional ischemia in transgenic mice which expressed an R16/17-deleted human micro-dystrophin gene in the dystrophic muscle. The palmitoylated R16/17.GFP fusion protein was conjugated to various cell-penetrating peptides and produced in the baculovirus-insect cell system. The best fusion protein was delivered to the transgenic mice and functional muscle ischemia was quantified. Results Among five candidate cell-penetrating peptides, the mutant HIV trans-acting activator of transcription (TAT) protein transduction domain (mTAT) was the best in transferring the R16/17.GFP protein to the muscle. Systemic delivery of the mTAT.R16/17.GFP protein to micro-dystrophin transgenic mice successfully restored sarcolemmal nNOS without inducing T cell infiltration. More importantly, R16/17 protein therapy effectively prevented treadmill challenge-induced force loss and improved muscle perfusion during contraction. Conclusions Our results suggest that R16/17 protein delivery is a highly promising therapy for muscle diseases involving sarcolemmal nNOS delocalizaton. Electronic supplementary material The online version of this article (10.1186/s10020-019-0101-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Hsiao Tung Yang
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Lakmini Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA. .,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Bioengineering, University of Missouri, Columbia, MO, 65212, USA.
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA.
| |
Collapse
|
27
|
Abstract
Nitric oxide (NO) plays a plethora of important roles in the human body. Insufficient production of NO (for example, during older age and in various disease conditions) can adversely impact health and physical performance. In addition to its endogenous production through the oxidation of l-arginine, NO can be formed nonenzymatically via the reduction of nitrate and nitrite, and the storage of these anions can be augmented by the consumption of nitrate-rich foodstuffs such as green leafy vegetables. Recent studies indicate that dietary nitrate supplementation, administered most commonly in the form of beetroot juice, can ( a) improve muscle efficiency by reducing the O2 cost of submaximal exercise and thereby improve endurance exercise performance and ( b) enhance skeletal muscle contractile function and thereby improve muscle power and sprint exercise performance. This review describes the physiological mechanisms potentially responsible for these effects, outlines the circumstances in which ergogenic effects are most likely to be evident, and discusses the effects of dietary nitrate supplementation on physical performance in a range of human populations.
Collapse
Affiliation(s)
- Andrew M Jones
- Department of Sport and Health Sciences, University of Exeter, Exeter EX1 2LU, United Kingdom;
| | - Christopher Thompson
- Department of Sport and Health Sciences, University of Exeter, Exeter EX1 2LU, United Kingdom;
| | - Lee J Wylie
- Department of Sport and Health Sciences, University of Exeter, Exeter EX1 2LU, United Kingdom;
| | - Anni Vanhatalo
- Department of Sport and Health Sciences, University of Exeter, Exeter EX1 2LU, United Kingdom;
| |
Collapse
|
28
|
Randazzo D, Khalique U, Belanto JJ, Kenea A, Talsness DM, Olthoff JT, Tran MD, Zaal KJ, Pak K, Pinal-Fernandez I, Mammen AL, Sackett D, Ervasti JM, Ralston E. Persistent upregulation of the β-tubulin tubb6, linked to muscle regeneration, is a source of microtubule disorganization in dystrophic muscle. Hum Mol Genet 2019; 28:1117-1135. [PMID: 30535187 DOI: 10.1093/hmg/ddy418] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/26/2018] [Accepted: 12/02/2018] [Indexed: 12/20/2022] Open
Abstract
In healthy adult skeletal muscle fibers microtubules form a three-dimensional grid-like network. In the mdx mouse, a model of Duchenne muscular dystrophy (DMD), microtubules are mostly disordered, without periodicity. These microtubule defects have been linked to the mdx mouse pathology. We now report that increased expression of the beta 6 class V β-tubulin (tubb6) contributes to the microtubule changes of mdx muscles. Wild-type muscle fibers overexpressing green fluorescent protein (GFP)-tubb6 (but not GFP-tubb5) have disorganized microtubules whereas mdx muscle fibers depleted of tubb6 (but not of tubb5) normalize their microtubules, suggesting that increasing tubb6 is toxic. However, tubb6 increases spontaneously during differentiation of mouse and human muscle cultures. Furthermore, endogenous tubb6 is not uniformly expressed in mdx muscles but is selectively increased in fiber clusters, which we identify as regenerating. Similarly, mdx-based rescued transgenic mice that retain a higher than expected tubb6 level show focal expression of tubb6 in subsets of fibers. Tubb6 is also upregulated in cardiotoxin-induced mouse muscle regeneration, in human myositis and DMD biopsies, and the tubb6 level correlates with that of embryonic myosin heavy chain, a regeneration marker. In conclusion, modulation of a β-tubulin isotype plays a role in muscle differentiation and regeneration. Increased tubb6 expression and microtubule reorganization are not pathological per se but reflect a return to an earlier developmental stage. However, chronic elevation of tubb6, as occurs in the mdx mouse, may contribute to the repeated cycles of regeneration and to the pathology of the disease.
Collapse
Affiliation(s)
- Davide Randazzo
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Umara Khalique
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Joseph J Belanto
- Department of Biochemistry, Molecular Biology, and Biophysics, and Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Aster Kenea
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Dana M Talsness
- Department of Biochemistry, Molecular Biology, and Biophysics, and Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - John T Olthoff
- Department of Biochemistry, Molecular Biology, and Biophysics, and Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Michelle D Tran
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kristien J Zaal
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Katherine Pak
- Laboratory of Muscle Stem Cells and Gene Regulation, Muscle Disease Unit, NIAMS, NIH, Bethesda, MD, USA
| | - Iago Pinal-Fernandez
- Laboratory of Muscle Stem Cells and Gene Regulation, Muscle Disease Unit, NIAMS, NIH, Bethesda, MD, USA.,Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew L Mammen
- Laboratory of Muscle Stem Cells and Gene Regulation, Muscle Disease Unit, NIAMS, NIH, Bethesda, MD, USA.,Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan Sackett
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, and Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Evelyn Ralston
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
29
|
Normal increases in insulin-stimulated glucose uptake after ex vivo contraction in neuronal nitric oxide synthase mu (nNOSμ) knockout mice. Pflugers Arch 2019; 471:961-969. [DOI: 10.1007/s00424-019-02268-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 11/25/2022]
|
30
|
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: 2.9] [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
|
31
|
Patel A, Zhao J, Yue Y, Zhang K, Duan D, Lai Y. Dystrophin R16/17-syntrophin PDZ fusion protein restores sarcolemmal nNOSμ. Skelet Muscle 2018; 8:36. [PMID: 30466494 PMCID: PMC6251231 DOI: 10.1186/s13395-018-0182-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022] Open
Abstract
Background Loss of sarcolemmal nNOSμ is a common manifestation in a wide variety of muscle diseases and contributes to the dysregulation of multiple muscle activities. Given the critical role sarcolemmal nNOSμ plays in muscle, restoration of sarcolemmal nNOSμ should be considered as an important therapeutic goal. Methods nNOSμ is anchored to the sarcolemma by dystrophin spectrin-like repeats 16 and 17 (R16/17) and the syntrophin PDZ domain (Syn PDZ). To develop a strategy that can independently restore sarcolemmal nNOSμ, we engineered an R16/17-Syn PDZ fusion construct and tested whether this construct alone is sufficient to anchor nNOSμ to the sarcolemma in three different mouse models of Duchenne muscular dystrophy (DMD). Results Membrane-associated nNOSμ is completely lost in DMD. Adeno-associated virus (AAV)-mediated delivery of the R16/17-Syn PDZ fusion construct successfully restored sarcolemmal nNOSμ in all three models. Further, nNOS restoration was independent of the dystrophin-associated protein complex. Conclusions Our results suggest that the R16/17-Syn PDZ fusion construct is sufficient to restore sarcolemmal nNOSμ in the dystrophin-null muscle. Electronic supplementary material The online version of this article (10.1186/s13395-018-0182-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aman Patel
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA. .,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Department of Bioengineering, University of Missouri, Columbia, MO, 65212, USA.
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA.
| |
Collapse
|
32
|
Wang L, Almeida LEF, Kamimura S, van der Meulen JH, Nagaraju K, Quezado M, Wakim P, Quezado ZMN. The role of nitrite in muscle function, susceptibility to contraction injury, and fatigability in sickle cell mice. Nitric Oxide 2018; 80:70-81. [PMID: 30114530 PMCID: PMC6186197 DOI: 10.1016/j.niox.2018.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 07/05/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022]
Abstract
Sickle cell disease (SCD) patients can have limited exercise capacity and muscle dysfunction characterized by decreased force, atrophy, microvascular abnormalities, fiber distribution changes, and skeletal muscle energetics abnormalities. Growing evidence suggests that in SCD there is alteration in nitric oxide (NO) availability/signaling and that nitrate/nitrite can serve as a NO reservoir and enhance muscle performance. Here, we examined effects of nitrite on muscle strength, exercise capacity, and on contractile properties of fast-(extensor digitorum longus, EDL) and slow-twitch (soleus) muscles in SCD mice. Compared to controls, homozygotes (sickling) had decreased grip strength, impaired wheel running performance, and decreased muscle mass of fast-twitch, but not slow-twitch muscle. Nitrite treatment yielded increases in nitrite plasma levels in controls, heterozygotes, and homozygotes but decreases in muscle nitrite levels in heterozygotes and homozygotes. Regardless of genotype, nitrite yielded increases in grip strength, which were coupled with increases in specific force in EDL, but not in soleus muscle. Further, nitrite increased EDL, but not soleus, fatigability in all genotypes. Conversely, in controls, nitrite decreased, whereas in homozygotes, it increased EDL susceptibility to contraction-induced injury. Interestingly, nitrite yielded no changes in distances ran on the running wheel. These differential effects of nitrite in fast- and slow-twitch muscles suggest that its ergogenic effects would be observed in high-intensity/short exercises as found with grip force increases but no changes on wheel running distances. Further, the differential effects of nitrite in homozygotes and control animals suggests that sickling mice, which have altered NO availability/signaling, handle nitrite differently than do control animals.
Collapse
Affiliation(s)
- Li Wang
- The Sheikh Zayed Institute for Pediatric Surgical Innovation and Center for Neuroscience Research, Children's Research Institute, Washington, DC, 20010, USA
| | - Luis E F Almeida
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sayuri Kamimura
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jack H van der Meulen
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Health System, Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20010, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Health System, Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20010, USA
| | - Martha Quezado
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Paul Wakim
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health Clinical Center, Bethesda, MD, 20892, USA
| | - Zenaide M N Quezado
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
33
|
Lechado I Terradas A, Vitadello M, Traini L, Namuduri AV, Gastaldello S, Gorza L. Sarcolemmal loss of active nNOS (Nos1) is an oxidative stress-dependent, early event driving disuse atrophy. J Pathol 2018; 246:433-446. [PMID: 30066461 DOI: 10.1002/path.5149] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/28/2018] [Accepted: 07/27/2018] [Indexed: 01/08/2023]
Abstract
Skeletal muscle atrophy following unloading or immobilization represents a major invalidating event in bedridden patients. Among mechanisms involved in atrophy development, a controversial role is played by neuronal NOS (nNOS; NOS1), whose dysregulation at the protein level and/or subcellular distribution also characterizes other neuromuscular disorders. This study aimed to investigate unloading-induced changes in nNOS before any evidence of myofiber atrophy, using vastus lateralis biopsies obtained from young healthy subjects after a short bed-rest and rat soleus muscles after exposure to short unloading periods. Our results showed that (1) changes in nNOS subcellular distribution using NADPH-diaphorase histochemistry to detect enzyme activity were observed earlier than using immunofluorescence to visualize the protein; (2) loss of active nNOS from the physiological subsarcolemmal localization occurred before myofiber atrophy, i.e. in 8-day bed-rest biopsies and in 6 h-unloaded rat soleus, and was accompanied by increased nNOS activity in the sarcoplasm; (3) nNOS (Nos1) transcript and protein levels decreased significantly in the rat soleus after 6 h and 1 day unloading, respectively, to return to ambulatory levels after 4 and 7 days of unloading, respectively; (4) unloading-induced nNOS redistribution appeared dependent on mitochondrial-derived oxidant species, indirectly measured by tropomyosin disulfide bonds which had increased significantly in the rat soleus already after a 6 h-unloading bout; (5) activity of displaced nNOS molecules is required for translocation of the FoxO3 transcription factor to myofiber nuclei. FoxO3 nuclear localization in rat soleus increased after 6 h unloading (about four-fold the ambulatory level), whereas it did not when nNOS expression and activity were inhibited in vivo before and during 6 h unloading. In conclusion, this study demonstrates that the redistribution of active nNOS molecules from sarcolemma to sarcoplasm not only is ahead of the atrophy of unloaded myofibers, and is induced by increased production of mitochondrial superoxide anion, but also drives FoxO3 activation to initiate muscle atrophy. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | | | - Leonardo Traini
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | | | - Stefano Gastaldello
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden.,Precision Medicine Research Center (Department), Binzhou Medical University, Shandong Province, Yantai, PR China
| | - Luisa Gorza
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
34
|
Percival JM. Perspective: Spectrin-Like Repeats in Dystrophin Have Unique Binding Preferences for Syntrophin Adaptors That Explain the Mystery of How nNOSμ Localizes to the Sarcolemma. Front Physiol 2018; 9:1369. [PMID: 30349485 PMCID: PMC6186803 DOI: 10.3389/fphys.2018.01369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/10/2018] [Indexed: 11/13/2022] Open
Abstract
Dystrophin is a massive multi-domain protein composed of specialized amino and carboxyl termini that are separated by 24 spectrin-like repeats. Dystrophin performs critical structural and signaling roles that are indispensable for the functional integrity of skeletal muscle. Indeed, the loss of dystrophin protein expression causes the muscle wasting disease, Duchenne muscular dystrophy (DMD). Substantial progress has been made in defining the functions of the domains of dystrophin, which has proven invaluable for the development of miniaturized dystrophin gene and exon skipping therapies for DMD. However, a long-standing mystery regarding dystrophin function is how dystrophin, and its adaptor and neuronal nitric oxide synthase mu (nNOSμ) binding partner α-syntrophin, cooperate to localize nNOSμ to the sarcolemma. Only when localized to the sarcolemma can nNOSμ override sympathetic vasoconstriction and prevent functional ischemia in contracting muscles. Current evidence suggests that spectrin-like repeat 17 of dystrophin and α-syntrophin cooperate to localize nNOSμ to the sarcolemma. However, the exact mechanism remains unclear and controversial because of equivocal evidence for direct binding of dystrophin and nNOSμ. Recently, an important study identified a novel α-syntrophin binding site within spectrin-like repeat 17, leading to a new model whereby α-syntrophin recruits nNOSμ to the sarcolemmal dystrophin complex by binding spectrin-like repeat 17. This model finally appears to solve the mystery of the dual requirement for dystrophin and α-syntrophin for sarcolemmal nNOSμ localization. The aim of the current perspective is to highlight this major advance in understanding of dystrophin’s role in localizing nNOSμ and its implications for current trials.
Collapse
Affiliation(s)
- Justin M Percival
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
35
|
Dombernowsky NW, Ölmestig JNE, Witting N, Kruuse C. Role of neuronal nitric oxide synthase (nNOS) in Duchenne and Becker muscular dystrophies - Still a possible treatment modality? Neuromuscul Disord 2018; 28:914-926. [PMID: 30352768 DOI: 10.1016/j.nmd.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/07/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023]
Abstract
Neuronal nitric oxide synthase (nNOS) is involved in nitric oxide (NO) production and suggested to play a crucial role in blood flow regulation of skeletal muscle. During activation of the muscle, NO helps attenuate the sympathetic vasoconstriction to accommodate increased metabolic demands, a phenomenon known as functional sympatholysis. In inherited myopathies such as the dystrophinopathies Duchenne and Becker muscle dystrophies (DMD and BMD), nNOS is lost from the sarcolemma. The loss of nNOS may cause functional ischemia contributing to skeletal and cardiac muscle cell injury. Effects of NO is augmented by inhibiting degradation of the second messenger cyclic guanosine monophosphate (cGMP) using sildenafil and tadalafil, both of which inhibit the enzyme phosphodiesterase 5 (PDE5). In animal models of DMD, PDE5-inhibitors prevent functional ischemia, reduce post-exercise skeletal muscle pathology and fatigue, show amelioration of cardiac muscle cell damage and increase cardiac performance. However, effect on clinical outcomes in DMD and BMD patients have been disappointing with minor effects on upper limb performance and none on ambulation. This review aims to summarize the current knowledge of nNOS function related to functional sympatholysis in skeletal muscle and studies on PDE5-inhibitor treatment in nNOS-deficient animal models and patients.
Collapse
Affiliation(s)
- Nanna W Dombernowsky
- Department of Neurology, Rigshospitalet Glostrup, University of Copenhagen, Denmark
| | - Joakim N E Ölmestig
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, University of Copenhagen, Denmark
| | - Nanna Witting
- Department of Neurology, Rigshospitalet Glostrup, University of Copenhagen, Denmark
| | - Christina Kruuse
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, University of Copenhagen, Denmark; PDE Research Group, Lundbeck Foundation Center for Neurovascular Research (LUCENS), Denmark.
| |
Collapse
|
36
|
Kerrick WGL, Xu Y, Percival JM. nNOS splice variants differentially regulate myofilament function but are dispensable for intracellular calcium and force transients in cardiac papillary muscles. PLoS One 2018; 13:e0200834. [PMID: 30028847 PMCID: PMC6054407 DOI: 10.1371/journal.pone.0200834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/02/2018] [Indexed: 11/18/2022] Open
Abstract
Cardiac muscle expresses three neuronal nitric oxide synthase (nNOS) splice variants: nNOSα, nNOSμ and nNOSβ. The functions of these nNOS splice variants in cardiac muscle, particularly myofilament-associated nNOSβ are unclear. To decipher cardiac nNOS splice variant function we investigated myofilament function and intracellular calcium and force transients in demembranated and intact papillary muscles from two lines of nNOS knockout mice. The first line (KN1) lacks nNOSα and nNOSμ. The second line (KN2) lacks active nNOSα, nNOSμ and nNOSβ. Demembranated KN1 papillary muscles exhibited reduced myofilament ATPase activity (-35%) and specific force (-10%) relative to controls. Demembranated KN2 muscles exhibited a smaller decrease in myofilament ATPase activity (-21%), but a greater reduction in specific force (-26%) relative to controls. Myofilament calcium sensitivity in demembranated KN1 and KN2 papillary muscles was similar to controls. Thus, papillary muscle-expressed nNOS splice variants are necessary for control levels of myofilament ATPase activity and force generation, but dispensable for myofilament calcium sensitivity. The greater reduction in myofilament ATPase relative to specific force in KN1, but not KN2 muscle, reduced the energy cost of muscle contraction, suggesting that nNOSβ increased the energetic efficiency of contraction in the absence of nNOSμ and nNOSα. Analyses of intact KN1 and KN2 papillary muscles showed that both intracellular calcium transients and their evoked force transients were similar to controls at stimulation frequencies between 1 and 3 Hz. Therefore, nNOS was dispensable for baseline excitation-contraction coupling. In summary, these data suggest that nNOS splice variants differentially regulate myofilament function, but not baseline calcium handling in papillary muscles. More importantly, they suggest that nNOSβ is a novel modulator of myofilament function, and ultimately the energetic efficiency of cardiac papillary muscle contraction.
Collapse
Affiliation(s)
- W Glenn L Kerrick
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Yuanyuan Xu
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Justin M Percival
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| |
Collapse
|
37
|
Guilbaud M, Gentil C, Peccate C, Gargaun E, Holtzmann I, Gruszczynski C, Falcone S, Mamchaoui K, Ben Yaou R, Leturcq F, Jeanson-Leh L, Piétri-Rouxel F. miR-708-5p and miR-34c-5p are involved in nNOS regulation in dystrophic context. Skelet Muscle 2018; 8:15. [PMID: 29703249 PMCID: PMC5924477 DOI: 10.1186/s13395-018-0161-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/30/2022] Open
Abstract
Background Duchenne (DMD) and Becker (BMD) muscular dystrophies are caused by mutations in the DMD gene coding for dystrophin, a protein being part of a large sarcolemmal protein scaffold that includes the neuronal nitric oxide synthase (nNOS). The nNOS was shown to play critical roles in a variety of muscle functions and alterations of its expression and location in dystrophic muscle fiber leads to an increase of the muscle fatigability. We previously revealed a decrease of nNOS expression in BMD patients all presenting a deletion of exons 45 to 55 in the DMD gene (BMDd45-55), impacting the nNOS binding site of dystrophin. Since several studies showed deregulation of microRNAs (miRNAs) in dystrophinopathies, we focused on miRNAs that could target nNOS in dystrophic context. Methods By a screening of 617 miRNAs in BMDd45-55 muscular biopsies using TLDA and an in silico study to determine which one could target nNOS, we selected four miRNAs. In order to select those that targeted a sequence of 3′UTR of NOS1, we performed luciferase gene reporter assay in HEK393T cells. Finally, expression of candidate miRNAs was modulated in control and DMD human myoblasts (DMDd45-52) to study their ability to target nNOS. Results TLDA assay and the in silico study allowed us to select four miRNAs overexpressed in muscle biopsies of BMDd45-55 compared to controls. Among them, only the overexpression of miR-31, miR-708, and miR-34c led to a decrease of luciferase activity in an NOS1-3′UTR-luciferase assay, confirming their interaction with the NOS1-3′UTR. The effect of these three miRNAs was investigated on control and DMDd45-52 myoblasts. First, we showed a decrease of nNOS expression when miR-708 or miR-34c were overexpressed in control myoblasts. We then confirmed that DMDd45-52 cells displayed an endogenous increased of miR-31, miR-708, and miR-34c and a decreased of nNOS expression, the same characteristics observed in BMDd45-55 biopsies. In DMDd45-52 cells, we demonstrated that the inhibition of miR-708 and miR-34c increased nNOS expression, confirming that both miRNAs can modulate nNOS expression in human myoblasts. Conclusion These results strongly suggest that miR-708 and miR-34c, overexpressed in dystrophic context, are new actors involved in the regulation of nNOS expression in dystrophic muscle. Electronic supplementary material The online version of this article (10.1186/s13395-018-0161-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marine Guilbaud
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Christel Gentil
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Cécile Peccate
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Elena Gargaun
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Isabelle Holtzmann
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Carole Gruszczynski
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Sestina Falcone
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Kamel Mamchaoui
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Rabah Ben Yaou
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France.,AP-HP, Centre de Référence Maladies Neuromusculaire Nord, Est, Ile-de-France, G.H. Pitié-Salpêtrière, F-75013, Paris, France
| | - France Leturcq
- Laboratoire de Génétique et Biologie Moléculaire, Hôpital Cochin, Paris, France
| | | | - France Piétri-Rouxel
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France.
| |
Collapse
|
38
|
Relation of nNOS isoforms to mitochondrial density and PGC-1alpha expression in striated muscles of mice. Nitric Oxide 2018; 77:35-43. [PMID: 29678764 DOI: 10.1016/j.niox.2018.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 04/10/2018] [Accepted: 04/16/2018] [Indexed: 01/16/2023]
Abstract
The expression of neuronal NO synthase (nNOS) alpha- and beta-isoforms in skeletal muscle is well documented but only little information is available about their regulation/functions. Using different mouse models, we now assessed whether the expression of nNOS-isoforms in muscle fibers is related to mitochondria content/activity and regulated by peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha). Catalytic histochemistry revealed highest nNOS-concentrations to be present in type-2 oxidative muscle fibers. Differences in mitochondrial density between nNOS-KO-mice and WT-littermates established by morphometry after transmission electron microscopy were significant in the oxidative portion of the tibialis anterior muscle (TA) but not in rectus femoris muscle (RF) indicating an nNOS-dependent mitochondrial pool in TA. Quantitative immunoblotting displayed the nNOS alpha-isoform to preponderate in those striated muscles of C57BL/6-mice that comprise of many type-2 oxidative fibers, e.g. TA, while roughly even levels of the two nNOS-isoforms were expressed in those muscles that mainly consist of type-2 glycolytic fibers, e.g. RF. Differences in citrate synthase-activity in muscle homogenates between nNOS-KO-mice and WT-littermates were positively related to nNOS alpha-isoform levels. In transgenic-mice over-expressing muscular PGC-1alpha compared to WT-littermates, immunoblotting revealed a significant shift in nNOS-expression in favor of the alpha-isoform in six out of eight striated muscles (exceptions: soleus muscle and tongue) without consistent relationship to changes in the expression of mitochondrial markers. In summary, our study demonstrated the nNOS alpha-isoform expression to be related to mitochondrial content/activity and to be up-regulated by up-stream PGC-1alpha in striated muscles, particularly in those enriched with type-2 oxidative fibers implying a functional convergence of the two signaling systems in these fibers.
Collapse
|
39
|
Zhang X, Hiam D, Hong YH, Zulli A, Hayes A, Rattigan S, McConell GK. Nitric oxide is required for the insulin sensitizing effects of contraction in mouse skeletal muscle. J Physiol 2017; 595:7427-7439. [PMID: 29071734 DOI: 10.1113/jp275133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/18/2017] [Indexed: 01/15/2023] Open
Abstract
KEY POINTS People with insulin resistance or type 2 diabetes can substantially increase their skeletal muscle glucose uptake during exercise and insulin sensitivity after exercise. Skeletal muscle nitric oxide (NO) is important for glucose uptake during exercise, although how prior exercise increases insulin sensitivity is unclear. In the present study, we examined whether NO is necessary for normal increases in skeletal muscle insulin sensitivity after contraction ex vivo in mouse muscle. The present study uncovers, for the first time, a novel role for NO in the insulin sensitizing effects of ex vivo contraction, which is independent of blood flow. ABSTRACT The factors regulating the increase in skeletal muscle insulin sensitivity after exercise are unclear. We examined whether nitric oxide (NO) is required for the increase in insulin sensitivity after ex vivo contractions. Isolated C57BL/6J mouse EDL muscles were contracted for 10 min or remained at rest (basal) with or without the NO synthase (NOS) inhibition (NG -monomethyl-l-arginine; l-NMMA; 100 μm). Then, 3.5 h post contraction/basal, muscles were exposed to saline or insulin (120 μU ml-1 ) with or without l-NMMA during the last 30 min. l-NMMA had no effect on basal skeletal muscle glucose uptake. The increase in muscle glucose uptake with insulin (57%) was significantly (P < 0.05) greater after prior contraction (140% increase). NOS inhibition during the contractions had no effect on this insulin-sensitizing effect of contraction, whereas NOS inhibition during insulin prevented the increase in skeletal muscle insulin sensitivity post-contraction. Soluble guanylate cyclase inhibition, protein kinase G (PKG) inhibition or cyclic nucleotide phosphodiesterase inhibition each had no effect on the insulin-sensitizing effect of prior contraction. In conclusion, NO is required for increases in insulin sensitivity several hours after contraction of mouse skeletal muscle via a cGMP/PKG independent pathway.
Collapse
Affiliation(s)
- Xinmei Zhang
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia.,School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Danielle Hiam
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia
| | - Yet-Hoi Hong
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia
| | - Anthony Zulli
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Alan Hayes
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia.,College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Stephen Rattigan
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Glenn K McConell
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia.,College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| |
Collapse
|
40
|
|
41
|
Kellogg DL, McCammon KM, Hinchee-Rodriguez KS, Adamo ML, Roman LJ. Neuronal nitric oxide synthase mediates insulin- and oxidative stress-induced glucose uptake in skeletal muscle myotubes. Free Radic Biol Med 2017; 110:261-269. [PMID: 28666850 PMCID: PMC5554434 DOI: 10.1016/j.freeradbiomed.2017.06.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/13/2017] [Accepted: 06/26/2017] [Indexed: 10/19/2022]
Abstract
Previously published studies strongly suggested that insulin- and exercise-induced skeletal muscle glucose uptake require nitric oxide (NO) production. However, the signal transduction mechanisms by which insulin and contraction regulated NO production and subsequent glucose transport are not known. In the present study, we utilized the myotube cell lines treated with insulin or hydrogen peroxide, the latter to mimic contraction-induced oxidative stress, to characterize these mechanisms. We found that insulin stimulation of neuronal nitric oxide synthase (nNOS) phosphorylation, NO production, and GLUT4 translocation were all significantly reduced by inhibition of either nNOS or Akt2. Hydrogen peroxide (H2O2) induced phosphorylation of nNOS at the same residue as did insulin, and also stimulated NO production and GLUT4 translocation. nNOS inhibition prevented H2O2-induced GLUT4 translocation. AMP activated protein kinase (AMPK) inhibition prevented H2O2 activation and phosphorylation of nNOS, leading to reduced NO production and significantly attenuated GLUT4 translocation. We conclude that nNOS phosphorylation and subsequently increased NO production are required for both insulin- and H2O2-stimulated glucose transport. Although the two stimuli result in phosphorylation of the same residue on nNOS, they do so through distinct protein kinases. Thus, insulin and H2O2-activated signaling pathways converge on nNOS, which is a common mediator of glucose uptake in both pathways. However, the fact that different kinases are utilized provides a basis for the use of exercise to activate glucose transport in the face of insulin resistance.
Collapse
Affiliation(s)
- Dean L Kellogg
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, United States
| | - Karen M McCammon
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, United States
| | - Kathryn S Hinchee-Rodriguez
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, United States
| | - Martin L Adamo
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, United States
| | - Linda J Roman
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, United States.
| |
Collapse
|
42
|
Moon Y, Balke JE, Madorma D, Siegel MP, Knowels G, Brouckaert P, Buys ES, Marcinek DJ, Percival JM. Nitric Oxide Regulates Skeletal Muscle Fatigue, Fiber Type, Microtubule Organization, and Mitochondrial ATP Synthesis Efficiency Through cGMP-Dependent Mechanisms. Antioxid Redox Signal 2017; 26:966-985. [PMID: 27393340 PMCID: PMC5467110 DOI: 10.1089/ars.2016.6630] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIM Skeletal muscle nitric oxide-cyclic guanosine monophosphate (NO-cGMP) pathways are impaired in Duchenne and Becker muscular dystrophy partly because of reduced nNOSμ and soluble guanylate cyclase (GC) activity. However, GC function and the consequences of reduced GC activity in skeletal muscle are unknown. In this study, we explore the functions of GC and NO-cGMP signaling in skeletal muscle. RESULTS GC1, but not GC2, expression was higher in oxidative than glycolytic muscles. GC1 was found in a complex with nNOSμ and targeted to nNOS compartments at the Golgi complex and neuromuscular junction. Baseline GC activity and GC agonist responsiveness was reduced in the absence of nNOS. Structural analyses revealed aberrant microtubule directionality in GC1-/- muscle. Functional analyses of GC1-/- muscles revealed reduced fatigue resistance and postexercise force recovery that were not due to shifts in type IIA-IIX fiber balance. Force deficits in GC1-/- muscles were also not driven by defects in resting mitochondrial adenosine triphosphate (ATP) synthesis. However, increasing muscle cGMP with sildenafil decreased ATP synthesis efficiency and capacity, without impacting mitochondrial content or ultrastructure. INNOVATION GC may represent a new target for alleviating muscle fatigue and that NO-cGMP signaling may play important roles in muscle structure, contractility, and bioenergetics. CONCLUSIONS These findings suggest that GC activity is nNOS dependent and that muscle-specific control of GC expression and differential GC targeting may facilitate NO-cGMP signaling diversity. They suggest that nNOS regulates muscle fiber type, microtubule organization, fatigability, and postexercise force recovery partly through GC1 and suggest that NO-cGMP pathways may modulate mitochondrial ATP synthesis efficiency. Antioxid. Redox Signal. 26, 966-985.
Collapse
Affiliation(s)
- Younghye Moon
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| | - Jordan E Balke
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| | - Derik Madorma
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| | - Michael P Siegel
- 2 Department of Bioengineering, University of Washington , Seattle, Washington
| | - Gary Knowels
- 2 Department of Bioengineering, University of Washington , Seattle, Washington
| | - Peter Brouckaert
- 3 Department for Molecular Biomedical Research and Biomedical Molecular Biology, Ghent University , Ghent, Belgium
| | - Emmanuel S Buys
- 4 Department of Anesthesia, Critical Care and Pain Medicine, Anesthesia Center for Critical Care Research , Massachusetts General Hospital, Boston, Massachusetts
| | - David J Marcinek
- 2 Department of Bioengineering, University of Washington , Seattle, Washington.,5 Department of Radiology, University of Washington , Seattle, Washington
| | - Justin M Percival
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|
43
|
Abstract
Nitric oxide (NO) is an imperative regulator of the cardiovascular system and is a critical mechanism in preventing the pathogenesis and progression of the diseased heart. The scenario of bioavailable NO in the myocardium is complex: 1) NO is derived from both endogenous NO synthases (endothelial, neuronal, and/or inducible NOSs [eNOS, nNOS, and/or iNOS]) and exogenous sources (entero-salivary NO pathway) and the amount of NO from exogenous sources varies significantly; 2) NOSs are located at discrete compartments of cardiac myocytes and are regulated by distinctive mechanisms under stress; 3) NO regulates diverse target proteins through different modes of post-transcriptional modification (soluble guanylate cyclase [sGC]/cyclic guanosine monophosphate [cGMP]/protein kinase G [PKG]-dependent phosphorylation,
S-nitrosylation, and transnitrosylation); 4) the downstream effectors of NO are multidimensional and vary from ion channels in the plasma membrane to signalling proteins and enzymes in the mitochondria, cytosol, nucleus, and myofilament; 5) NOS produces several radicals in addition to NO (e.g. superoxide, hydrogen peroxide, peroxynitrite, and different NO-related derivatives) and triggers redox-dependent responses. However, nNOS inhibits cardiac oxidases to reduce the sources of oxidative stress in diseased hearts. Recent consensus indicates the importance of nNOS protein in cardiac protection under pathological stress. In addition, a dietary regime with high nitrate intake from fruit and vegetables together with unsaturated fatty acids is strongly associated with reduced cardiovascular events. Collectively, NO-dependent mechanisms in healthy and diseased hearts are better understood and shed light on the therapeutic prospects for NO and NOSs in clinical applications for fatal human heart diseases.
Collapse
Affiliation(s)
- Yin Hua Zhang
- Department of Physiology & Biomedical Sciences, College of Medicine, Seoul National University, 103 Dae Hak Ro, Chong No Gu, 110-799 Seoul, Korea, South.,Yanbian University Hospital, Yanji, Jilin Province, 133000, China.,Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
44
|
Leitner LM, Wilson RJ, Yan Z, Gödecke A. Reactive Oxygen Species/Nitric Oxide Mediated Inter-Organ Communication in Skeletal Muscle Wasting Diseases. Antioxid Redox Signal 2017; 26:700-717. [PMID: 27835923 PMCID: PMC5421600 DOI: 10.1089/ars.2016.6942] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Cachexia is defined as a complex metabolic syndrome that is associated with underlying illness and a loss of muscle with or without loss of fat mass. This disease is associated with a high incidence with chronic diseases such as heart failure, cancer, chronic obstructive pulmonary disease (COPD), and acquired immunodeficiency syndrome (AIDS), among others. Since there is currently no effective treatment available, cachectic patients have a poor prognosis. Elucidation of the underlying mechanisms is, therefore, an important medical task. Recent Advances: There is accumulating evidence that the diseased organs such as heart, lung, kidney, or cancer tissue secrete soluble factors, including Angiotensin II, myostatin (growth differentiation factor 8 [GDF8]), GDF11, tumor growth factor beta (TGFβ), which act on skeletal muscle. There, they induce a set of genes called atrogenes, which, among others, induce the ubiquitin-proteasome system, leading to protein degradation. Moreover, elevated reactive oxygen species (ROS) levels due to modulation of NADPH oxidases (Nox) and mitochondrial function contribute to disease progression, which is characterized by loss of muscle mass, exercise resistance, and frailty. CRITICAL ISSUES Although substantial progress was achieved to elucidate the pathophysiology of cachexia, effectice therapeutic strategies are urgently needed. FUTURE DIRECTIONS With the identification of key components of the aberrant inter-organ communication leading to cachexia, studies in mice and men to inhibit ROS formation, induction of anti-oxidative superoxide dismutases, and upregulation of muscular nitric oxide (NO) formation either by pharmacological tools or by exercise are promising approaches to reduce the extent of skeletal muscle wasting. Antioxid. Redox Signal. 26, 700-717.
Collapse
Affiliation(s)
- Lucia M Leitner
- 1 Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität Düsseldorf, Universitätsklinikum , Düsseldorf, Germany
| | - Rebecca J Wilson
- 2 Department of Medicine-Cardiovascular Medicine, University of Virginia , Charlottesville, Virginia
| | - Zhen Yan
- 2 Department of Medicine-Cardiovascular Medicine, University of Virginia , Charlottesville, Virginia.,3 Center for Skeletal Muscle Research at Robert Berne Cardiovascular Research Center, University of Virginia , Charlottesville, Virginia
| | - Axel Gödecke
- 1 Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität Düsseldorf, Universitätsklinikum , Düsseldorf, Germany
| |
Collapse
|
45
|
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: 1.6] [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
|
46
|
Moon Y, Cao Y, Zhu J, Xu Y, Balkan W, Buys ES, Diaz F, Kerrick WG, Hare JM, Percival JM. GSNOR Deficiency Enhances In Situ Skeletal Muscle Strength, Fatigue Resistance, and RyR1 S-Nitrosylation Without Impacting Mitochondrial Content and Activity. Antioxid Redox Signal 2017; 26:165-181. [PMID: 27412893 PMCID: PMC5278832 DOI: 10.1089/ars.2015.6548] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIM Nitric oxide (NO) plays important, but incompletely defined roles in skeletal muscle. NO exerts its regulatory effects partly though S-nitrosylation, which is balanced by denitrosylation by enzymes such as S-nitrosoglutathione reductase (GSNOR), whose functions in skeletal muscle remain to be fully deciphered. RESULTS GSNOR null (GSNOR-/-) tibialis anterior (TA) muscles showed normal growth and were stronger and more fatigue resistant than controls in situ. However, GSNOR-/- lumbrical muscles showed normal contractility and Ca2+ handling in vitro, suggesting important differences in GSNOR function between muscles or between in vitro and in situ environments. GSNOR-/- TA muscles exhibited normal mitochondrial content, and capillary densities, but reduced type IIA fiber content. GSNOR inhibition did not impact mitochondrial respiratory complex I, III, or IV activities. These findings argue that enhanced GSNOR-/- TA contractility is not driven by changes in mitochondrial content or activity, fiber type, or blood vessel density. However, loss of GSNOR led to RyR1 hypernitrosylation, which is believed to increase muscle force output under physiological conditions. cGMP synthesis by soluble guanylate cyclase (sGC) was decreased in resting GSNOR-/- muscle and was more responsive to agonist (DETANO, BAY 41, and BAY 58) stimulation, suggesting that GSNOR modulates cGMP production in skeletal muscle. INNOVATION GSNOR may act as a "brake" on skeletal muscle contractile performance under physiological conditions by modulating nitrosylation/denitrosylation balance. CONCLUSIONS GSNOR may play important roles in skeletal muscle contractility, RyR1 S-nitrosylation, fiber type specification, and sGC activity. Antioxid. Redox Signal. 26, 165-181.
Collapse
Affiliation(s)
- Younghye Moon
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| | - Yenong Cao
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida.,2 The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine , Miami, Florida
| | - Jingjing Zhu
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| | - Yuanyuan Xu
- 3 Department of Physiology and Biophysics, University of Miami Miller School of Medicine , Miami, Florida
| | - Wayne Balkan
- 2 The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine , Miami, Florida.,4 Department of Medicine, University of Miami Miller School of Medicine , Miami, Florida
| | - Emmanuel S Buys
- 5 Department of Anesthesia, Critical Care and Pain Medicine, Anesthesia Center for Critical Care Research , Harvard Medical School, Massachusetts General Hospital Boston, Boston, Massachusetts
| | - Francisca Diaz
- 6 Department of Neurology, University of Miami Miller School of Medicine , Miami, Florida
| | - W Glenn Kerrick
- 3 Department of Physiology and Biophysics, University of Miami Miller School of Medicine , Miami, Florida
| | - Joshua M Hare
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida.,2 The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine , Miami, Florida.,4 Department of Medicine, University of Miami Miller School of Medicine , Miami, Florida
| | - Justin M Percival
- 1 Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|
47
|
Spinazzola JM, Kunkel LM. Pharmacological therapeutics targeting the secondary defects and downstream pathology of Duchenne muscular dystrophy. Expert Opin Orphan Drugs 2016; 4:1179-1194. [PMID: 28670506 DOI: 10.1080/21678707.2016.1240613] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Since the identification of the dystrophin gene in 1986, a cure for Duchenne muscular dystrophy (DMD) has yet to be discovered. Presently, there are a number of genetic-based therapies in development aimed at restoration and/or repair of the primary defect. However, growing understanding of the pathophysiological consequences of dystrophin absence has revealed several promising downstream targets for the development of therapeutics. AREAS COVERED In this review, we discuss various strategies for DMD therapy targeting downstream consequences of dystrophin absence including loss of muscle mass, inflammation, fibrosis, calcium overload, oxidative stress, and ischemia. The rationale of each approach and the efficacy of drugs in preclinical and clinical studies are discussed. EXPERT OPINION For the last 30 years, effective DMD drug therapy has been limited to corticosteroids, which are associated with a number of negative side effects. Our knowledge of the consequences of dystrophin absence that contribute to DMD pathology has revealed several potential therapeutic targets. Some of these approaches may have potential to improve or slow disease progression independently or in combination with genetic-based approaches. The applicability of these pharmacological therapies to DMD patients irrespective of their genetic mutation, as well as the potential benefits even for advanced stage patients warrants their continued investigation.
Collapse
Affiliation(s)
- Janelle M Spinazzola
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115
| | - Louis M Kunkel
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115.,The Stem Cell Program at Boston Children's Hospital, Boston, MA 02115.,The Manton Center for Orphan Diseases, Boston, MA 02115.,Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
48
|
Mason SA, Morrison D, McConell GK, Wadley GD. Muscle redox signalling pathways in exercise. Role of antioxidants. Free Radic Biol Med 2016; 98:29-45. [PMID: 26912034 DOI: 10.1016/j.freeradbiomed.2016.02.022] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/05/2016] [Accepted: 02/17/2016] [Indexed: 01/01/2023]
Abstract
Recent research highlights the importance of redox signalling pathway activation by contraction-induced reactive oxygen species (ROS) and nitric oxide (NO) in normal exercise-related cellular and molecular adaptations in skeletal muscle. In this review, we discuss some potentially important redox signalling pathways in skeletal muscle that are involved in acute and chronic responses to contraction and exercise. Specifically, we discuss redox signalling implicated in skeletal muscle contraction force, mitochondrial biogenesis and antioxidant enzyme induction, glucose uptake and muscle hypertrophy. Furthermore, we review evidence investigating the impact of major exogenous antioxidants on these acute and chronic responses to exercise. Redox signalling pathways involved in adaptive responses in skeletal muscle to exercise are not clearly elucidated at present, and further research is required to better define important signalling pathways involved. Evidence of beneficial or detrimental effects of specific antioxidant compounds on exercise adaptations in muscle is similarly limited, particularly in human subjects. Future research is required to not only investigate effects of specific antioxidant compounds on skeletal muscle exercise adaptations, but also to better establish mechanisms of action of specific antioxidants in vivo. Although we feel it remains somewhat premature to make clear recommendations in relation to application of specific antioxidant compounds in different exercise settings, a bulk of evidence suggests that N-acetylcysteine (NAC) is ergogenic through its effects on maintenance of muscle force production during sustained fatiguing events. Nevertheless, a current lack of evidence from studies using performance tests representative of athletic competition and a potential for adverse effects with high doses (>70mg/kg body mass) warrants caution in its use for performance enhancement. In addition, evidence implicates high dose vitamin C (1g/day) and E (≥260 IU/day) supplementation in impairments to some skeletal muscle cellular adaptations to chronic exercise training. Thus, determining the utility of antioxidant supplementation in athletes likely requires a consideration of training and competition periodization cycles of athletes in addition to type, dose and duration of antioxidant supplementation.
Collapse
Affiliation(s)
- Shaun A Mason
- Centre for Physical Activity and Nutrition (C-PAN) Research, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Dale Morrison
- Centre for Physical Activity and Nutrition (C-PAN) Research, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Glenn K McConell
- Clinical Exercise Science Research Program, Institute for Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, Victoria, Australia
| | - Glenn D Wadley
- Centre for Physical Activity and Nutrition (C-PAN) Research, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia.
| |
Collapse
|
49
|
Hord JM, Botchlett R, Lawler JM. Age-related alterations in the sarcolemmal environment are attenuated by lifelong caloric restriction and voluntary exercise. Exp Gerontol 2016; 83:148-57. [PMID: 27534381 DOI: 10.1016/j.exger.2016.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 07/25/2016] [Accepted: 08/10/2016] [Indexed: 10/21/2022]
Abstract
Age-related loss of skeletal muscle mass and function, referred to as sarcopenia, is mitigated by lifelong calorie restriction as well as exercise. In aged skeletal muscle fibers there is compromised integrity of the cell membrane that may contribute to sarcopenia. The purpose of this study was to determine if lifelong mild (8%) caloric restriction (CR) and lifelong CR+voluntary wheel running (WR) could ameliorate disruption of membrane scaffolding and signaling proteins during the aging process, thus maintaining a favorable, healthy membrane environment in plantaris muscle fibers. Fischer-344 rats were divided into four groups: 24-month old adults fed ad libitum (OAL); 24-month old on 8% caloric restriction (OCR); 24month old 8% caloric restriction+wheel running (OCRWR); and 6-month old sedentary adults fed ad libitum (YAL) were used to determine age-related changes. Aging resulted in discontinuous membrane expression of dystrophin glycoprotein complex (DGC) proteins: dystrophin and α-syntrophin. Older muscle also displayed decreased content of neuronal nitric oxide synthase (nNOS), a key DGC signaling protein. In contrast, OCR and OCRWR provided significant protection against age-related DGC disruption. In conjunction with the age-related decline in membrane DGC patency, key membrane repair proteins (MG53, dysferlin, annexin A6, and annexin A2) were significantly increased in the OAL plantaris. However, lifelong CR and CRWR interventions were effective at maintaining membrane repair proteins near YAL levels of. OAL fibers also displayed reduced protein content of NADPH oxidase isoform 2 (Nox2) subunits (p67phox and p47phox), consistent with a perturbed sarcolemmal environment. Loss of Nox2 subunits was prevented by lifelong CR and CRWR. Our results are therefore consistent with the hypothesis that lifelong CR and WR are effective countermeasures against age-related alterations in the myofiber membrane environment.
Collapse
Affiliation(s)
- Jeffrey M Hord
- Redox Biology & Cell Signaling Laboratory, Department of Health and Kinesiology, College of Education and Human Development, Texas A&M University, College Station, TX, United States
| | - Rachel Botchlett
- Department of Nutrition & Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, United States
| | - John M Lawler
- Redox Biology & Cell Signaling Laboratory, Department of Health and Kinesiology, College of Education and Human Development, Texas A&M University, College Station, TX, United States; Department of Nutrition & Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, United States.
| |
Collapse
|
50
|
Pan F, Mi JY, Zhang Y, Pan XY, Rui YJ. Muscle fiber types composition and type identified endplate morphology of forepaw intrinsic muscles in the rat. J Muscle Res Cell Motil 2016; 37:95-100. [PMID: 27460929 DOI: 10.1007/s10974-016-9450-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/08/2016] [Indexed: 11/26/2022]
Abstract
The failure to accept reinnervation is considered to be one of the reasons for the poor motor functional recovery of intrinsic hand muscles (IHMs) after nerve injury. Rat could be a suitable model to be used in simulating motor function recovery of the IHMs after nerve injury as to the similarities in function and anatomy of the muscles between human and rat. However, few studies have reported the muscle fiber types composition and endplate morphologic characteristics of intrinsic forepaw muscles (IFMs) in the rat. In this study, the myosin heavy chain isoforms and acetylcholine receptors were stained by immunofluorescence to show the muscle fiber types composition and endplates on type-identified fibers of the lumbrical muscles (LMs), interosseus muscles (IMs), abductor digiti minimi (AM) and flexor pollicis brevis (FM) in rat forepaw. The majority of IFMs fibers were labeled positively for fast-switch fiber. However, the IMs were composed of only slow-switch fiber. With the exception of the IMs, the other IFMs had a part of hybrid fibers. Two-dimensional morphological characteristics of endplates on I and IIa muscle fiber had no significant differences among the IFMs. The LMs is the most suitable IFMs of rat to stimulate reinnervation of the IHMs after nerve injury. Gaining greater insight into the muscle fiber types composition and endplate morphology in the IFMs of rat may help understand the pathological and functional changes of IFMs in rat model stimulating reinnervation of IHMs after peripheral nerve injury.
Collapse
Affiliation(s)
- Feng Pan
- Department of Hand Surgery, Wuxi Number 9 People's Hospital Affiliated to Soochow University, Wuxi, 214062, Jiangsu, People's Republic of China
| | - Jing-Yi Mi
- Department of Hand Surgery, Wuxi Number 9 People's Hospital Affiliated to Soochow University, Wuxi, 214062, Jiangsu, People's Republic of China
| | - Yan Zhang
- Department of Hand Surgery, Wuxi Number 9 People's Hospital Affiliated to Soochow University, Wuxi, 214062, Jiangsu, People's Republic of China
| | - Xiao-Yun Pan
- Department of Hand Surgery, Wuxi Number 9 People's Hospital Affiliated to Soochow University, Wuxi, 214062, Jiangsu, People's Republic of China
| | - Yong-Jun Rui
- Department of Hand Surgery, Wuxi Number 9 People's Hospital Affiliated to Soochow University, Wuxi, 214062, Jiangsu, People's Republic of China.
| |
Collapse
|