bims-moremu Biomed News
on Molecular regulators of muscle mass
Issue of 2021‒04‒11
33 papers selected by
Anna Vainshtein
Craft Science Inc.


  1. FASEB J. 2021 May;35(5): e21511
      Hydrogen sulfide (H2 S) can be endogenously produced and belongs to the class of signaling molecules known as gasotransmitters. Cystathionine gamma-lyase (CSE)-derived H2 S is implicated in the regulation of cell differentiation and the aging process, but the involvements of the CSE/H2 S system in myogenesis upon aging and injury have not been explored. In this study, we demonstrated that CSE acts as a major H2 S-generating enzyme in skeletal muscles and is significantly down-regulated in aged skeletal muscles in mice. CSE deficiency exacerbated the age-dependent sarcopenia and cardiotoxin-induced injury/regeneration in mouse skeletal muscle, possibly attributed to inefficient myogenesis. In contrast, supplement of NaHS (an H2 S donor) induced the expressions of myogenic genes and promoted muscle regeneration in mice. In vitro, incubation of myoblast cells (C2C12) with H2 S promoted myogenesis, as evidenced by the inhibition of cell cycle progression and migration, altered expressions of myogenic markers, elongation of myoblasts, and formation of multinucleated myotubes. Myogenesis was also found to upregulate CSE expression, while blockage of CSE/H2 S signaling resulted in a suppression of myogenesis. Mechanically, H2 S significantly induced the heterodimer formation between MEF2c and MRF4 and promoted the binding of MEF2c/MRF4 to myogenin promoter. MEF2c was S-sulfhydrated at both cysteine 361 and 420 in the C-terminal transactivation domain, and blockage of MEF2c S-sulfhydration abolished the stimulatory role of H2 S on MEF2c/MRF4 heterodimer formation. These findings support an essential role for H2 S in maintaining myogenesis, presenting it as a potential candidate for the prevention of age-related sarcopenia and treatment of muscle injury.
    Keywords:  H2S; MEF2c; S-sulfhydration; aging; cystathionine gamma-lyase; myogenesis
    DOI:  https://doi.org/10.1096/fj.202002675R
  2. Biofabrication. 2021 Apr 09.
      Myotonic dystrophy type 1 (DM1) is the most common hereditary myopathy in the adult population. The disease is characterized by progressive skeletal muscle degeneration that produces severe disability. At present, there is still no effective treatment for DM1 patients, but the breakthroughs in understanding the molecular pathogenic mechanisms in DM1 have allowed the testing of new therapeutic strategies. Animal models and in vitro two-dimensional cell cultures have been essential for these advances. However, serious concerns exist regarding how faithfully these models reproduce the biological complexity of the disease. Biofabrication tools can be applied to engineer human three-dimensional (3D) culture systems that complement current preclinical research models. Here, we describe the development of the first in vitro 3D model of DM1 human skeletal muscle. Transdifferentiated myoblasts from patient-derived fibroblasts were encapsulated in micromolded gelatin methacryloyl-carboxymethyl cellulose methacrylate (GelMA-CMCMA) hydrogels through photomold patterning on functionalized glass coverslips. These hydrogels present a microstructured topography that promotes myoblasts alignment and differentiation resulting in highly aligned myotubes from both healthy and DM1 cells in a long-lasting cell culture. The DM1 3D microtissues recapitulate the molecular alterations detected in patient biopsies. Importantly, fusion index analyses demonstrate that 3D micropatterning significantly improved DM1 cell differentiation into multinucleated myotubes compared to standard cell cultures. Moreover, the characterization of the 3D cultures of DM1 myotubes detects phenotypes as the reduced thickness of myotubes that can be used for drug testing. Finally, we evaluated the therapeutic effect of antagomiR-23b administration on bioengineered DM1 skeletal muscle microtissues. AntagomiR-23b treatment rescues both molecular DM1 hallmarks and structural phenotype, restoring myotube diameter to healthy control sizes. Overall, these new microtissues represent an improvement over conventional cell culture models and can be used as biomimetic platforms to establish preclinical studies for myotonic dystrophy.
    Keywords:  3D cell culture; hydrogel micropatterning; myotonic dystrophy; skeletal muscle; tissue engineering
    DOI:  https://doi.org/10.1088/1758-5090/abf6ae
  3. Crit Rev Biochem Mol Biol. 2021 Apr 07. 1-17
      Muscle stem cells (MuSCs) also called satellite cells are the building blocks of skeletal muscle, the largest tissue in the human body which is formed primarily of myofibers. While MuSCs are the principal cells that directly contribute to the formation of the muscle fibers, their ability to do so depends on critical interactions with a vast array of nonmyogenic cells within their niche environment. Therefore, understanding the nature of communication between MuSCs and their niche is of key importance to understand how the skeletal muscle is maintained and regenerated after injury. MuSCs are rare and therefore difficult to study in vivo within the context of their niche environment. The advent of single-cell technologies, such as switching mechanism at 5' end of the RNA template (SMART) and tagmentation based technologies using hyperactive transposase, afford the unprecedented opportunity to perform whole transcriptome and epigenome studies on rare cells within their niche environment. In this review, we will delve into how single-cell technologies can be applied to the study of MuSCs and muscle-resident niche cells and the impact this can have on our understanding of MuSC biology and skeletal muscle regeneration.
    Keywords:  ATAC-Seq; SMART-Seq; Single cell technologies; chromatin; gene expression; muscle stem cells; niche environment; skeletal muscle
    DOI:  https://doi.org/10.1080/10409238.2021.1908950
  4. Mol Metab. 2021 Mar 31. pii: S2212-8778(21)00071-5. [Epub ahead of print] 101226
      MicroRNAs (miRNA) are known to regulate expression of genes involved in several physiological processes including metabolism, mitochondrial biogenesis, proliferation, differentiation, and cell death. Using "in silico" analyses, we identified 219 unique miRNAs that potentially bind to the 3'UTR region of a critical mitochondrial regulator, the peroxisome proliferator-activated receptor gamma coactivator (PGC) 1 alpha (Pgc1α). Out of the 219 candidate miRNAs, miR-696 had one of the highest interactions at the 3'UTR of Pgc1α, suggesting that miR-696 may be involved in the regulation of Pgc1α. Consistent with this hypothesis, we found that miR-696 was highly expressed in the skeletal muscle of both STZ-induced diabetic mice and chronic high-fat fed mice. C2C12 muscle cells exposed to palmitic acid also exhibited higher expression of miR-696. This increased expression corresponded with reduced expression of oxidative metabolism genes and reduced mitochondrial respiration. Importantly, reduction of miR-696 reversed decreases in mitochondrial activity in response to palmitic acid. Using C2C12 cells treated with the AMP-activated protein kinase (AMPK) activator AICAR and skeletal muscle from AMPKα2 dominant negative (DN) mice, we found that the signaling mechanism regulating miR-696 does not involve AMPK. In contrast, overexpression of SNF1-AMPK-related kinase (SNARK) in C2C12 cells increased miR-696 transcription while the knockdown of SNARK significantly decreased miR-696. Moreover, muscle-specific transgenic mice overexpressing SNARK exhibited lower expression of Pgc1α, elevated levels of miR-696, and reduced amounts of spontaneous activity. Our findings demonstrate that metabolic stress increases miR-696 expression in skeletal muscle cells, which in turn inhibits Pgc1α, reducing mitochondrial function. SNARK plays a role in this process as a metabolic stress signaling molecule inducing the expression of miR-696.
    Keywords:  SNARK; and Pgc1α; miR-696; mitochondrial function; skeletal muscle
    DOI:  https://doi.org/10.1016/j.molmet.2021.101226
  5. Cell Mol Life Sci. 2021 Apr 07.
      Duchenne muscular dystrophy (DMD) is a devastating chromosome X-linked disease that manifests predominantly in progressive skeletal muscle wasting and dysfunctions in the heart and diaphragm. Approximately 1/5000 boys and 1/50,000,000 girls suffer from DMD, and to date, the disease is incurable and leads to premature death. This phenotypic severity is due to mutations in the DMD gene, which result in the absence of functional dystrophin protein. Initially, dystrophin was thought to be a force transducer; however, it is now considered an essential component of the dystrophin-associated protein complex (DAPC), viewed as a multicomponent mechanical scaffold and a signal transduction hub. Modulating signal pathway activation or gene expression through epigenetic modifications has emerged at the forefront of therapeutic approaches as either an adjunct or stand-alone strategy. In this review, we propose a broader perspective by considering DMD to be a disease that affects myofibers and muscle stem (satellite) cells, as well as a disorder in which abrogated communication between different cell types occurs. We believe that by taking this systemic view, we can achieve safe and holistic treatments that can restore correct signal transmission and gene expression in diseased DMD tissues.
    Keywords:  DMD; Duchenne muscular dystrophy; Signaling pathways; Striated muscles
    DOI:  https://doi.org/10.1007/s00018-021-03821-x
  6. Hum Mol Genet. 2021 Apr 05. pii: ddab100. [Epub ahead of print]
      Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder characterized by progressive muscular weakness due to the loss of dystrophin. Extracellular Ca2+ flows into the cytoplasm through membrane tears in dystrophin-deficient myofibers, which leads to muscle contracture and necrosis. Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) takes up cytosolic Ca2+ into the sarcoplasmic reticulum (SR), but its activity is decreased in dystrophic muscle. Here, we show that an allosteric SERCA activator, CDN1163, ameliorates dystrophic phenotypes in dystrophin-deficient mdx mice. Administration of CDN1163 prevented exercise-induced muscular damage and restored mitochondrial function. In addition, treatment with CDN1163 for seven weeks enhanced muscular strength and reduced muscular degeneration and fibrosis in mdx mice. Our findings provide preclinical proof-of-concept evidence that pharmacological activation of SERCA could be a promising therapeutic strategy for DMD. Moreover, CDN1163 improved muscular strength surprisingly in wild-type mice, which may pave the new way for the treatment of muscular dysfunction.
    DOI:  https://doi.org/10.1093/hmg/ddab100
  7. Sci Transl Med. 2021 Apr 07. pii: eabb0319. [Epub ahead of print]13(588):
      Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy, and despite advances in genetic and pharmacological disease-modifying treatments, its management remains a major challenge. Mitochondrial dysfunction contributes to DMD, yet the mechanisms by which this occurs remain elusive. Our data in experimental models and patients with DMD show that reduced expression of genes involved in mitochondrial autophagy, or mitophagy, contributes to mitochondrial dysfunction. Mitophagy markers were reduced in skeletal muscle and in muscle stem cells (MuSCs) of a mouse model of DMD. Administration of the mitophagy activator urolithin A (UA) rescued mitophagy in DMD worms and mice and in primary myoblasts from patients with DMD, increased skeletal muscle respiratory capacity, and improved MuSCs' regenerative ability, resulting in the recovery of muscle function and increased survival in DMD mouse models. These data indicate that restoration of mitophagy alleviates symptoms of DMD and suggest that UA may have potential therapeutic applications for muscular dystrophies.
    DOI:  https://doi.org/10.1126/scitranslmed.abb0319
  8. Am J Physiol Cell Physiol. 2021 Apr 07.
      The miR-129 family is widely reported as tumor repressors, while, their roles in skeletal muscle have not been fully investigated. Here, the function and mechanism of miR-129-5p in skeletal muscle, a member of the miR-129 family, were explored using C2C12 cell line. Our study shown that miR-129-5p was widely detected in mouse tissues, with the highest expression in skeletal muscle. Gain- and loss-of-function study shown that miR-129-5p could negatively regulate myogenic differentiation, indicated by reduced ratio of MyHC-positive myofibers and repressed expression of myogenic genes, such as MyoD, MyoG and MyHC. Furthermore, miR-129-5p was more enriched in fast extensor digitalis lateralis (EDL) than in slow soleus (SOL). Enhanced miR-129-5p could significantly reduce the expression of mitochondrial cox family, together with that of MyHC I, and knockdown of miR-129-5p conversely increased the expression of cox genes and MyHC I. Mechanistically, miR-129-5p directly targeted the 3'-UTR of Mef2a, which was suppressed by miR-129-5p agomir at both mRNA and protein levels in C2C12 cells. Moreover, overexpression of Mef2a could rescue the inhibitory effects of miR-129-5p on the expression of myogenic factors and MyHC I. Collectively, our data revealed that miR-129-5p as a negative regulator of myogenic differentiation and slow fiber gene expression, thus affecting body metabolic homeostasis.
    Keywords:  C2C12 cell line; Mef2a; MiR-129-5p; myogenic differentiation
    DOI:  https://doi.org/10.1152/ajpcell.00578.2020
  9. J Cell Mol Med. 2021 Apr 09.
      Fibrosis after skeletal muscle injury is common in sports and can cause irreversible damage to the biomechanical properties of skeletal muscle. Long non-coding RNAs (lncRNAs) have been validated to act as important modulators in the fibrosis of various organs. Here, we reported a novel lncRNA (the skeletal muscle fibrosis-associated transcript 1, lnc-MFAT1), which was highly expressed in skeletal muscle fibrosis. We demonstrate that lnc-MFAT1 knockdown can reduce TGFβ-induced fibrosis in vitro and attenuate skeletal muscle fibrosis after acute contusion in mice. Further study showed that lnc-MFAT1 acted as a competitive endogenous RNA of miR-135a-5p. Besides, the miR-135a-5p inhibition obviously promoted TGFβ-induced fibrosis in vitro via enhancing its target genes Tgfbr2/Smad4. Moreover, we discovered that lnc-MFAT1 regulates Tgfbr2/Smad4 expression by sponging miR-135a-5p to exert competing endogenous RNA function, resulting in TGFβ pathway activation. In conclusion, our study identified a crucial role of lnc-MFAT1-miR-135a-Tgfbr2/Smad4 axis in skeletal muscle fibrosis, providing a promising treatment option against skeletal muscle fibrosis.
    Keywords:  TGFβ pathway; long noncoding RNA; miR-135a-5p; skeletal muscle fibrosis
    DOI:  https://doi.org/10.1111/jcmm.16508
  10. Nucleic Acids Res. 2021 Apr 09. pii: gkab226. [Epub ahead of print]
      Skeletal muscle is a dynamic tissue the size of which can be remodeled through the concerted actions of various cues. Here, we investigated the skeletal muscle transcriptional program and identified key tissue-specific regulatory genetic elements. Our results show that Myod1 is bound to numerous skeletal muscle enhancers in collaboration with the glucocorticoid receptor (GR) to control gene expression. Remarkably, transcriptional activation controlled by these factors occurs through direct contacts with the promoter region of target genes, via the CpG-bound transcription factor Nrf1, and the formation of Ctcf-anchored chromatin loops, in a myofiber-specific manner. Moreover, we demonstrate that GR negatively controls muscle mass and strength in mice by down-regulating anabolic pathways. Taken together, our data establish Myod1, GR and Nrf1 as key players of muscle-specific enhancer-promoter communication that orchestrate myofiber size regulation.
    DOI:  https://doi.org/10.1093/nar/gkab226
  11. Nat Commun. 2021 04 08. 12(1): 2099
      In Duchenne muscular dystrophy (DMD), sarcolemma fragility and myofiber necrosis produce cellular debris that attract inflammatory cells. Macrophages and T-lymphocytes infiltrate muscles in response to damage-associated molecular pattern signalling and the release of TNF-α, TGF-β and interleukins prevent skeletal muscle improvement from the inflammation. This immunological scenario was extended by the discovery of a specific response to muscle antigens and a role for regulatory T cells (Tregs) in muscle regeneration. Normally, autoimmunity is avoided by autoreactive T-lymphocyte deletion within thymus, while in the periphery Tregs monitor effector T-cells escaping from central regulatory control. Here, we report impairment of thymus architecture of mdx mice together with decreased expression of ghrelin, autophagy dysfunction and AIRE down-regulation. Transplantation of dystrophic thymus in recipient nude mice determine the up-regulation of inflammatory/fibrotic markers, marked metabolic breakdown that leads to muscle atrophy and loss of force. These results indicate that involution of dystrophic thymus exacerbates muscular dystrophy by altering central immune tolerance.
    DOI:  https://doi.org/10.1038/s41467-021-22305-x
  12. Cell Metab. 2021 Apr 06. pii: S1550-4131(21)00127-3. [Epub ahead of print]33(4): 758-780
      As the principal tissue for insulin-stimulated glucose disposal, skeletal muscle is a primary driver of whole-body glycemic control. Skeletal muscle also uniquely responds to muscle contraction or exercise with increased sensitivity to subsequent insulin stimulation. Insulin's dominating control of glucose metabolism is orchestrated by complex and highly regulated signaling cascades that elicit diverse and unique effects on skeletal muscle. We discuss the discoveries that have led to our current understanding of how insulin promotes glucose uptake in muscle. We also touch upon insulin access to muscle, and insulin signaling toward glycogen, lipid, and protein metabolism. We draw from human and rodent studies in vivo, isolated muscle preparations, and muscle cell cultures to home in on the molecular, biophysical, and structural elements mediating these responses. Finally, we offer some perspective on molecular defects that potentially underlie the failure of muscle to take up glucose efficiently during obesity and type 2 diabetes.
    Keywords:  GLUT4; diabetes; exercise-induced sensitization to insulin; glucose uptake; glycogen; insulin; insulin resistance; insulin signaling; skeletal muscle
    DOI:  https://doi.org/10.1016/j.cmet.2021.03.020
  13. Eur J Appl Physiol. 2021 Apr 08.
      Blood flow-restricted (BFR) exercise can induce training adaptations comparable to those observed following training in free flow conditions. However, little is known about the acute responses within skeletal muscle following BFR aerobic exercise (AE). Moreover, although preliminary evidence suggests chronic BFR AE may augment certain training adaptations in skeletal muscle mitochondria more than non-BFR AE, the underlying mechanisms are poorly understood. In this review, we summarise the acute BFR AE literature examining mitochondrial biogenic signalling pathways and provide insight into mechanisms linked to skeletal muscle remodelling following BFR AE. Specifically, we focus on signalling pathways potentially contributing to augmented peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) mRNA following work-rate-matched BFR AE compared with non-BFR AE. We present evidence suggesting reductions in muscle oxygenation during acute BFR AE lead to increased intracellular energetic stress, AMP-activated protein kinase (AMPK) activation and PGC-1α mRNA. In addition, we briefly discuss mitochondrial adaptations to BFR aerobic training, and we assess the risk of bias using the Cochrane Collaboration risk of bias assessment tool. We ultimately call for several straightforward modifications to help minimise bias in future BFR AE studies.
    Keywords:  AMPK; Aerobic exercise; BFR; Ischemic training; Mitochondria; Occlusion training; PGC-1α; Risk of bias
    DOI:  https://doi.org/10.1007/s00421-021-04669-6
  14. Aging Pathobiol Ther. 2020 ;2(1): 45-51
      Objective: Mitochondrial dysfunction comprises part of the etiology of myriad health issues, particularly those that occur with advancing age. Methionine sulfoxide reductase A (MsrA) is a ubiquitous protein oxidation repair enzyme that specifically and catalytically reduces a specific epimer of oxidized methionine: methionine sulfoxide. In this study, we tested the ways in which mitochondrial bioenergetic functions are affected by increasing MsrA expression in different cellular compartments.Methods: In this study, we tested the function of isolated mitochondria, including free radical generation, ATP production, and respiration, from the skeletal muscle of two lines of transgenic mice with increased MsrA expression: mitochondria-targeted MsrA overexpression or cytosol-targeted MsrA overexpression.
    Results: Surprisingly, in the samples from mice with mitochondrial-targeted MsrA overexpression, we found dramatically increased free radical production though no specific defect in respiration, ATP production, or membrane potential. Among the electron transport chain complexes, we found the activity of complex I was specifically reduced in mitochondrial MsrA transgenic mice. In mice with cytosolic-targeted MsrA overexpression, we found no significant alteration made to any of these parameters of mitochondrial energetics.
    Conclusions: There is also a growing amount of evidence that MsrA is a functional requirement for sustaining optimal mitochondrial respiration and free radical generation. MsrA is also known to play a partial role in maintaining normal protein homeostasis by specifically repairing oxidized proteins. Our studies highlight a potential novel role for MsrA in regulating the activity of mitochondrial function through its interaction with the mitochondrial proteome.
    Keywords:  Superoxide; electron transport chain; mitochondria; oxidative stress; protein homeostasis
    DOI:  https://doi.org/10.31491/apt.2020.03.012
  15. Cell Death Discov. 2021 Apr 09. 7(1): 73
      Long non-coding RNAs (lncRNAs) are well-known to participate in a variety of important regulatory processes in myogenesis. In our previous RNA-seq study (accession number GSE58755), we found that lncRNA-FKBP1C was differentially expressed between White Recessive Rock (WRR) and Xinghua (XH) chicken. Here, we have further demonstrated that lncRNA-FKBP1C interacted directly with MYH1B by biotinylated RNA pull-down assay and RNA immunoprecipitation (RIP). Protein stability and degradation experiments identified that lncRNA-FKBP1C enhanced the protein stability of MYH1B. Overexpression of lncRNA-FKBP1C inhibited myoblasts proliferation, promoted myoblasts differentiation, and participated in the formation of skeletal muscle fibers. LncRNA-FKBP1C could downregulate the fast muscle genes and upregulate slow muscle genes. Conversely, its interference promoted cell proliferation, repressed cell differentiation, and drove the transformation of slow-twitch muscle fibers to fast-twitch muscle fibers. Similar results were observed after knockdown of the MYH1B gene, but the difference was that the MYH1B gene had no effects on fast muscle fibers. In short, these data demonstrate that lncRNA-FKBP1C could bound with MYH1B and enhance its protein stability, thus affecting proliferation, differentiation of myoblasts and conversion of skeletal muscle fiber types.
    DOI:  https://doi.org/10.1038/s41420-021-00463-7
  16. Clin Sci (Lond). 2021 Apr 07. pii: CS20210128. [Epub ahead of print]
      Exercise training improves muscle fitness in many aspects, including induction of mitochondrial biogenesis and maintenance of mitochondrial dynamics. The insulin-like growth factors were recently proposed as key regulators of myogenic factors to regulate muscle development. This study aimed to investigate the physical exercise impact on insulin-like growth factor 2 (IGF2) and analyzed its functions on skeletal muscle cells in vitro. Using online databases, we stated that IGF2 was relatively highly expressed in skeletal muscle cells and increased after exercise training. Then, IGF2 deficiency in myotubes from C2C12 and primary skeletal muscle cells (PMSCs) led to impaired mitochondrial function, reduced mitochondrial-related protein content, and decreased mitochondrial biogenesis. Furthermore, we explored the possible regulatory pathway and found that mitochondrial regulation in skeletal muscle cells might occur through IGF2-SIRT1-PGC1α signaling pathway. Therefore, this study first demonstrated the relationship between IGF2 and mitochondria in skeletal muscle.
    Keywords:  Insulin-like growth factor 2; Sirtuin 1; mitochondrial biogenesis; mitochondrial function; peroxisome proliferator-activated receptor-γ co-activator-1α; skeletal muscle
    DOI:  https://doi.org/10.1042/CS20210128
  17. Front Immunol. 2021 ;12 586429
      Angiogenesis is an important process under both physiological and pathophysiological conditions. Here we investigated the role and the underlying mechanism of PD-1 in hindlimb ischemia-induced inflammation and angiogenesis in mice. We found that inhibition of PD-1 by genetic PD-1 knockout or pharmacological PD-1 blocking antibodies dramatically attenuated hindlimb blood perfusion, angiogenesis, and exercise capacity in mice after femoral artery ligation. Mechanistically, we found that PD-1 knockout significantly exacerbated ischemia-induced muscle oxidative stress, leukocyte infiltration and IFN-γ production before abnormal angiogenesis in these mice. In addition, we found that the percentages of IFN-γ positive macrophages and CD8 T cells were significantly increased in P-1 knockout mice after hindlimb ischemia. Macrophages were the major leukocyte subset infiltrated in skeletal muscle, which were responsible for the enhanced muscle leukocyte-derived IFN-γ production in PD-1 knockout mice after hindlimb ischemia. Moreover, we demonstrated that IFN-γ significantly attenuated vascular endothelial cell proliferation, tube formation and migration in vitro. IFN-γ also significantly enhanced vascular endothelial cell apoptosis. In addition, the total number of TNF-α positive leukocytes/muscle weight were significantly increased in PD-1-/- mice after hindlimb ischemia. These data indicate that PD-1 exerts an important role in ischemia-induced muscle inflammation and angiogenesis.
    Keywords:  PD-1; angiogenesis; hindlimb ischemia; inflammation; oxidative stress
    DOI:  https://doi.org/10.3389/fimmu.2021.586429
  18. Vet Clin North Am Equine Pract. 2021 Apr;pii: S0749-0739(20)30072-9. [Epub ahead of print]37(1): 139-175
      Skeletal muscle comprises 40% to 55% of mature body weight in horses, and its mass is determined largely by rates of muscle protein synthesis. In order to support exercise, appropriate energy sources are essential: glucose can support both anaerobic and aerobic exercise, whereas fat can only be metabolized aerobically. Following exercise, ingestion of nonfiber carbohydrates and protein can aid muscle growth and recovery. Muscle glycogen replenishment is slow in horses, regardless of dietary interventions. Several heritable muscle disorders, including type 1 and 2 polysaccharide storage myopathy and recurrent exertional rhabdomyolysis, can be managed in part by restricting dietary nonstructural carbohydrate intake.
    Keywords:  Exercise; Growth; Muscle disorders; Skeletal muscle
    DOI:  https://doi.org/10.1016/j.cveq.2020.12.005
  19. Sci Rep. 2021 Apr 08. 11(1): 7733
      Hamstring muscle injury is highly prevalent in sports involving repeated maximal sprinting. Although neuromuscular fatigue is thought to be a risk factor, the mechanisms underlying the fatigue response to repeated maximal sprints are unclear. Here, we show that repeated maximal sprints induce neuromuscular fatigue accompanied with a prolonged strength loss in hamstring muscles. The immediate hamstring strength loss was linked to both central and peripheral fatigue, while prolonged strength loss was associated with indicators of muscle damage. The kinematic changes immediately after sprinting likely protected fatigued hamstrings from excess elongation stress, while larger hamstring muscle physiological cross-sectional area and lower myoblast:fibroblast ratio appeared to protect against fatigue/damage and improve muscle recovery within the first 48 h after sprinting. We have therefore identified novel mechanisms that likely regulate the fatigue/damage response and initial recovery following repeated maximal sprinting in humans.
    DOI:  https://doi.org/10.1038/s41598-021-87195-x
  20. J Biol Chem. 2021 Jan 07. pii: S0021-9258(20)00343-9. [Epub ahead of print]296 100228
      The conserved C-terminal end segment of troponin I (TnI) plays a critical role in regulating muscle relaxation. This function is retained in the isolated C-terminal 27 amino acid peptide (residues 184-210) of human cardiac TnI (HcTnI-C27): When added to skinned muscle fibers, HcTnI-C27 reduces the Ca2+-sensitivity of activated myofibrils and facilitates relaxation without decreasing the maximum force production. However, the underlying mechanism of HcTnI-C27 function is unknown. We studied the conformational preferences of HcTnI-C27 and a myopathic mutant, Arg192His, (HcTnI-C27-H). Both peptides were mainly disordered in aqueous solution with a nascent helix involving residues from Trp191 to Ile195, as shown by NMR analysis and molecular dynamics simulations. The population of nascent helix was smaller in HcTnI-C27-H than in HcTnI-C27, as shown by circular dichroism (CD) titrations. Fluorescence and isothermal titration calorimetry (ITC) showed that both peptides bound tropomyosin (αTm), with a detectably higher affinity (∼10 μM) of HcTnI-C27 than that of HcTnI-C27-H (∼15 μM), consistent with an impaired Ca2+-desensitization effect of the mutant peptide on skinned muscle strips. Upon binding to αTm, HcTnI-C27 acquired a weakly stable helix-like conformation involving residues near Trp191, as shown by transferred nuclear Overhauser effect spectroscopy and hydrogen/deuterium exchange experiments. With the potent Ca2+-desensitization effect of HcTnI-C27 on skinned cardiac muscle from a mouse model of hypertrophic cardiomyopathy, the data support that the C-terminal end domain of TnI can function as an isolated peptide with the intrinsic capacity of binding tropomyosin, providing a promising therapeutic approach to selectively improve diastolic function of the heart.
    Keywords:  computer modeling; hypertrophic cardiomyopathy; muscle contractility; peptide conformation; skinned cardiac muscle; troponin I
    DOI:  https://doi.org/10.1074/jbc.RA120.016012
  21. FEBS J. 2021 Apr 07.
      Desmin is the primary intermediate filament (IF) protein of cardiac, skeletal and smooth muscle. By linking the contractile myofibrils to the sarcolemma and cellular organelles, desmin IF contribute to muscle structural and cellular integrity, force transmission, and mitochondrial homeostasis. Mutations in desmin cause myofibril misalignment, mitochondrial dysfunction, and impaired mechanical integrity leading to cardiac and skeletal myopathies in humans, often characterized by the accumulation of protein aggregates. Recent evidence indicates that desmin filaments also regulate proteostasis and cell size. In skeletal muscle, changes in desmin filament dynamics can facilitate catabolic events as an adaptive response to a changing environment. In addition, post-translational modifications of desmin and its misfolding in the heart have emerged as key determinants of homeostasis and disease. In this review, we provide an overview of the structural and cellular roles of desmin, and propose new models for its novel functions in preserving the homeostasis of striated muscles.
    Keywords:  GSK3; desmin intermediate filaments; heart failure; metabolism; muscle atrophy; protein degradation; protein misfolding
    DOI:  https://doi.org/10.1111/febs.15864
  22. Aging Dis. 2021 Apr;12(2): 494-514
      Action potential is transmitted to muscle fibers through specialized synaptic interfaces called neuromuscular junctions (NMJs). These structures are capped by terminal Schwann cells (tSCs), which play essential roles during formation and maintenance of the NMJ. tSCs are implicated in the correct communication between nerves and muscles, and in reinnervation upon injury. During aging, loss of muscle mass and strength (sarcopenia and dynapenia) are due, at least in part, to the progressive loss of contacts between muscle fibers and nerves. Despite the important role of tSCs in NMJ function, very little is known on their implication in the NMJ-aging process and in age-associated denervation. This review summarizes the current knowledge about the implication of tSCs in the age-associated degeneration of NMJs. We also speculate on the possible mechanisms underlying the observed phenotypes.
    Keywords:  aging; frailty; muscle denervation; neuromuscular junction (NMJ); peripheral nervous system; sarcopenia; terminal Schwann cell (tSC)
    DOI:  https://doi.org/10.14336/AD.2020.0708
  23. Sci Rep. 2021 Apr 05. 11(1): 7505
      Desmin is a muscle-specific intermediate filament protein that has fundamental role in muscle structure and force transmission. Whereas human desmin protein is encoded by a single gene, two desmin paralogs (desma and desmb) exist in zebrafish. Desma and desmb show differential spatiotemporal expression during zebrafish embryonic and larval development, being similarly expressed in skeletal muscle until hatching, after which expression of desmb shifts to gut smooth muscle. We generated knockout (KO) mutant lines carrying loss-of-function mutations for each gene by using CRISPR/Cas9. Mutants are viable and fertile, and lack obvious skeletal muscle, heart or intestinal defects. In contrast to morphants, knockout of each gene did not cause any overt muscular phenotype, but did alter calcium flux in myofibres. These results point to a possible compensation mechanism in these mutant lines generated by targeting nonsense mutations to the first coding exon.
    DOI:  https://doi.org/10.1038/s41598-021-86974-w
  24. FEBS Lett. 2021 Apr 08.
      We have previously shown evidence that α-syntrophin plays an important role in myoblast differentiation. In this study, we focused on abnormal myotube formation of the α-syntrophin-knockdown C2 cell line (SNKD). The overall amount of intracellular protein as well as muscle-specific proteins in SNKD cells were significantly lower than those in the control. Akt-mTOR signaling, an important pathway for protein synthesis and muscle hypertrophy, was downregulated. In addition, the levels of endoplasmic reticulum (ER) stress markers increased in SNKD cells. The decrease in intracellular protein synthesis and reduction of the myotube diameter in SNKD cells were restored by 4-phenylbutyric acid, a chemical chaperone, or overexpression of α-syntrophin. These results suggest a novel role for α-syntrophin in protein homeostasis during myoblast differentiation.
    Keywords:  Endoplasmic reticulum stress; Muscle differentiation; Protein homeostasis; Protein synthesis; α-Syntrophin
    DOI:  https://doi.org/10.1002/1873-3468.14088
  25. J Gen Physiol. 2021 May 03. pii: e202012701. [Epub ahead of print]153(5):
      Cannabidiol (CBD) is the primary nonpsychotropic phytocannabinoid found in Cannabis sativa, which has been proposed to be therapeutic against many conditions, including muscle spasms. Among its putative targets are voltage-gated sodium channels (Navs), which have been implicated in many conditions. We investigated the effects of CBD on Nav1.4, the skeletal muscle Nav subtype. We explored direct effects, involving physical block of the Nav pore, as well as indirect effects, involving modulation of membrane elasticity that contributes to Nav inhibition. MD simulations revealed CBD's localization inside the membrane and effects on bilayer properties. Nuclear magnetic resonance (NMR) confirmed these results, showing CBD localizing below membrane headgroups. To determine the functional implications of these findings, we used a gramicidin-based fluorescence assay to show that CBD alters membrane elasticity or thickness, which could alter Nav function through bilayer-mediated regulation. Site-directed mutagenesis in the vicinity of the Nav1.4 pore revealed that removing the local anesthetic binding site with F1586A reduces the block of INa by CBD. Altering the fenestrations in the bilayer-spanning domain with Nav1.4-WWWW blocked CBD access from the membrane into the Nav1.4 pore (as judged by MD). The stabilization of inactivation, however, persisted in WWWW, which we ascribe to CBD-induced changes in membrane elasticity. To investigate the potential therapeutic value of CBD against Nav1.4 channelopathies, we used a pathogenic Nav1.4 variant, P1158S, which causes myotonia and periodic paralysis. CBD reduces excitability in both wild-type and the P1158S variant. Our in vitro and in silico results suggest that CBD may have therapeutic value against Nav1.4 hyperexcitability.
    DOI:  https://doi.org/10.1085/jgp.202012701
  26. Nat Commun. 2021 04 07. 12(1): 2091
      Complex animals build specialised muscles to match specific biomechanical and energetic needs. Hence, composition and architecture of sarcomeres and mitochondria are muscle type specific. However, mechanisms coordinating mitochondria with sarcomere morphogenesis are elusive. Here we use Drosophila muscles to demonstrate that myofibril and mitochondria morphogenesis are intimately linked. In flight muscles, the muscle selector spalt instructs mitochondria to intercalate between myofibrils, which in turn mechanically constrain mitochondria into elongated shapes. Conversely in cross-striated leg muscles, mitochondria networks surround myofibril bundles, contacting myofibrils only with thin extensions. To investigate the mechanism causing these differences, we manipulated mitochondrial dynamics and found that increased mitochondrial fusion during myofibril assembly prevents mitochondrial intercalation in flight muscles. Strikingly, this causes the expression of cross-striated muscle specific sarcomeric proteins. Consequently, flight muscle myofibrils convert towards a partially cross-striated architecture. Together, these data suggest a biomechanical feedback mechanism downstream of spalt synchronizing mitochondria with myofibril morphogenesis.
    DOI:  https://doi.org/10.1038/s41467-021-22058-7
  27. Sports Med. 2021 Apr 05.
      BACKGROUND: The fatigue of a muscle or muscle group can produce global responses to a variety of systems (i.e., cardiovascular, endocrine, and others). There are also reported strength and endurance impairments of non-exercised muscles following the fatigue of another muscle; however, the literature is inconsistent.OBJECTIVE: To examine whether non-local muscle fatigue (NLMF) occurs following the performance of a fatiguing bout of exercise of a different muscle(s).
    DESIGN: Systematic review and meta-analysis.
    SEARCH AND INCLUSION: A systematic literature search using a Boolean search strategy was conducted with PubMed, SPORTDiscus, Web of Science, and Google Scholar in April 2020, and was supplemented with additional 'snowballing' searches up to September 2020. To be included in our analysis, studies had to include at least one intentional performance measure (i.e., strength, endurance, or power), which if reduced could be considered evidence of muscle fatigue, and also had to include the implementation of a fatiguing protocol to a location (i.e., limb or limbs) that differed to those for which performance was measured. We excluded studies that measured only mechanistic variables such as electromyographic activity, or spinal/supraspinal excitability. After search and screening, 52 studies were eligible for inclusion including 57 groups of participants (median sample = 11) and a total of 303 participants.
    RESULTS: The main multilevel meta-analysis model including all effects sizes (278 across 50 clusters [median = 4, range = 1 to 18 effects per cluster) revealed a trivial point estimate with high precision for the interval estimate [- 0.02 (95% CIs = - 0.14 to 0.09)], yet with substantial heterogeneity (Q(277) = 642.3, p < 0.01), I2 = 67.4%). Subgroup and meta-regression analyses showed that NLMF effects were not moderated by study design (between vs. within-participant), homologous vs. heterologous effects, upper or lower body effects, participant training status, sex, age, the time of post-fatigue protocol measurement, or the severity of the fatigue protocol. However, there did appear to be an effect of type of outcome measure where both strength [0.11 (95% CIs = 0.01-0.21)] and power outcomes had trivial effects [- 0.01 (95% CIs = - 0.24 to 0.22)], whereas endurance outcomes showed moderate albeit imprecise effects [- 0.54 (95% CIs = - 0.95 to - 0.14)].
    CONCLUSIONS: Overall, the findings do not support the existence of a general NLMF effect; however, when examining specific types of performance outcomes, there may be an effect specifically upon endurance-based outcomes (i.e., time to task failure). However, there are relatively fewer studies that have examined endurance effects or mechanisms explaining this possible effect, in addition to fewer studies including women or younger and older participants, and considering causal effects of prior training history through the use of longitudinal intervention study designs. Thus, it seems pertinent that future research on NLMF effects should be redirected towards these still relatively unexplored areas.
    DOI:  https://doi.org/10.1007/s40279-021-01456-3
  28. Front Bioeng Biotechnol. 2021 ;9 650289
      Volumetric muscle loss (VML) injuries after extremity trauma results in an important clinical challenge often associated with impaired healing, significant fibrosis, and long-term pain and functional deficits. While acute muscle injuries typically display a remarkable capacity for regeneration, critically sized VML defects present a dysregulated immune microenvironment which overwhelms innate repair mechanisms leading to chronic inflammation and pro-fibrotic signaling. In this series of studies, we developed an immunomodulatory biomaterial therapy to locally modulate the sphingosine-1-phosphate (S1P) signaling axis and resolve the persistent pro-inflammatory injury niche plaguing a critically sized VML defect. Multiparameter pseudo-temporal 2D projections of single cell cytometry data revealed subtle distinctions in the altered dynamics of specific immune subpopulations infiltrating the defect that were critical to muscle regeneration. We show that S1P receptor modulation via nanofiber delivery of Fingolimod (FTY720) was characterized by increased numbers of pro-regenerative immune subsets and coincided with an enriched pool of muscle stem cells (MuSCs) within the injured tissue. This FTY720-induced priming of the local injury milieu resulted in increased myofiber diameter and alignment across the defect space followed by enhanced revascularization and reinnervation of the injured muscle. These findings indicate that localized modulation of S1P receptor signaling via nanofiber scaffolds, which resemble the native extracellular matrix ablated upon injury, provides great potential as an immunotherapy for bolstering endogenous mechanisms of regeneration following VML injury.
    Keywords:  immunomodulation; inflammation; regeneration; sphingolipid; tissue engineering
    DOI:  https://doi.org/10.3389/fbioe.2021.650289
  29. Regen Eng Transl Med. 2021 Mar;7(1): 1-9
      Rotator cuff tears (RCTs) are a common cause of disability and pain in the adult population. Despite the successful repair of the torn tendon, the delay between the time of injury and time of repair can cause muscle atrophy. The goal of the study was to engineer an electroconductive nanofibrous matrix with an aligned orientation to enhance muscle regeneration after rotator cuff (RC) repair. The electroconductive nanofibrous matrix was fabricated by coating Poly(3,4-ethylenedioxythiophene): poly(styrenesulfonate) (PEDOT:PSS) nanoparticles onto the aligned poly(ε-caprolactone) (PCL) electrospun nanofibers. The regenerative potential of the matrix was evaluated using two repair models of RCTs include acute and sub-acute. Sprague-Dawley rats (n=39) were randomly assigned to 1 of 8 groups. For the acute model, the matrix was implanted on supraspinatus muscle immediately after the injury. The repair surgery for the sub-acute model was conducted 6 weeks after injury. The supraspinatus muscle was harvested for histological analysis two and six weeks after repair. The results demonstrated the efficacy of electrical and topographical cues on the treatment of muscle atrophy in vivo. In both acute and sub-acute models, the stimulus effects of topographical and electrical cues reduced the gap area between muscle fibers. This study showed that muscle atrophy can be alleviated by successful surgical repair using an electroconductive nanofibrous matrix in a rat RC model.
    Keywords:  Rotator cuff; electroconductive matrix; muscle atrophy; nanofibers
    DOI:  https://doi.org/10.1007/s40883-020-00186-8
  30. Mol Ther. 2021 Apr 03. pii: S1525-0016(21)00190-8. [Epub ahead of print]
      Motor neuron diseases are untreatable with common pharmacological approaches. Spinal Muscular Atrophy (SMA) is caused by SMN1 gene mutations leading to lowered SMN expression. Symptoms are alleviated in infants with a higher copy number of the SMN2 gene, which, however, displays a splicing defect resulting in low SMN levels. Amyotrophic Lateral Sclerosis (ALS) is caused by a number of mutations with C9orf72 repeat expansions the most common genetic cause and SOD1 gain-of-function mutations the first genetic cause identified for this disease. Genetic therapies based on oligonucleotides that enhance SMN2 splicing and SMN production or lower SOD1 expression have shown promise in initial clinical trials for SMA and ALS patients harboring SOD1 mutations, respectively. Gene addition/silencing approaches using AAV are also currently under clinical investigation in trials for SMA and ALS. Here, we provide a brief overview of these current efforts alongside with their advantages and challenges. We also review genome editing approaches aimed either at correcting the disease-causing mutations or at modulating the expression of genetic modifiers, e.g. by repairing SOD1 mutations or SMN2 splicing defect, or deleting C9orf72 expanded repeats. This recent and accumulating body of work has shown promising results to approach therapeutic trials that should significantly lower the progression of these deadly disorders.
    Keywords:  ALS; CRISPR; Motor neuron diseases; SMA; base editing; genome editing
    DOI:  https://doi.org/10.1016/j.ymthe.2021.04.003
  31. Physiol Rep. 2021 Apr;9(7): e14815
      OBJECTIVE: Cardiopulmonary exercise testing (CPET) is often used to assess pre-operative fitness in elderly patients, in whom peripheral arterial disease (PAD) is highly prevalent, but may affect the results of CPET by early lactate release due to muscle ischemia. This study investigated the effect revascularization of PAD on oxygen delivery (VO2 ) during CPET.METHOD: We conducted a prospective cohort study of 30 patients, who underwent CPET before and after treatment of ilio-femoral PAD. The primary outcome measure was difference in VO2 at the lactate threshold (LT) before and after revascularization. Secondary outcome measures were the relationship between change in VO2 at LT and peak exercise and change in ankle-brachial index (ABI) differential.
    RESULTS: The study was approved by the North West-Lancaster Research and Ethics committee (reference 15/NW/0801) and registered in clinicaltrial.gov (reference NCT02657278). As specified in the study protocol, 30 patients were recruited but only 20 (15 men), with a mean age of 62 years, completed pre- and post-treatment CPETs. Twelve patients demonstrated an improvement in VO2 at LT after revascularization, but the difference did not achieve statistical significance (mean difference (95% CI) = 1.43 (-0.21 to 3.08) ml/kg/min; (p = 0.085). There was, however, a significant improvement in VO2 , VE/CO2 , workload and Borg breathlessness and leg fatigue score at peak exercise after revascularization. There was no significant correlation between change in VO2 at LT (r = -0.11, p = 0.65) or change in VO2 at peak and ABI differential (r = -0.14, p = 0.55).
    CONCLUSION: Revascularization of PAD led to significant improvement in multiple peak/maximal exercise parameters within a few weeks and without exercise training. We were unable to demonstrate a statistically significant improvement in VO2 at LT albeit in a majority of subjects this exceeded what we pre-defined as clinically significant.
    Keywords:  aerobic threshold; atherosclerosis; skeletal muscle
    DOI:  https://doi.org/10.14814/phy2.14815
  32. J Gerontol A Biol Sci Med Sci. 2021 Apr 09. pii: glab104. [Epub ahead of print]
      BACKGROUND: The ProMuscle in Practice intervention, comprising resistance exercise and an increased protein intake, was effective in improving muscle strength, lean body mass, and physical functioning in older adults aged ≥65 years (N=168). However, a heterogeneous response to such interventions is common. Therefore, we explored the differences in responsiveness to the intervention in subgroups based on demographic characteristics and mobility-impairing disorders.METHODS: Multiple regression analyses were performed to study mean changes between baseline and 12 weeks on the Short Physical Performance Battery, chair rise test, lean body mass, knee extension strength, leg press strength, and leg extension strength. The interaction term treatment x subgroup was included to study differences in effects between subgroups. Subgroups comprised age (≤75 vs. >75 years), sex (men vs. women), presence of frailty, presence of sarcopenia, and presence of osteoarthritis.
    RESULTS: A significant interaction effect including age was found on lean body mass (β=-0.8, 95% CI: -1.5; -0.2), favoring participants aged ≤75 years. A significant interaction effect including sex was found on leg press strength (β=15.5, 95% CI: 0.6; 30.3), favoring women. Participants with or without frailty, sarcopenia, or osteoarthritis responded equally to the intervention in terms of absolute effects.
    CONCLUSIONS: Participants aged ≤75 years and women benefited to a great extent from the intervention, as they improved significantly on nearly every outcome. Effects in participants with and without a mobility-impairing disorder were comparable, indicating that the intervention is suitable for both groups.
    Keywords:  Lifestyle intervention; muscle mass; muscle strength; physical functioning; responsiveness
    DOI:  https://doi.org/10.1093/gerona/glab104
  33. Mol Ther. 2021 Apr 03. pii: S1525-0016(21)00189-1. [Epub ahead of print]
      The emerging clustered regularly interspaced short palindromic repeats (CRISPR)-mediated genome editing technologies have progressed remarkably in recent years, opening up the potential of precise genome editing as a therapeutic approach to treat various diseases. The CRISPR/CRISPR associated (Cas) system is an attractive platform for the treatment of Duchenne muscular dystrophy (DMD), which is a neuromuscular disease caused by mutations in the DMD gene. CRISPR/Cas can be used to permanently repair the mutated DMD gene, leading to expression of the encoded protein, dystrophin, in systems ranging from cells derived from DMD patients to animal models of DMD. However, the development of more efficient therapeutic approaches and delivery methods remains a great challenge for DMD. Herein, we review various therapeutic strategies that use CRISPR/Cas to correct or bypass DMD mutations and discuss their therapeutic potential, as well as obstacles that lie ahead.
    Keywords:  CRISPR; Cas; Duchenne muscular dystrophy; genome editing; neuromuscular disorder
    DOI:  https://doi.org/10.1016/j.ymthe.2021.04.002