banner



How Does Muscle Tissue Repair Itself

  • Periodical List
  • J Exp Orthop
  • v.three; 2022 December
  • PMC4958098

J Exp Orthop. 2022 Dec; three: 15.

Muscle injuries and strategies for improving their repair

Thomas Laumonier

Department of Orthopaedic Surgery, Geneva University Hospitals & Faculty of Medicine, 4, Rue Gabrielle Perret-Gentil, 1211 Geneva 14, Switzerland

Jacques Menetrey

Department of Orthopaedic Surgery, Geneva University Hospitals & Faculty of Medicine, iv, Rue Gabrielle Perret-Gentil, 1211 Geneva fourteen, Switzerland

Received 2022 Mar 15; Accepted 2022 Jul 15.

Abstract

Satellite cells are tissue resident muscle stem cells required for postnatal skeletal musculus growth and repair through replacement of damaged myofibers. Muscle regeneration is coordinated through dissimilar mechanisms, which imply jail cell-cell and cell-matrix interactions besides every bit extracellular secreted factors. Cellular dynamics during musculus regeneration are highly complex. Immune, fibrotic, vascular and myogenic cells appear with distinct temporal and spatial kinetics afterward musculus injury. Three primary phases accept been identified in the process of muscle regeneration; a destruction stage with the initial inflammatory response, a regeneration phase with activation and proliferation of satellite cells and a remodeling phase with maturation of the regenerated myofibers. Whereas relatively small musculus injuries, such every bit strains, heal spontaneously, severe muscle injuries class fibrotic tissue that impairs musculus part and atomic number 82 to muscle contracture and chronic hurting. Current therapeutic approaches accept limited effectiveness and optimal strategies for such lesions are non known notwithstanding. Various strategies, including growth factors injections, transplantation of muscle stalk cells in combination or not with biological scaffolds, anti-fibrotic therapies and mechanical stimulation, may become therapeutic alternatives to improve functional muscle recovery.

Keywords: Skeletal muscle, Injury, Regeneration, Stem cell, Fibrosis, Scaffolds, Growth factors

Introduction

Human skeletal muscle is about 40 % of the body mass and is formed by bundle of contractile multinucleated muscle fibers, resulting from the fusion of myoblasts. Satellite cells (SC) are skeletal muscle stalk cell located between the plasma membrane of myofibers and the basal lamina. Their regenerative capabilities are essential to repair skeletal muscle subsequently injury (Hurme and Kalimo 1992; Lipton and Schultz 1979) (Sambasivan et al. 2022; Dumont et al. 2022a). In adult muscles, SC are found in a quiescent land and represent, depending on species, historic period, muscle location, and muscle type, around five to 10 % of skeletal muscle cells (Rocheteau et al. 2022). After injury, SC go activated, proliferate and give ascent to myogenic forerunner cells, known as myoblasts. After entering the differentiation process, myoblasts form new myotubes or fuse with damaged myofibers, ultimately mature in functional myofibers.

Skeletal muscle injuries can stem from a multifariousness of events, including straight trauma such equally muscle lacerations and contusions, indirect insults such as strains and also from degenerative diseases such as muscular dystrophies (Huard et al. 2002; Kasemkijwattana et al. 2000; Kasemkijwattana et al. 1998; Menetrey et al. 2000; Menetrey et al. 1999; Crisco et al. 1994; Garrett et al. 1984; Lehto and Jarvinen 1991; Jarvinen et al. 2005; Cossu and Sampaolesi 2007). Skeletal muscle can regenerate completely and spontaneously in response to minor injuries, such as strain. In contrast, after astringent injuries, muscle healing is incomplete, often resulting in the formation of fibrotic tissue that impairs muscle function. Although researchers accept extensively investigated various approaches to improve muscle healing, there is still no gold standard treatment.

This concise review provides a sight about the various phases of muscle repair and regeneration, namely degeneration, inflammation, regeneration, remodeling and maturation. We also give an overview of enquiry efforts that accept focused on the utilise of stem cell therapy, growth factors and/or biological scaffolds to ameliorate muscle regeneration and repair. We also accost the therapeutic potential of mechanical stimulation and of anti-fibrotic therapy to enhance muscle regeneration and repair.

Review

Musculus healing process

Skeletal muscle has a robust innate capability for repair after injury through the presence of adult musculus stalk cells known as satellite cells (SC). The disruption of muscle tissue homeostasis, caused by injury, generates sequential involvement of various players effectually three primary phases (Fig.1).

  • (1, two) Degeneration/inflammation phase: characterized past rupture and necrosis of the myofibers, formation of a hematoma and an of import inflammatory reaction.

  • (3) Regeneration phase: phagocytosis of damaged tissue, followed past myofibers regeneration, leading to satellite cell activation.

  • (4, 5) Remodeling phase: maturation of regenerated myofibers with recovery of muscle functional capacity (four) and also fibrosis and scar tissue formation (5).

An external file that holds a picture, illustration, etc.  Object name is 40634_2016_51_Fig1_HTML.jpg

Sequential cycle of muscle healing phases after laceration. Histological images adapted from Menetrey et al, Am J Sports Med 1999. (sp: superficial portion, de: deepest part)

Muscle degeneration and inflammation

Active musculus degeneration and inflammation occur inside the offset few days after injury. The initial consequence is necrosis of the musculus fibers, which is triggered by disruption of local homeostasis and particularly past unregulated influx of calcium through sarcolemma lesions (Tidball 2022). Backlog in cytoplasmic calcium causes proteases and hydrolases activation that contribute to musculus damage and also causes activation of enzymes that drive the product of mitogenic substances for muscle and allowed cells (Tidball 2005). Later musculus degeneration, neutrophils are the first inflammatory cells infiltrating the lesion. A large number of pro-inflammatory molecules such as cytokines (TNF-α, IL-6), chemokine (CCL17, CCL2) and growth factors (FGF, HGF, IGF-I, VEGF; TGF-β1) are secreted by neutrophils in social club to create a chemoattractive microenvironment for other inflammatory cells such as monocytes and macrophages (Tidball 1995; Toumi and Best 2003). Two types of macrophages are identified during muscle regeneration (McLennan 1996), which announced sequentially during muscle repair (Arnold et al. 2007). M1 macrophages, defined as pro-inflammatory macrophages, act during the first few days after injury,. contribute to cell lysis, removal of cellular droppings and stimulate myoblast proliferation. Conversely, M2 macrophages, defined every bit anti-inflammatory macrophages, act 2 to 4 days after injury, attenuate the inflammatory response and favor musculus repair past promoting myotubes formation (Tidball and Wehling-Henricks 2007; Chazaud 2022; Chazaud et al. 2003). Macrophages, infiltrating injured muscle, are fundamental players of the healing process (Zhao et al. 2022), able to participate in the musculus regeneration process or to favor fibrosis (Munoz-Canoves and Serrano 2022; Lemos et al. 2022).

Muscle regeneration, remodeling and maturation

Muscle regeneration ordinarily starts during the first 4–v days afterwards injury, peaks at 2 weeks, and and so gradually diminishes 3 to 4 weeks later injury. It's a multiple steps process including activation/proliferation of SC, repair and maturation of damaged muscle fibers and connective tissue formation. A fine residuum between these mechanisms is essential for a total recovery of the contractile muscle function.

Muscle fibers are post-mitotic cells, which do not have the capacity to divide. Post-obit an injury, damaged muscle fibers can't be repaired without the presence of adult muscle stem cells, the satellite cells (SC) (Relaix and Zammit 2022; Sambasivan et al. 2022). Following activation, SC proliferate and generate a population of myoblasts that can either differentiate to repair damaged fibers or, for a modest proportion, self-renew to maintain the SC pool for possible futurity demands of muscle regeneration (Collins 2006; Dhawan and Rando 2005). SC bike progression and jail cell fate determination are control past circuitous regulatory mechanisms in which, intrinsic and extrinsic factors are involved (Dumont et al. 2022a; Dumont et al. 2022b).

Connective tissue/fibrosis

Connective tissue remodeling is an important step of the regenerative muscle process. Chop-chop later on muscle injury, a gap is formed betwixt damaged musculus fibers and filled with a hematoma. Muscle injuries can be clinically classified depending of the nature of the hematoma (size, location). Tardily elimination of the hematoma is known to delay skeletal muscle regeneration, to improve fibrosis and to reduce biomechanical properties of the healing musculus (Beiner et al. 1999). In rare complication, major musculus injuries may atomic number 82 to the development of myositis ossificans that will impair muscle regeneration and repair (Beiner and Jokl 2002) (Walczak et al. 2022).

The presence of fibrin and fibronectin at the injury site, initiate the germination of an extracellular matrix that is rapidly invaded by fibroblasts (Darby et al. 2022; Desmouliere and Gabbiani 1995). Fibrogenic cytokines such as transforming growth factor β1 (TGF-β1) participate to excessive fibroblasts/myofibroblasts proliferation and to an increment in type I/III collagens, laminin and fibronectin production (Lehto et al. 1985). In its initial phase, the fibrotic response is beneficial, stabilizing the tissue and acting as a scaffold for myofibers regeneration. Nevertheless, an excessive collagen synthesis mail service injury, oftentimes outcome in an increase of scar tissue size over time that can prevent normal muscle function (Mann et al. 2022). Many growth factors are involved in the development of fibrosis, such as Connective Tissue Growth Factor (CTGF), Platelet-Derived Growth Factor (PDGF) or myostatin. TGF-β1, by stimulating fibroblasts/myofibroblasts to produce extracellular proteins such as fibronectin and type I/Iii collagen, has been identified as the cardinal element in this process (Mann et al. 2022),. Although fibroblasts are the major collagen-producing cells in skeletal muscle, TGF-β1 have besides an upshot directly on myoblasts causing their conversion to myofibroblasts. Thus myoblasts initially interim to repair damaged myofibers, will produce significant level of collagen and will contribute to muscle fibrosis (Li and Huard 2002).

R evascularization

The restoration of the blood supply in the injured skeletal muscle is one of the outset signs of musculus regeneration and is essential to its success. Without revascularization, muscle regeneration is incomplete and a significant fibrosis occurs (Best et al. 2022; Ota et al. 2022). Afterwards muscle trauma, blood vessels rupture induces tissue hypoxia at the injury site (Jarvinen et al. 2005). New capillaries formation rapidly after injury is therefore necessary (Scholz et al. 2003) for a functional muscle recovery. Secretion of angiogenic factors such every bit vascular endothelial growth factor (VEGF) at the lesion site is important and several studies have shown that VEGF, by favoring angiogenesis, improve skeletal muscle repair (Deasy et al. 2009; Frey et al. 2022).

Innervation

Muscle repair is complete when injured myofibers are fully regenerated and become innervated. The synaptic contact between a motor neuron and its target muscle fiber, often accept place at a specific site in the central region of myofibers, the neuromuscular junction (NMJ) (Wu et al. 2010). NMJ are essential for maturation and functional activity of regenerating muscles. Within 2–iii weeks afterwards muscle damage, the presence of newly formed NMJ is observed in regenerative muscle (Rantanen et al. 1995; Vaittinen et al. 2001).

Strategies to improve muscle regeneration and repair

Growth factors

Growth factors play a variety of roles in the different stages of muscle regeneration (Grounds 1999; Menetrey et al. 2000). These biologically active molecules, synthetized by the injured tissue or by other cell types present at the inflammatory site, are release in the extracellular infinite and modulate the regenerative response (Tabular arrayi). Although hepatocyte growth factor (HGF), fibroblast growth cistron (FGF) and platelet-derived growth cistron (PDGF) are of involvement considering of their chapters to stimulate satellite cells (Sheehan et al. 2000; Allen and Boxhorn 1989; Yablonka-Reuveni et al. 1990), insulin like growth factor-1 (IGF-I) appears to be of particular importance for the muscle regeneration procedure. IGF-I stimulates myoblasts proliferation and differentiation (Engert et al. 1996) and is implicated in the regulation of muscle growth (Schiaffino and Mammucari 2022). In a mouse model, straight injections of man recombinant IGF-I at ii, v, and vii days after injury enhanced musculus healing in lacerated, contused, and strain-injured muscles (Menetrey et al. 2000; Kasemkijwattana et al. 2000). However, the efficacy of directly injection of recombinant proteins is express by the high concentration of the cistron typically required to elicit a measurable issue. This is mainly due to the bloodstream's rapid clearance of these molecules and their relatively short biological half-lives. Gene therapy may be an effective method past which to deliver high, maintainable concentrations of growth factor to injured muscle (Barton-Davis et al. 1998; Barton et al. 2002; Musaro et al. 2001). Although IGF-I improved muscle healing, histology of the injected muscle revealed fibrosis within the lacerated site, despite high level of IGF-I production (Lee et al. 2000). Some other growth factor, VEGF, past favoring angiogenesis, is known to enhance skeletal muscle repair (Deasy et al. 2009; Frey et al. 2022; Messina et al. 2007). By targeting simultaneously angiogenesis and myogenesis, it was shown that combined delivery of VEGF and IGF-I enhance muscle regenerative process (Borselli et al. 2010). In this direction, the use of platelet-rich plasma (PRP) is considered every bit a possible alternative approach based on the power of autologous growth factors to meliorate skeletal muscle regeneration (Hamid et al. 2022; Hammond et al. 2009). Considered as safe products, autologous PRP injections are increasingly used in patients with sports-related injuries (Engebretsen et al. 2010). Notwithstanding, a recent randomized clinical trial show no significant positive effects of PRP injections, as compared with placebo injections, in patients with muscle injuries, upward to one year after injections (Reurink et al. 2022; Reurink et al. 2022). Customization of PRP preparation, equally recently demonstrated by the use of TGF-β1 neutralizing antibodies, is a promising alternative to promote muscle regeneration while significantly reducing fibrosis (Li et al. 2022).

Tabular array i

The role of growth factors in skeletal muscle regeneration

Growth factors Physiological effects, potential benefits Shortcomings Commentary
IGF-1 - Essential for muscle growth during development and regeneration.
- Promote myoblast proliferation and differentiation in vitro (Huard et al. 2002)
- Hypertrophic outcome of IGF-1 (Barton-Davis et al. 1999)
- Serial injections of IGF-ane improve musculus healing in vivo (Menetrey et al. 2000).
- Existence of a muscle specific isoform of IGF-i (mIGF-1) (Musaro et al. 1999; Musaro et al. 2004)
- Chemotactic for fibroblasts, increase collagen production, enhance fibrosis development - IGF-i play a primal role in the enhancement of muscle regeneration-
- Anti-inflammatory actions of IGF-i (Mourkioti and Rosenthal 2005; Tidball and Welc 2022)
HGF - Promote myoblast proliferation and inhibit myoblast differentiation (Anderson 2022; Yin et al. 2022)
- Of import role for satellite prison cell activation. Balance between the activation of satellite cells and their return to quiescence. (Chazaud 2010)
- Recently, information technology was shown that a 2nd set of HGF production is crucial for inflammation resolution subsequently injury (Proto et al. 2022)
- Injection of HGF into injured muscle increased myoblast numbers only blocked the regeneration process (Miller et al. 2000) - HGF is of import during the early phase of muscle regeneration, actuate satellite cells
VEGF - Of import signaling protein that favor angiogenesis.
- Promote myoblast migration, proliferation and survival. (Arsic et al. 2004)
- VEGF assistants improves muscle regeneration. (Messina et al. 2007; Deasy et al. 2009)
- Non regulated VEGF expression promote abnormal angiogenesis and fibrosis in skeletal muscle (Karvinen et al. 2022) - Importance of the proximity betwixt satellite cells and the microvasculature during musculus regeneration, role of VEGF
FGF - Large family of mitogen involved in prison cell growth and survival
- FGF-half-dozen has a muscle specific expression, stimulates satellite cell proliferation and promotes myogenic terminal differentiation (Floss et al. 1997)
- FGF-2 promote satellite cell proliferation and inhibit myogenic differentiation (Menetrey et al. 2000; Kastner et al. 2000)
- Stimulate fibroblast proliferation, - FGF signaling plays a primal role in muscle repair, blocking FGF signaling delay muscle regeneration (Saera-Vila et al. 2022).
TGF-β1 - Primal regulator of the balance betwixt muscle fibrosis and muscle regeneration
- Inhibits satellite cell proliferation and differentiation in vitro
- Excessive TGFβ1-induced deposition of ECM at the site of injury, fibrosis (Garg et al. 2022). - Anti fibrotic therapy past blocking overexpression of TGF-β1 amend musculus regeneration. (Burks et al. 2022; Hwang et al. 2022)
PDGF-BB - PDGF isoforms can regulate myoblast proliferation and differentiation in vitro (Yablonka-Reuveni et al. 1990)
- PDGF-BB stimulates satellite cell proliferation and inhibit their differentiation (Accuse and Rudnicki 2004)
- Stiff mitogen for fibroblasts - Release from injured vessels and platelets, PDGF stimulates early on skeletal musculus regeneration

Stem cells

Transplantation of satellite cell-derived myoblasts has long been explored equally a promising approach for treatment of skeletal musculus disorders. After an initial demonstration that normal myoblasts can restore dystrophin expression in mdx mice (Partridge et al. 1989), clinical trials, in which allogeneic normal homo myoblasts were injected intramuscularly several times in dystrophic young boys muscles, accept not been successful (Law et al. 1990; Mendell et al. 1995). Fifty-fifty recently, despite articulate improvement in methodologies that raise the success of myoblast transplantation in Duchenne patients (Skuk et al. 2007), outcomes of clinical trials are still disappointing. These experiments have raised concerns virtually the express migratory and proliferative capacities of homo myoblasts, as well equally their limited life span in vivo. It led to the investigations of other muscle stem cells sources that could overcome these limitations and outperform the success of muscle cell transplantation. Among all these non-satellite myogenic stalk cells, human mesoangioblasts, human being myogenic-endothelial cells and homo muscle–derived CD133+ accept shown myogenic potentials in vitro and in vivo (Sampaolesi et al. 2006; Zheng et al. 2007; Meng et al. 2022). The utilize of such myogenic progenitors cells for improving musculus healing may become an interesting therapeutic alternative (Tedesco and Cossu 2022; Tedesco et al. 2010; Chen et al. 2022). A beginning stage I/IIa clinical trial has recently demonstrated that intra arterial injections of homo mesoangioblasts are safe but display only very limited clinical efficacy in Duchenne patients (Cossu et al. 2022).

Scaffolds

Myogenic forerunner cell survival and migration is greatly increased by using appropriate scaffold composition and growth factor commitment (Hill et al. 2006) (Boldrin et al. 2007). Controlling the microenvironment of injected myogenic cells using biological scaffolds enhance musculus regeneration (Borselli et al. 2022). Ideally, using an appropriate extracellular matrix (ECM) composition and stiffness, scaffolds should best replicate the in vivo milieu and mechanical microenvironment (Gilbert et al. 2010) (Engler et al. 2006). A combination of stem cells, biomaterial-based scaffolds and growth factors may provide a therapeutic pick to improve regeneration of injured skeletal muscles (Jeon and Elisseeff 2022).

Anti-fibrotic therapy

TGF-β1 is expressed at high levels and plays an of import role in the fibrotic cascade that occurs after the onset of muscle injury (Bernasconi et al. 1995; Li et al. 2004). Therefore, neutralization of TGF-β1 expression in injured skeletal musculus should inhibit the formation of scar tissue. Indeed, the employ of anti-fibrotic agents (ie decorin, relaxin, antibody against TGF-β1…) that inactivate TGF-β1 signaling pathways reduces muscle fibrosis and, consequently, meliorate muscle healing, leading to a well-nigh complete recovery of lacerated muscle (Fukushima et al. 2001; Li et al. 2007). Losartan, an angiotensin II receptor antagonist, neutralize the outcome of TGF-β1 and reduce fibrosis, making information technology the treatment of choice, since information technology already has FDA approval to be used clinically (Bedair et al. 2008; Park et al. 2022; Terada et al. 2022). Suramin, also canonical by the FDA, blocks TGF-β1 pathway and reduces muscle fibrosis in experimental model (Chan et al. 2003; Taniguti et al. 2022).

Mechanical stimulation

Mechanical stimulation may offer a simple and effective approach to enhance skeletal musculus regeneration. Stretch activation, mechanical conditioning but also massage therapy or concrete manipulation of injured skeletal muscles have shown multiple do good effects on musculus biological science and function in vitro and in vivo (Tatsumi et al. 2001);(Best et al. 2022) (Crane et al. 2022; Kumar et al. 2002; Gilbert et al. 2010; Powell et al. 2002). Recently, Cezar and colleagues demonstrates that mechanical forces are equally of import biological regulators as chemicals and genes, and underlines the immense potential of developing mechano-therapies to treat musculus damage (Cezar et al. 2022). A recent study also demonstrated that a treatment based on ultrasound-guided intra-tissue percutaneous electrolysis (EPI technique) enhances the treatment of muscle injuries (Abat et al. 2022). Altogether, these results propose that mechanical stimulation should exist considered as a possible therapy to improve muscle regeneration and repair.

Conclusions

Skeletal muscle injuries are very frequently present in sports medicine and pose challenging problems in traumatology. Despite their clinical importance, the optimal rehabilitation strategies for treating these injuries are non well defined. Later on a trauma, skeletal muscles have the capacity to regenerate and repair in a complex and well-coordinated response. This process required the presence of diverse prison cell populations, up and down-regulation of diverse gene expressions and participation of multiples growth factors. Strategies based on the combination of stem cells, growth factors and biological scaffolds take already shown promising results in animal models. A better understanding of the cellular and molecular pathways also as a improve definition of the interactions (prison cell-cell and cell-matrix) that are essential for constructive muscle regeneration, should contribute to the development of new therapies in humans. In this direction, a recent newspaper from Sadtler et al demonstrated that specific biological scaffold implanted in injured mice muscles trigger a pro-regenerative immune response that stimulate skeletal muscle repair (Sadtler et al. 2022).

Abridgement

CTGF, connective tissue growth factor; FGF, fibroblast growth factor; HGF, hepatocyte growth cistron; IGF-I, insulin similar growth gene-I; NMJ, neuromuscular junction; PDGF, platelet derived growth factor; PRP, platelet rich plasma; SC, satellite cells; TGF-β1, transforming growth factor β1; VEGF, vascular endothelial growth factor

Footnotes

Competing interests

The authors declare that they take no competing interests.

Authors' contributions

TL and JM participated equally in drafting the manuscript. Both authors read and approved the concluding manuscript.

Contributor Information

Thomas Laumonier, Phone: +41 22 3795393, hc.eginu@reinomual.samoht.

Jacques Menetrey, hc.eguch@yertenem.seuqcaj.

References

  • Abat F, Valles SL, Gelber PE, Polidori F, Jorda A, Garcia-Herreros S, Monllau JC, Sanchez-Ibanez JM. An experimental study of muscular injury repair in a mouse model of notexin-induced lesion with EPI(R) technique. BMC Sports Sci Med Rehabil. 2015;vii:7. doi: 10.1186/s13102-015-0002-0. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Allen RE, Boxhorn LK. Regulation of skeletal muscle satellite cell proliferation and differentiation by transforming growth factor-beta, insulin-similar growth factor I, and fibroblast growth gene. J Prison cell Physiol. 1989;138(ii):311–315. doi: x.1002/jcp.1041380213. [PubMed] [CrossRef] [Google Scholar]
  • Anderson JE. Hepatocyte growth gene and satellite cell activation. Adv Exp Med Biol. 2016;900:1–25. doi: 10.1007/978-iii-319-27511-6_1. [PubMed] [CrossRef] [Google Scholar]
  • Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, Gherardi RK, Chazaud B. Inflammatory monocytes recruited after skeletal musculus injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med. 2007;204(five):1057–1069. doi: x.1084/jem.20070075. [PMC complimentary article] [PubMed] [CrossRef] [Google Scholar]
  • Arsic N, Zacchigna S, Zentilin 50, Ramirez-Correa G, Pattarini Fifty, Salvi A, Sinagra G, Giacca Yard. Vascular endothelial growth factor stimulates skeletal muscle regeneration in vivo. Mol Ther. 2004;10(5):844–854. doi: x.1016/j.ymthe.2004.08.007. [PubMed] [CrossRef] [Google Scholar]
  • Barton ER, Morris 50, Musaro A, Rosenthal N, Sweeney HL. Muscle-specific expression of insulin-like growth factor I counters musculus reject in mdx mice. J Jail cell Biol. 2002;157(1):137–148. doi: 10.1083/jcb.200108071. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • Barton-Davis ER, Shoturma DI, Musaro A, Rosenthal N, Sweeney HL. Viral mediated expression of insulin-similar growth factor I blocks the aging-related loss of skeletal muscle function. Proc Natl Acad Sci U S A. 1998;95(26):15603–15607. doi: 10.1073/pnas.95.26.15603. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
  • Barton-Davis ER, Shoturma DI, Sweeney HL. Contribution of satellite cells to IGF-I induced hypertrophy of skeletal musculus. Acta Physiol Scand. 1999;167(iv):301–305. doi: ten.1046/j.1365-201x.1999.00618.x. [PubMed] [CrossRef] [Google Scholar]
  • Bedair HS, Karthikeyan T, Quintero A, Li Y, Huard J. Angiotensin Ii receptor occludent administered after injury improves muscle regeneration and decreases fibrosis in normal skeletal muscle. Am J Sports Med. 2008;36(viii):1548–1554. doi: 10.1177/0363546508315470. [PubMed] [CrossRef] [Google Scholar]
  • Beiner JM, Jokl P (2002) Musculus contusion injury and myositis ossificans traumatica. Clin Orthop Relat Res (403 Suppl):S110-119 [PubMed]
  • Beiner JM, Jokl P, Cholewicki J, Punjabi MM. The effect of anabolic steroids and corticosteroids on healing of muscle contusion injury. Am J Sports Med. 1999;27(1):two–9. [PubMed] [Google Scholar]
  • Bernasconi P, Torchiana E, Confalonieri P, Brugnoni R, Barresi R, Mora M, Cornelio F, Morandi Fifty, Mantegazza R. Expression of transforming growth factor-beta 1 in dystrophic patient muscles correlates with fibrosis. Pathogenetic role of a fibrogenic cytokine. J Clin Invest. 1995;96(two):1137–1144. doi: 10.1172/JCI118101. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
  • Best TM, Gharaibeh B, Huard J. Stem cells, angiogenesis and muscle healing: a potential role in massage therapies? Br J Sports Med. 2012 [PubMed] [Google Scholar]
  • Boldrin Fifty, Elvassore N, Malerba A, Flaibani M, Cimetta E, Piccoli M, Baroni Physician, Gazzola MV, Messina C, Gamba P, Vitiello L, de Coppi P. Satellite cells delivered by micro-patterned scaffolds: a new strategy for prison cell transplantation in musculus diseases. Tissue Eng. 2007;13(2):253–262. doi: ten.1089/ten.2006.0093. [PubMed] [CrossRef] [Google Scholar]
  • Borselli C, Storrie H, Benesch-Lee F, Shvartsman D, Cezar C, Lichtman JW, Vandenburgh HH, Mooney DJ. Functional musculus regeneration with combined delivery of angiogenesis and myogenesis factors. Proc Natl Acad Sci U S A. 2010;107(8):3287–3292. doi: x.1073/pnas.0903875106. [PMC costless commodity] [PubMed] [CrossRef] [Google Scholar]
  • Borselli C, Cezar CA, Shvartsman D, Vandenburgh HH, Mooney DJ. The part of multifunctional delivery scaffold in the power of cultured myoblasts to promote muscle regeneration. Biomaterials. 2011;32(34):8905–8914. doi: 10.1016/j.biomaterials.2011.08.019. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Burks TN, Andres-Mateos East, Marx R, Mejias R, Van Erp C, Simmers JL, Walston JD, Ward CW, Cohn RD. Losartan restores skeletal muscle remodeling and protects against disuse atrophy in sarcopenia. Sci Transl Med. 2011;three(82):82ra37. doi: 10.1126/scitranslmed.3002227. [PMC complimentary article] [PubMed] [CrossRef] [Google Scholar]
  • Cezar CA, Roche ET, Vandenburgh HH, Duda GN, Walsh CJ, Mooney DJ. Biologic-free mechanically induced muscle regeneration. Proc Natl Acad Sci U S A. 2016;113(6):1534–1539. doi: 10.1073/pnas.1517517113. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
  • Chan YS, Li Y, Foster Westward, Horaguchi T, Somogyi K, Fu FH, Huard J. Antifibrotic effects of suramin in injured skeletal muscle subsequently laceration. J Appl Physiol. 2003;95(2):771–780. doi: 10.1152/japplphysiol.00915.2002. [PubMed] [CrossRef] [Google Scholar]
  • Charge SB, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiol Rev. 2004;84(1):209–238. doi: ten.1152/physrev.00019.2003. [PubMed] [CrossRef] [Google Scholar]
  • Chazaud B. Dual effect of HGF on satellite/myogenic cell quiescence. Focus on "High concentrations of HGF inhibit skeletal muscle satellite cell proliferation in vitro past inducing expression of myostatin: a possible machinery for reestablishing satellite prison cell quiescence in vivo". Am J Physiol Cell Physiol. 2010;298(iii):C448–C449. doi: 10.1152/ajpcell.00561.2009. [PubMed] [CrossRef] [Google Scholar]
  • Chazaud B. Macrophages: supportive cells for tissue repair and regeneration. Immunobiology. 2014;219(iii):172–178. doi: 10.1016/j.imbio.2013.09.001. [PubMed] [CrossRef] [Google Scholar]
  • Chazaud B, Sonnet C, Lafuste P, Bassez Thou, Rimaniol AC, Poron F, Authier FJ, Dreyfus PA, Gherardi RK. Satellite cells attract monocytes and use macrophages equally a support to escape apoptosis and heighten muscle growth. J Cell Biol. 2003;163(5):1133–1143. doi: 10.1083/jcb.200212046. [PMC gratis article] [PubMed] [CrossRef] [Google Scholar]
  • Chen CW, Corselli M, Peault B, Huard J. Human blood-vessel-derived stalk cells for tissue repair and regeneration. J Biomed Biotechnol. 2012;2012:597439. [PMC costless article] [PubMed] [Google Scholar]
  • Collins CA. Satellite jail cell cocky-renewal. Curr Opin Pharmacol. 2006;6(3):301–306. doi: ten.1016/j.coph.2006.01.006. [PubMed] [CrossRef] [Google Scholar]
  • Cossu G, Sampaolesi M. New therapies for Duchenne muscular dystrophy: challenges, prospects and clinical trials. Trends Mol Med. 2007;13(12):520–526. doi: 10.1016/j.molmed.2007.10.003. [PubMed] [CrossRef] [Google Scholar]
  • Cossu G, Previtali SC, Napolitano S, Cicalese MP, Tedesco FS, Nicastro F, Noviello Grand, Roostalu U, Natali Sora MG, Scarlato Chiliad, De Pellegrin M, Godi C, Giuliani S, Ciotti F, Tonlorenzi R, Lorenzetti I, Rivellini C, Benedetti S, Gatti R, Marktel Southward, Mazzi B, Tettamanti A, Ragazzi M, Imro MA, Marano G, Ambrosi A, Fiori R, Sormani MP, Bonini C, Venturini M, Politi LS, Torrente Y, Ciceri F. Intra-arterial transplantation of HLA-matched donor mesoangioblasts in Duchenne muscular dystrophy. EMBO Mol Med. 2015 [PMC free article] [PubMed] [Google Scholar]
  • Crane JD, Ogborn DI, Cupido C, Melov S, Hubbard A, Bourgeois JM, Tarnopolsky MA. Massage therapy attenuates inflammatory signaling later do-induced musculus damage. Sci Transl Med. 2012;iv(119):119ra113. doi: 10.1126/scitranslmed.3002882. [PubMed] [CrossRef] [Google Scholar]
  • Crisco JJ, Jokl P, Heinen GT, Connell Medico, Panjabi MM. A muscle contusion injury model. Biomechanics, physiology, and histology. Am J Sports Med. 1994;22(5):702–710. doi: 10.1177/036354659402200521. [PubMed] [CrossRef] [Google Scholar]
  • Darby IA, Zakuan N, Billet F, Desmouliere A. The myofibroblast, a primal prison cell in normal and pathological tissue repair. Cell Mol Life Sci. 2016;73(6):1145–1157. doi: 10.1007/s00018-015-2110-0. [PubMed] [CrossRef] [Google Scholar]
  • Deasy BM, Feduska JM, Payne TR, Li Y, Ambrosio F, Huard J. Effect of VEGF on the regenerative capacity of muscle stem cells in dystrophic skeletal muscle. Mol Ther. 2009;17(10):1788–1798. doi: ten.1038/mt.2009.136. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Desmouliere A, Gabbiani One thousand. Myofibroblast differentiation during fibrosis. Exp Nephrol. 1995;3(2):134–139. [PubMed] [Google Scholar]
  • Dhawan J, Rando TA. Stem cells in postnatal myogenesis: molecular mechanisms of satellite jail cell quiescence, activation and replenishment. Trends Prison cell Biol. 2005;15(12):666–673. doi: 10.1016/j.tcb.2005.10.007. [PubMed] [CrossRef] [Google Scholar]
  • Dumont NA, Bentzinger CF, Sincennes MC, Rudnicki MA. Satellite cells and skeletal muscle regeneration. Compr Physiol. 2015;5(3):1027–1059. doi: 10.1002/cphy.c140068. [PubMed] [CrossRef] [Google Scholar]
  • Dumont NA, Wang YX, Rudnicki MA. Intrinsic and extrinsic mechanisms regulating satellite cell part. Evolution. 2015;142(9):1572–1581. doi: x.1242/dev.114223. [PMC costless article] [PubMed] [CrossRef] [Google Scholar]
  • Engebretsen 50, Steffen G, Alsousou J, Anitua Eastward, Bachl N, Devilee R, Everts P, Hamilton B, Huard J, Jenoure P, Kelberine F, Kon E, Maffulli N, Matheson G, Mei-Dan O, Menetrey J, Philippon Chiliad, Randelli P, Schamasch P, Schwellnus M, Vernec A, Verrall G. IOC consensus paper on the use of platelet-rich plasma in sports medicine. Br J Sports Med. 2010;44(15):1072–1081. doi: 10.1136/bjsm.2010.079822. [PubMed] [CrossRef] [Google Scholar]
  • Engert JC, Berglund EB, Rosenthal Northward. Proliferation precedes differentiation in IGF-I-stimulated myogenesis. J Jail cell Biol. 1996;135(2):431–440. doi: 10.1083/jcb.135.2.431. [PMC gratuitous commodity] [PubMed] [CrossRef] [Google Scholar]
  • Engler AJ, Sen Southward, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126(4):677–689. doi: 10.1016/j.cell.2006.06.044. [PubMed] [CrossRef] [Google Scholar]
  • Floss T, Arnold HH, Braun T. A function for FGF-6 in skeletal muscle regeneration. Genes Dev. 1997;xi(sixteen):2040–2051. doi: 10.1101/gad.eleven.16.2040. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
  • Frey SP, Jansen H, Raschke MJ, Meffert RH, Ochman Southward. VEGF improves skeletal musculus regeneration later on acute trauma and reconstruction of the limb in a rabbit model. Clin Orthop Relat Res. 2012;470(12):3607–3614. doi: x.1007/s11999-012-2456-7. [PMC gratis commodity] [PubMed] [CrossRef] [Google Scholar]
  • Fukushima K, Badlani N, Usas A, Riano F, Fu F, Huard J. The use of an antifibrosis agent to improve muscle recovery after laceration. Am J Sports Med. 2001;29(four):394–402. [PubMed] [Google Scholar]
  • Garg K, Corona BT, Walters TJ. Therapeutic strategies for preventing skeletal muscle fibrosis later on injury. Front Pharmacol. 2015;6:87. doi: x.3389/fphar.2015.00087. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
  • Garrett WE, Jr, Seaber AV, Boswick J, Urbaniak JR, Goldner JL. Recovery of skeletal muscle after laceration and repair. J Mitt Surg. 1984;9(5):683–692. doi: 10.1016/S0363-5023(84)80014-3. [PubMed] [CrossRef] [Google Scholar]
  • Gilbert PM, Havenstrite KL, Magnusson KE, Sacco A, Leonardi NA, Kraft P, Nguyen NK, Thrun S, Lutolf MP, Blau HM. Substrate elasticity regulates skeletal muscle stalk cell self-renewal in civilisation. Science. 2010;329(5995):1078–1081. doi: 10.1126/scientific discipline.1191035. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Grounds MD. Musculus regeneration: molecular aspects and therapeutic implications. Curr Opin Neurol. 1999;12(5):535–543. doi: 10.1097/00019052-199910000-00007. [PubMed] [CrossRef] [Google Scholar]
  • Hamid MS, Yusof A, Mohamed Ali MR. Platelet-rich plasma (PRP) for astute musculus injury: a systematic review. PLoS One. 2014;9(ii) doi: 10.1371/journal.pone.0090538. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hammond JW, Hinton RY, Coil LA, Muriel JM, Lovering RM. Use of autologous platelet-rich plasma to care for muscle strain injuries. Am J Sports Med. 2009;37(half dozen):1135–1142. doi: x.1177/0363546508330974. [PMC costless article] [PubMed] [CrossRef] [Google Scholar]
  • Loma East, Boontheekul T, Mooney DJ. Designing scaffolds to heighten transplanted myoblast survival and migration. Tissue Eng. 2006;12(v):1295–1304. doi: 10.1089/10.2006.12.1295. [PubMed] [CrossRef] [Google Scholar]
  • Huard J, Li Y, Fu FH. Musculus injuries and repair: current trends in inquiry. J Os Joint Surg Am. 2002;84-A(5):822–832. [PubMed] [Google Scholar]
  • Hurme T, Kalimo H. Activation of myogenic precursor cells after muscle injury. Med Sci Sports Exerc. 1992;24(2):197–205. doi: 10.1249/00005768-199202000-00007. [PubMed] [CrossRef] [Google Scholar]
  • Hwang OK, Park JK, Lee EJ, Lee EM, Kim AY, Jeong KS. Therapeutic consequence of losartan, an angiotensin II blazon 1 receptor antagonist, on CCl(4)-induced skeletal muscle injury. Int J Mol Sci. 2016;17(2):227. doi: 10.3390/ijms17020227. [PMC costless article] [PubMed] [CrossRef] [Google Scholar]
  • Jarvinen TA, Jarvinen TL, Kaariainen M, Kalimo H, Jarvinen M. Muscle injuries: biology and treatment. Am J Sports Med. 2005;33(five):745–764. doi: 10.1177/0363546505274714. [PubMed] [CrossRef] [Google Scholar]
  • Jeon OH, Elisseeff J. Orthopedic tissue regeneration: cells, scaffolds, and small molecules. Drug Deliv Transl Res. 2016;6(2):105–120. doi: 10.1007/s13346-015-0266-7. [PubMed] [CrossRef] [Google Scholar]
  • Karvinen H, Pasanen E, Rissanen TT, Korpisalo P, Vahakangas E, Jazwa A, Giacca Thou, Yla-Herttuala S. Long-term VEGF-A expression promotes aberrant angiogenesis and fibrosis in skeletal musculus. Gene Ther. 2011;18(12):1166–1172. doi: x.1038/gt.2011.66. [PubMed] [CrossRef] [Google Scholar]
  • Kasemkijwattana C, Menetrey J, Somogyl K, Moreland MS, Fu FH, Buranapanitkit B, Watkins SC, Huard J. Evolution of approaches to improve the healing following musculus contusion. Cell Transplant. 1998;7(half-dozen):585–598. doi: 10.1016/S0963-6897(98)00037-2. [PubMed] [CrossRef] [Google Scholar]
  • Kasemkijwattana C, Menetrey J, Bosch P, Somogyi Grand, Moreland MS, Fu FH, Buranapanitkit B, Watkins SS, Huard J. Utilise of growth factors to meliorate muscle healing later strain injury. Clin Orthop. 2000;370:272–285. doi: x.1097/00003086-200001000-00028. [PubMed] [CrossRef] [Google Scholar]
  • Kastner South, Elias MC, Rivera AJ, Yablonka-Reuveni Z. Gene expression patterns of the fibroblast growth factors and their receptors during myogenesis of rat satellite cells. J Histochem Cytochem. 2000;48(8):1079–1096. doi: 10.1177/002215540004800805. [PubMed] [CrossRef] [Google Scholar]
  • Kumar A, Chaudhry I, Reid MB, Boriek AM. Singled-out signaling pathways are activated in response to mechanical stress applied axially and transversely to skeletal muscle fibers. J Biol Chem. 2002;277(48):46493–46503. doi: 10.1074/jbc.M203654200. [PubMed] [CrossRef] [Google Scholar]
  • Law PK, Bertorini TE, Goodwin TG, Chen Thousand, Fang QW, Li HJ, Kirby DS, Florendo JA, Herrod HG, Golden GS. Dystrophin production induced by myoblast transfer therapy in Duchenne muscular dystrophy. Lancet. 1990;336(8707):114–115. doi: 10.1016/0140-6736(xc)91628-N. [PubMed] [CrossRef] [Google Scholar]
  • Lee C, Fukushima K, Usas A, Xin 50, Pelinkovic D, Martinek Five, Huard J. Biological intervention based on cell and gene therapy to better muscle healing after laceration. J Musculoskelet Res. 2000;4(iv):256–277. doi: 10.1142/S0218957700000264. [CrossRef] [Google Scholar]
  • Lehto MU, Jarvinen MJ. Muscle injuries, their healing procedure and treatment. Ann Chir Gynaecol. 1991;eighty(ii):102–108. [PubMed] [Google Scholar]
  • Lehto K, Sims TJ, Bailey AJ. Skeletal muscle injury--molecular changes in the collagen during healing. Res Exp Med. 1985;185(2):95–106. doi: 10.1007/BF01854894. [PubMed] [CrossRef] [Google Scholar]
  • Lemos DR, Babaeijandaghi F, Depression M, Chang CK, Lee ST, Fiore D, Zhang RH, Natarajan A, Nedospasov SA, Rossi FM. Nilotinib reduces musculus fibrosis in chronic muscle injury by promoting TNF-mediated apoptosis of fibro/adipogenic progenitors. Nat Med. 2015;21(vii):786–794. doi: 10.1038/nm.3869. [PubMed] [CrossRef] [Google Scholar]
  • Li Y, Huard J. Differentiation of muscle-derived cells into myofibroblasts in injured skeletal muscle. Am J Pathol. 2002;161(3):895–907. doi: 10.1016/S0002-9440(10)64250-two. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
  • Li Y, Foster W, Deasy BM, Chan Y, Prisk V, Tang Y, Cummins J, Huard J. Transforming growth gene-beta1 induces the differentiation of myogenic cells into fibrotic cells in injured skeletal muscle: a key event in muscle fibrogenesis. Am J Pathol. 2004;164(3):1007–1019. doi: 10.1016/S0002-9440(10)63188-4. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Li Y, Li J, Zhu J, Sun B, Branca 1000, Tang Y, Foster Westward, Xiao X, Huard J. Decorin factor transfer promotes musculus cell differentiation and muscle regeneration. Mol Ther. 2007;fifteen(9):1616–1622. doi: 10.1038/sj.mt.6300250. [PubMed] [CrossRef] [Google Scholar]
  • Li H, Hicks JJ, Wang Fifty, Oyster North, Philippon MJ, Hurwitz Due south, Hogan MV, Huard J. Customized platelet-rich plasma with transforming growth factor beta1 neutralization antibody to reduce fibrosis in skeletal muscle. Biomaterials. 2016;87:147–156. doi: 10.1016/j.biomaterials.2016.02.017. [PubMed] [CrossRef] [Google Scholar]
  • Lipton BH, Schultz E. Developmental fate of skeletal musculus satellite cells. Science. 1979;205(4412):1292–1294. doi: x.1126/science.472747. [PubMed] [CrossRef] [Google Scholar]
  • Mann CJ, Perdiguero E, Kharraz Y, Aguilar S, Pessina P, Serrano AL, Munoz-Canoves P. Aberrant repair and fibrosis development in skeletal muscle. Skelet Muscle. 2011;1(i):21. doi: ten.1186/2044-5040-one-21. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • McLennan IS. Degenerating and regenerating skeletal muscles contain several subpopulations of macrophages with distinct spatial and temporal distributions. J Anat. 1996;188(Pt i):17–28. [PMC free article] [PubMed] [Google Scholar]
  • Mendell JR, Kissel JT, Amato AA, King Westward, Signore 50, Prior TW, Sahenk Z, Benson S, McAndrew PE, Rice R, Nagaraja H, Stephens R, Lantry L, Morris GE, Burghes AH. Myoblast transfer in the treatment of Duchenne's muscular dystrophy. N Engl J Med. 1995;333(13):832–838. doi: 10.1056/NEJM199509283331303. [PubMed] [CrossRef] [Google Scholar]
  • Menetrey J, Kasemkijwattana C, Fu FH, Moreland MS, Huard J. Suturing versus immobilization of a muscle laceration. A morphological and functional study in a mouse model. Am J Sports Med. 1999;27(ii):222–229. [PubMed] [Google Scholar]
  • Menetrey J, Kasemkijwattana C, Day CS, Bosch P, Vogt M, Fu FH, Moreland MS, Huard J. Growth factors better muscle healing in vivo. J Bone Joint Surg Br. 2000;82(1):131–137. doi: 10.1302/0301-620X.82B1.8954. [PubMed] [CrossRef] [Google Scholar]
  • Meng J, Chun Southward, Asfahani R, Lochmuller H, Muntoni F, Morgan J. Man skeletal muscle-derived CD133(+) cells form functional satellite cells after intramuscular transplantation in immunodeficient host mice. Mol Ther. 2014;22(5):1008–1017. doi: 10.1038/mt.2014.26. [PMC gratis article] [PubMed] [CrossRef] [Google Scholar]
  • Messina Due south, Mazzeo A, Bitto A, Aguennouz Grand, Migliorato A, De Pasquale MG, Minutoli L, Altavilla D, Zentilin L, Giacca M, Squadrito F, Vita Thousand. VEGF overexpression via adeno-associated virus factor transfer promotes skeletal muscle regeneration and enhances muscle role in mdx mice. FASEB J. 2007;21(13):3737–3746. doi: 10.1096/fj.07-8459com. [PubMed] [CrossRef] [Google Scholar]
  • Miller KJ, Thaloor D, Matteson S, Pavlath GK. Hepatocyte growth gene affects satellite cell activation and differentiation in regenerating skeletal muscle. Am J Physiol Cell Physiol. 2000;278(i):C174–C181. [PubMed] [Google Scholar]
  • Mourkioti F, Rosenthal North. IGF-1, inflammation and stem cells: interactions during muscle regeneration. Trends Immunol. 2005;26(10):535–542. doi: 10.1016/j.information technology.2005.08.002. [PubMed] [CrossRef] [Google Scholar]
  • Munoz-Canoves P, Serrano AL. Macrophages make up one's mind between regeneration and fibrosis in muscle. Trends Endocrinol Metab. 2015;26(ix):449–450. doi: 10.1016/j.tem.2015.07.005. [PubMed] [CrossRef] [Google Scholar]
  • Musaro A, McCullagh KJ, Naya FJ, Olson EN, Rosenthal N. IGF-one induces skeletal myocyte hypertrophy through calcineurin in clan with GATA-2 and NF-ATc1. Nature. 1999;400(6744):581–585. doi: 10.1038/23060. [PubMed] [CrossRef] [Google Scholar]
  • Musaro A, McCullagh K, Paul A, Houghton 50, Dobrowolny G, Molinaro M, Barton ER, Sweeney HL, Rosenthal Due north. Localized Igf-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal musculus. Nat Genet. 2001;27(2):195–200. doi: 10.1038/84839. [PubMed] [CrossRef] [Google Scholar]
  • Musaro A, Giacinti C, Borsellino 1000, Dobrowolny G, Pelosi L, Cairns L, Ottolenghi S, Cossu Chiliad, Bernardi G, Battistini L, Molinaro M, Rosenthal N. Stem jail cell-mediated muscle regeneration is enhanced by local isoform of insulin-like growth factor one. Proc Natl Acad Sci U S A. 2004;101(5):1206–1210. doi: 10.1073/pnas.0303792101. [PMC costless article] [PubMed] [CrossRef] [Google Scholar]
  • Ota S, Uehara K, Nozaki M, Kobayashi T, Terada S, Tobita K, Fu FH, Huard J. Intramuscular transplantation of muscle-derived stem cells accelerates skeletal muscle healing after contusion injury via enhancement of angiogenesis. Am J Sports Med. 2011;39(nine):1912–1922. doi: 10.1177/0363546511415239. [PubMed] [CrossRef] [Google Scholar]
  • Park JK, Ki MR, Lee EM, Kim AY, You SY, Han SY, Lee EJ, Hong IH, Kwon SH, Kim SJ, Rando TA, Jeong KS. Losartan improves adipose tissue-derived stem cell niche by inhibiting transforming growth factor-beta and fibrosis in skeletal muscle injury. Cell Transplant. 2012;21(xi):2407–2424. doi: 10.3727/096368912X637055. [PubMed] [CrossRef] [Google Scholar]
  • Partridge TA, Morgan JE, Coulton GR, Hoffman EP, Kunkel LM. Conversion of mdx myofibres from dystrophin-negative to -positive by injection of normal myoblasts. Nature. 1989;337(6203):176–179. doi: 10.1038/337176a0. [PubMed] [CrossRef] [Google Scholar]
  • Powell CA, Smiley BL, Mills J, Vandenburgh HH. Mechanical stimulation improves tissue-engineered human skeletal muscle. Am J Physiol Prison cell Physiol. 2002;283(5):C1557–C1565. doi: 10.1152/ajpcell.00595.2001. [PubMed] [CrossRef] [Google Scholar]
  • Proto JD, Tang Y, Lu A, Chen WC, Stahl E, Poddar Grand, Beckman SA, Robbins PD, Nidernhofer LJ, Imbrogno K, Hannigan T, Mars WM, Wang B, Huard J. NF-kappaB inhibition reveals a novel role for HGF during skeletal musculus repair. Cell Death Dis. 2015;6 doi: 10.1038/cddis.2015.66. [PMC gratuitous commodity] [PubMed] [CrossRef] [Google Scholar]
  • Rantanen J, Ranne J, Hurme T, Kalimo H. Denervated segments of injured skeletal muscle fibers are reinnervated by newly formed neuromuscular junctions. J Neuropathol Exp Neurol. 1995;54(2):188–194. doi: 10.1097/00005072-199503000-00005. [PubMed] [CrossRef] [Google Scholar]
  • Relaix F, Zammit PS. Satellite cells are essential for skeletal musculus regeneration: the jail cell on the edge returns eye stage. Development. 2012;139(16):2845–2856. doi: 10.1242/dev.069088. [PubMed] [CrossRef] [Google Scholar]
  • Reurink M, Goudswaard GJ, Moen MH, Weir A, Verhaar JA, Bierma-Zeinstra SM, Maas M, Tol JL, Dutch Hamstring Injection Therapy Study I Platelet-rich plasma injections in astute muscle injury. N Engl J Med. 2014;370(26):2546–2547. doi: 10.1056/NEJMc1402340. [PubMed] [CrossRef] [Google Scholar]
  • Reurink Yard, Goudswaard GJ, Moen MH, Weir A, Verhaar JA, Bierma-Zeinstra SM, Maas M, Tol JL, Dutch HITsI Rationale, secondary outcome scores and 1-year follow-up of a randomised trial of platelet-rich plasma injections in acute hamstring muscle injury: the Dutch Hamstring Injection Therapy study. Br J Sports Med. 2015;49(18):1206–1212. doi: x.1136/bjsports-2014-094250. [PubMed] [CrossRef] [Google Scholar]
  • Rocheteau P, Vinet One thousand, Chretien F. Dormancy and quiescence of skeletal muscle stem cells. Results Probl Cell Differ. 2015;56:215–235. doi: ten.1007/978-3-662-44608-9_10. [PubMed] [CrossRef] [Google Scholar]
  • Sadtler Thou, Estrellas K, Allen BW, Wolf MT, Fan H, Tam AJ, Patel CH, Luber BS, Wang H, Wagner KR, Powell JD, Housseau F, Pardoll DM, Elisseeff JH. Developing a pro-regenerative biomaterial scaffold microenvironment requires T helper ii cells. Science. 2016;352(6283):366–370. doi: 10.1126/science.aad9272. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Saera-Vila A, Kish PE, Kahana A. Fgf regulates dedifferentiation during skeletal muscle regeneration in adult zebrafish. Cell Signal. 2016;28(9):1196–1204. doi: 10.1016/j.cellsig.2016.06.001. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sambasivan R, Yao R, Kissenpfennig A, Van Wittenberghe 50, Paldi A, Gayraud-Morel B, Guenou H, Malissen B, Tajbakhsh S, Galy A. Pax7-expressing satellite cells are indispensable for adult skeletal muscle regeneration. Evolution. 2011;138(17):3647–3656. doi: 10.1242/dev.067587. [PubMed] [CrossRef] [Google Scholar]
  • Sampaolesi One thousand, Blot S, D'Antona Thou, Granger Northward, Tonlorenzi R, Innocenzi A, Mognol P, Thibaud JL, Galvez BG, Barthelemy I, Perani Fifty, Mantero S, Guttinger M, Pansarasa O, Rinaldi C, Cusella De Angelis MG, Torrente Y, Bordignon C, Bottinelli R, Cossu Grand. Mesoangioblast stalk cells ameliorate musculus function in dystrophic dogs. Nature. 2006;444(7119):574–579. doi: 10.1038/nature05282. [PubMed] [CrossRef] [Google Scholar]
  • Schiaffino S, Mammucari C. Regulation of skeletal muscle growth by the IGF1-Akt/PKB pathway: insights from genetic models. Skelet Muscle. 2011;1(1):4. doi: x.1186/2044-5040-ane-4. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Scholz D, Thomas Southward, Sass South, Podzuweit T. Angiogenesis and myogenesis every bit 2 facets of inflammatory post-ischemic tissue regeneration. Mol Cell Biochem. 2003;246(one-2):57–67. doi: 10.1023/A:1023403928385. [PubMed] [CrossRef] [Google Scholar]
  • Sheehan SM, Tatsumi R, Temm-Grove CJ, Allen RE. HGF is an autocrine growth factor for skeletal muscle satellite cells in vitro. Muscle Nervus. 2000;23(2):239–245. doi: 10.1002/(SICI)1097-4598(200002)23:two<239::AID-MUS15>3.0.CO;2-U. [PubMed] [CrossRef] [Google Scholar]
  • Skuk D, Goulet M, Roy B, Piette V, Cote CH, Chapdelaine P, Hogrel JY, Paradis M, Bouchard JP, Sylvain M, Lachance JG, Tremblay JP. Offset test of a "loftier-density injection" protocol for myogenic prison cell transplantation throughout large volumes of muscles in a Duchenne muscular dystrophy patient: eighteen months follow-up. Neuromuscul Disord. 2007;17(1):38–46. doi: 10.1016/j.nmd.2006.10.003. [PubMed] [CrossRef] [Google Scholar]
  • Taniguti AP, Pertille A, Matsumura CY, Santo Neto H, Marques MJ. Prevention of muscle fibrosis and myonecrosis in mdx mice by suramin, a TGF-beta1 blocker. Muscle Nerve. 2011;43(1):82–87. doi: x.1002/mus.21869. [PubMed] [CrossRef] [Google Scholar]
  • Tatsumi R, Sheehan SM, Iwasaki H, Hattori A, Allen RE. Mechanical stretch induces activation of skeletal muscle satellite cells in vitro. Exp Jail cell Res. 2001;267(1):107–114. doi: 10.1006/excr.2001.5252. [PubMed] [CrossRef] [Google Scholar]
  • Tedesco FS, Cossu G. Stem cell therapies for muscle disorders. Curr Opin Neurol. 2012;25(5):597–603. doi: 10.1097/WCO.0b013e328357f288. [PubMed] [CrossRef] [Google Scholar]
  • Tedesco FS, Dellavalle A, Diaz-Manera J, Messina G, Cossu G. Repairing skeletal musculus: regenerative potential of skeletal muscle stem cells. J Clin Invest. 2010;120(1):11–19. doi: 10.1172/JCI40373. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • Terada Due south, Ota Southward, Kobayashi G, Kobayashi T, Mifune Y, Takayama K, Witt Thou, Vadala G, Oyster N, Otsuka T, Fu FH, Huard J. Apply of an antifibrotic agent improves the effect of platelet-rich plasma on muscle healing later on injury. J Os Joint Surg Am. 2013;95(eleven):980–988. doi: x.2106/JBJS.L.00266. [PubMed] [CrossRef] [Google Scholar]
  • Tidball JG. Inflammatory cell response to acute muscle injury. Med Sci Sports Exerc. 1995;27(vii):1022–1032. doi: 10.1249/00005768-199507000-00011. [PubMed] [CrossRef] [Google Scholar]
  • Tidball JG. Inflammatory processes in musculus injury and repair. Am J Physiol Regul Integr Comp Physiol. 2005;288(two):R345–R353. doi: 10.1152/ajpregu.00454.2004. [PubMed] [CrossRef] [Google Scholar]
  • Tidball JG. Mechanisms of muscle injury, repair, and regeneration. Compr Physiol. 2011;one(iv):2029–2062. [PubMed] [Google Scholar]
  • Tidball JG, Wehling-Henricks Chiliad. Macrophages promote musculus membrane repair and muscle fibre growth and regeneration during modified muscle loading in mice in vivo. J Physiol. 2007;578(Pt 1):327–336. doi: 10.1113/jphysiol.2006.118265. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tidball JG, Welc SS. Macrophage-Derived IGF-1 Is a Stiff Coordinator of Myogenesis and Inflammation in Regenerating Muscle. Mol Ther. 2015;23(7):1134–1135. doi: 10.1038/mt.2015.97. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • Toumi H, All-time TM. The inflammatory response: friend or enemy for muscle injury? Br J Sports Med. 2003;37(4):284–286. doi: 10.1136/bjsm.37.four.284. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • Vaittinen S, Lukka R, Sahlgren C, Hurme T, Rantanen J, Lendahl U, Eriksson JE, Kalimo H. The expression of intermediate filament protein nestin as related to vimentin and desmin in regenerating skeletal muscle. J Neuropathol Exp Neurol. 2001;sixty(6):588–597. doi: 10.1093/jnen/sixty.six.588. [PubMed] [CrossRef] [Google Scholar]
  • Walczak BE, Johnson CN, Howe BM. Myositis Ossificans. J Am Acad Orthop Surg. 2015;23(10):612–622. doi: 10.5435/JAAOS-D-14-00269. [PubMed] [CrossRef] [Google Scholar]
  • Wu H, Xiong WC, Mei Fifty. To build a synapse: signaling pathways in neuromuscular junction assembly. Development. 2010;137(vii):1017–1033. doi: 10.1242/dev.038711. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yablonka-Reuveni Z, Balestreri TM, Bowen-Pope DF. Regulation of proliferation and differentiation of myoblasts derived from developed mouse skeletal musculus by specific isoforms of PDGF. J Prison cell Biol. 1990;111(4):1623–1629. doi: 10.1083/jcb.111.iv.1623. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • Yin H, Toll F, Rudnicki MA. Satellite cells and the muscle stem cell niche. Physiol Rev. 2013;93(1):23–67. doi: 10.1152/physrev.00043.2011. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhao Westward, Lu H, Wang X, Ransohoff RM, Zhou 50. CX3CR1 deficiency delays acute skeletal muscle injury repair by impairing macrophage functions. FASEB J. 2016;30(1):380–393. doi: 10.1096/fj.14-270090. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zheng B, Cao B, Crisan M, Lord's day B, Li G, Logar A, Yap S, Pollett JB, Drowley 50, Cassino T, Gharaibeh B, Deasy BM, Huard J, Peault B. Prospective identification of myogenic endothelial cells in human skeletal musculus. Nat Biotechnol. 2007;25(9):1025–1034. doi: ten.1038/nbt1334. [PubMed] [CrossRef] [Google Scholar]

Articles from Journal of Experimental Orthopaedics are provided here courtesy of Springer-Verlag


How Does Muscle Tissue Repair Itself,

Source: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4958098/

Posted by: mcalisterandister.blogspot.com

0 Response to "How Does Muscle Tissue Repair Itself"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel