banner



How Long Does It Take Muscles To Grow Back Together After Being Repaired

  • Journal List
  • J Exp Orthop
  • v.3; 2022 December
  • PMC4958098

J Exp Orthop. 2022 Dec; 3: 15.

Muscle injuries and strategies for improving their repair

Thomas Laumonier

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

Jacques Menetrey

Section 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 xv.

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 different mechanisms, which imply prison cell-prison cell and jail cell-matrix interactions besides as extracellular secreted factors. Cellular dynamics during muscle regeneration are highly complex. Allowed, fibrotic, vascular and myogenic cells announced with distinct temporal and spatial kinetics later muscle injury. Iii main phases take been identified in the procedure of musculus regeneration; a destruction phase 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 minor muscle injuries, such as strains, heal spontaneously, severe muscle injuries class fibrotic tissue that impairs muscle function and lead to muscle contracture and chronic pain. Current therapeutic approaches have limited effectiveness and optimal strategies for such lesions are non known yet. 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, Stalk 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 stem jail cell located between the plasma membrane of myofibers and the basal lamina. Their regenerative capabilities are essential to repair skeletal muscle after 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 musculus blazon, around 5 to 10 % of skeletal muscle cells (Rocheteau et al. 2022). Afterward injury, SC become activated, proliferate and requite rising to myogenic precursor 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 variety of events, including direct trauma such as 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, later on severe injuries, musculus healing is incomplete, oft resulting in the formation of fibrotic tissue that impairs muscle function. Although researchers take extensively investigated various approaches to better muscle healing, there is nevertheless no golden standard treatment.

This curtailed 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 research efforts that have focused on the use of stem jail cell therapy, growth factors and/or biological scaffolds to improve muscle regeneration and repair. Nosotros likewise accost the therapeutic potential of mechanical stimulation and of anti-fibrotic therapy to enhance muscle regeneration and repair.

Review

Muscle healing procedure

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

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

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

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

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

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

Musculus degeneration and inflammation

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

Musculus regeneration, remodeling and maturation

Muscle regeneration usually starts during the get-go 4–5 days after injury, peaks at 2 weeks, and then gradually diminishes three to iv weeks afterward injury. It's a multiple steps procedure including activation/proliferation of SC, repair and maturation of damaged muscle fibers and connective tissue formation. A fine balance betwixt these mechanisms is essential for a total recovery of the contractile muscle function.

Muscle fibers are post-mitotic cells, which do not have the chapters to divide. Following 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 tin can either differentiate to repair damaged fibers or, for a small proportion, self-renew to maintain the SC pool for possible time to come demands of musculus regeneration (Collins 2006; Dhawan and Rando 2005). SC cycle progression and jail cell fate decision are control by complex 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 of import footstep of the regenerative muscle process. Rapidly later musculus injury, a gap is formed betwixt damaged muscle fibers and filled with a hematoma. Muscle injuries can be clinically classified depending of the nature of the hematoma (size, location). Late elimination of the hematoma is known to filibuster skeletal muscle regeneration, to better fibrosis and to reduce biomechanical properties of the healing muscle (Beiner et al. 1999). In rare complexity, major muscle injuries may lead 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 speedily invaded by fibroblasts (Darby et al. 2022; Desmouliere and Gabbiani 1995). Fibrogenic cytokines such every bit transforming growth factor β1 (TGF-β1) participate to excessive fibroblasts/myofibroblasts proliferation and to an increase 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, ofttimes result in an increase of scar tissue size over time that can prevent normal muscle office (Mann et al. 2022). Many growth factors are involved in the development of fibrosis, such as Connective Tissue Growth Cistron (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 every bit the key element in this process (Isle of man et al. 2022),. Although fibroblasts are the major collagen-producing cells in skeletal muscle, TGF-β1 have also an consequence straight on myoblasts causing their conversion to myofibroblasts. Thus myoblasts initially acting to repair damaged myofibers, will produce significant level of collagen and volition contribute to muscle fibrosis (Li and Huard 2002).

R evascularization

The restoration of the blood supply in the injured skeletal musculus is one of the first signs of muscle regeneration and is essential to its success. Without revascularization, musculus regeneration is incomplete and a significant fibrosis occurs (Best et al. 2022; Ota et al. 2022). Later muscle trauma, blood vessels rupture induces tissue hypoxia at the injury site (Jarvinen et al. 2005). New capillaries formation quickly after injury is therefore necessary (Scholz et al. 2003) for a functional muscle recovery. Secretion of angiogenic factors such as vascular endothelial growth factor (VEGF) at the lesion site is of import and several studies take 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 take place at a specific site in the cardinal region of myofibers, the neuromuscular junction (NMJ) (Wu et al. 2010). NMJ are essential for maturation and functional activity of regenerating muscles. Within 2–three weeks after musculus harm, the presence of newly formed NMJ is observed in regenerative muscle (Rantanen et al. 1995; Vaittinen et al. 2001).

Strategies to improve musculus 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 past other prison cell types nowadays at the inflammatory site, are release in the extracellular space and attune the regenerative response (Tabular array1). Although hepatocyte growth cistron (HGF), fibroblast growth factor (FGF) and platelet-derived growth factor (PDGF) are of interest because of their chapters to stimulate satellite cells (Sheehan et al. 2000; Allen and Boxhorn 1989; Yablonka-Reuveni et al. 1990), insulin like growth gene-one (IGF-I) appears to be of item importance for the muscle regeneration process. 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, direct injections of human recombinant IGF-I at ii, five, and seven days later on injury enhanced muscle healing in lacerated, contused, and strain-injured muscles (Menetrey et al. 2000; Kasemkijwattana et al. 2000). Yet, the efficacy of directly injection of recombinant proteins is limited by the high concentration of the factor typically required to elicit a measurable event. This is mainly due to the bloodstream's rapid clearance of these molecules and their relatively short biological one-half-lives. Gene therapy may be an effective method past which to deliver loftier, 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). Another growth factor, VEGF, by 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 as a possible culling approach based on the ability of autologous growth factors to improve skeletal muscle regeneration (Hamid et al. 2022; Hammond et al. 2009). Considered equally prophylactic 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, upwardly to one year afterward 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).

Table 1

The role of growth factors in skeletal muscle regeneration

Growth factors Physiological effects, potential benefits Shortcomings Commentary
IGF-one - Essential for muscle growth during development and regeneration.
- Promote myoblast proliferation and differentiation in vitro (Huard et al. 2002)
- Hypertrophic outcome of IGF-one (Barton-Davis et al. 1999)
- Serial injections of IGF-1 improve musculus healing in vivo (Menetrey et al. 2000).
- Existence of a muscle specific isoform of IGF-1 (mIGF-1) (Musaro et al. 1999; Musaro et al. 2004)
- Chemotactic for fibroblasts, increment collagen production, enhance fibrosis evolution - IGF-1 play a central role in the enhancement of muscle regeneration-
- Anti-inflammatory actions of IGF-1 (Mourkioti and Rosenthal 2005; Tidball and Welc 2022)
HGF - Promote myoblast proliferation and inhibit myoblast differentiation (Anderson 2022; Yin et al. 2022)
- Important role for satellite cell activation. Balance between the activation of satellite cells and their return to quiescence. (Chazaud 2010)
- Recently, it was shown that a second set of HGF production is crucial for inflammation resolution afterwards injury (Proto et al. 2022)
- Injection of HGF into injured muscle increased myoblast numbers but blocked the regeneration procedure (Miller et al. 2000) - HGF is of import during the early phase of muscle regeneration, actuate satellite cells
VEGF - Important signaling protein that favor angiogenesis.
- Promote myoblast migration, proliferation and survival. (Arsic et al. 2004)
- VEGF administration improves muscle regeneration. (Messina et al. 2007; Deasy et al. 2009)
- Non regulated VEGF expression promote aberrant angiogenesis and fibrosis in skeletal muscle (Karvinen et al. 2022) - Importance of the proximity betwixt satellite cells and the microvasculature during muscle regeneration, role of VEGF
FGF - Large family of mitogen involved in prison cell growth and survival
- FGF-6 has a muscle specific expression, stimulates satellite cell proliferation and promotes myogenic concluding 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 key role in muscle repair, blocking FGF signaling delay musculus regeneration (Saera-Vila et al. 2022).
TGF-β1 - Primal regulator of the residuum between musculus 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 improve muscle regeneration. (Burks et al. 2022; Hwang et al. 2022)
PDGF-BB - PDGF isoforms tin 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)
- Potent mitogen for fibroblasts - Release from injured vessels and platelets, PDGF stimulates early skeletal muscle regeneration

Stem cells

Transplantation of satellite cell-derived myoblasts has long been explored equally a promising approach for treatment of skeletal musculus disorders. Later on an initial demonstration that normal myoblasts tin 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 immature boys muscles, take non been successful (Law et al. 1990; Mendell et al. 1995). Even recently, despite clear comeback in methodologies that raise the success of myoblast transplantation in Duchenne patients (Skuk et al. 2007), outcomes of clinical trials are nonetheless disappointing. These experiments have raised concerns near the express migratory and proliferative capacities of human myoblasts, too every bit their limited life span in vivo. It led to the investigations of other muscle stalk cells sources that could overcome these limitations and outperform the success of muscle cell transplantation. Among all these non-satellite myogenic stem cells, human mesoangioblasts, human myogenic-endothelial cells and human muscle–derived CD133+ have shown myogenic potentials in vitro and in vivo (Sampaolesi et al. 2006; Zheng et al. 2007; Meng et al. 2022). The use of such myogenic progenitors cells for improving muscle healing may become an interesting therapeutic alternative (Tedesco and Cossu 2022; Tedesco et al. 2010; Chen et al. 2022). A first stage I/IIa clinical trial has recently demonstrated that intra arterial injections of human being mesoangioblasts are prophylactic but display only very limited clinical efficacy in Duchenne patients (Cossu et al. 2022).

Scaffolds

Myogenic precursor cell survival and migration is greatly increased by using advisable scaffold limerick and growth cistron commitment (Hill et al. 2006) (Boldrin et al. 2007). Controlling the microenvironment of injected myogenic cells using biological scaffolds raise muscle regeneration (Borselli et al. 2022). Ideally, using an appropriate extracellular matrix (ECM) limerick 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 choice to amend regeneration of injured skeletal muscles (Jeon and Elisseeff 2022).

Anti-fibrotic therapy

TGF-β1 is expressed at high levels and plays an important 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 muscle should inhibit the formation of scar tissue. Indeed, the use of anti-fibrotic agents (ie decorin, relaxin, antibody against TGF-β1…) that inactivate TGF-β1 signaling pathways reduces musculus fibrosis and, consequently, improve musculus healing, leading to a virtually complete recovery of lacerated muscle (Fukushima et al. 2001; Li et al. 2007). Losartan, an angiotensin Ii receptor antagonist, neutralize the event of TGF-β1 and reduce fibrosis, making it the treatment of choice, since it already has FDA approval to be used clinically (Bedair et al. 2008; Park et al. 2022; Terada et al. 2022). Suramin, too 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 elementary and effective approach to enhance skeletal musculus regeneration. Stretch activation, mechanical conditioning merely also massage therapy or concrete manipulation of injured skeletal muscles accept shown multiple benefit effects on muscle biological science and part 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 important biological regulators equally chemicals and genes, and underlines the immense potential of developing mechano-therapies to care for muscle damage (Cezar et al. 2022). A recent written report 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 be considered equally a possible therapy to improve muscle regeneration and repair.

Conclusions

Skeletal musculus injuries are very oftentimes present in sports medicine and pose challenging problems in traumatology. Despite their clinical importance, the optimal rehabilitation strategies for treating these injuries are not well defined. After a trauma, skeletal muscles have the chapters to regenerate and repair in a complex and well-coordinated response. This process required the presence of various prison cell populations, upwards and downwards-regulation of various cistron expressions and participation of multiples growth factors. Strategies based on the combination of stem cells, growth factors and biological scaffolds have already shown promising results in animal models. A amend understanding of the cellular and molecular pathways besides as a ameliorate definition of the interactions (jail cell-cell and jail cell-matrix) that are essential for constructive musculus regeneration, should contribute to the development of new therapies in humans. In this direction, a recent paper 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).

Abbreviation

CTGF, connective tissue growth factor; FGF, fibroblast growth factor; HGF, hepatocyte growth factor; IGF-I, insulin similar growth factor-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 have no competing interests.

Authors' contributions

TL and JM participated equally in drafting the manuscript. Both authors read and approved the final 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-like growth gene I, and fibroblast growth factor. J Cell Physiol. 1989;138(two):311–315. doi: 10.1002/jcp.1041380213. [PubMed] [CrossRef] [Google Scholar]
  • Anderson JE. Hepatocyte growth factor and satellite cell activation. Adv Exp Med Biol. 2016;900:1–25. doi: 10.1007/978-3-319-27511-6_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 afterwards skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med. 2007;204(five):1057–1069. doi: 10.1084/jem.20070075. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
  • Arsic N, Zacchigna South, Zentilin L, Ramirez-Correa One thousand, Pattarini 50, Salvi A, Sinagra G, Giacca Grand. Vascular endothelial growth factor stimulates skeletal muscle regeneration in vivo. Mol Ther. 2004;x(5):844–854. doi: 10.1016/j.ymthe.2004.08.007. [PubMed] [CrossRef] [Google Scholar]
  • Barton ER, Morris 50, Musaro A, Rosenthal N, Sweeney HL. Muscle-specific expression of insulin-similar growth factor I counters musculus pass up in mdx mice. J Jail cell Biol. 2002;157(1):137–148. doi: x.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 crumbling-related loss of skeletal musculus function. Proc Natl Acad Sci U S A. 1998;95(26):15603–15607. doi: 10.1073/pnas.95.26.15603. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Barton-Davis ER, Shoturma DI, Sweeney HL. Contribution of satellite cells to IGF-I induced hypertrophy of skeletal muscle. Acta Physiol Scand. 1999;167(iv):301–305. doi: 10.1046/j.1365-201x.1999.00618.ten. [PubMed] [CrossRef] [Google Scholar]
  • Bedair HS, Karthikeyan T, Quintero A, Li Y, Huard J. Angiotensin II receptor blockade administered later injury improves muscle regeneration and decreases fibrosis in normal skeletal muscle. Am J Sports Med. 2008;36(8):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(ane):2–9. [PubMed] [Google Scholar]
  • Bernasconi P, Torchiana Eastward, Confalonieri P, Brugnoni R, Barresi R, Mora M, Cornelio F, Morandi L, Mantegazza R. Expression of transforming growth cistron-beta 1 in dystrophic patient muscles correlates with fibrosis. Pathogenetic function of a fibrogenic cytokine. J Clin Invest. 1995;96(2):1137–1144. doi: 10.1172/JCI118101. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Best TM, Gharaibeh B, Huard J. Stem cells, angiogenesis and musculus healing: a potential part in massage therapies? Br J Sports Med. 2012 [PubMed] [Google Scholar]
  • Boldrin Fifty, Elvassore North, Malerba A, Flaibani M, Cimetta E, Piccoli M, Baroni Medico, Gazzola MV, Messina C, Gamba P, Vitiello L, de Coppi P. Satellite cells delivered by micro-patterned scaffolds: a new strategy for cell transplantation in musculus diseases. Tissue Eng. 2007;13(ii):253–262. doi: 10.1089/10.2006.0093. [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 South A. 2010;107(eight):3287–3292. doi: 10.1073/pnas.0903875106. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Borselli C, Cezar CA, Shvartsman D, Vandenburgh HH, Mooney DJ. The role of multifunctional commitment 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 E, 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 free article] [PubMed] [CrossRef] [Google Scholar]
  • Cezar CA, Roche ET, Vandenburgh HH, Duda GN, Walsh CJ, Mooney DJ. Biologic-costless mechanically induced muscle regeneration. Proc Natl Acad Sci U S A. 2016;113(6):1534–1539. doi: x.1073/pnas.1517517113. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chan YS, Li Y, Foster W, Horaguchi T, Somogyi G, Fu FH, Huard J. Antifibrotic furnishings of suramin in injured skeletal muscle after laceration. J Appl Physiol. 2003;95(2):771–780. doi: x.1152/japplphysiol.00915.2002. [PubMed] [CrossRef] [Google Scholar]
  • Charge SB, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiol Rev. 2004;84(i):209–238. doi: 10.1152/physrev.00019.2003. [PubMed] [CrossRef] [Google Scholar]
  • Chazaud B. Dual event of HGF on satellite/myogenic cell quiescence. Focus on "High concentrations of HGF inhibit skeletal muscle satellite cell proliferation in vitro by inducing expression of myostatin: a possible machinery for reestablishing satellite cell quiescence in vivo". Am J Physiol Cell Physiol. 2010;298(three):C448–C449. doi: ten.1152/ajpcell.00561.2009. [PubMed] [CrossRef] [Google Scholar]
  • Chazaud B. Macrophages: supportive cells for tissue repair and regeneration. Immunobiology. 2014;219(3):172–178. doi: x.1016/j.imbio.2013.09.001. [PubMed] [CrossRef] [Google Scholar]
  • Chazaud B, Sonnet C, Lafuste P, Bassez G, Rimaniol Air-conditioning, Poron F, Authier FJ, Dreyfus PA, Gherardi RK. Satellite cells concenter monocytes and use macrophages as a support to escape apoptosis and enhance muscle growth. J Cell Biol. 2003;163(5):1133–1143. doi: 10.1083/jcb.200212046. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chen CW, Corselli Chiliad, Peault B, Huard J. Human blood-vessel-derived stem cells for tissue repair and regeneration. J Biomed Biotechnol. 2012;2012:597439. [PMC free article] [PubMed] [Google Scholar]
  • Collins CA. Satellite prison cell cocky-renewal. Curr Opin Pharmacol. 2006;half dozen(3):301–306. doi: 10.1016/j.coph.2006.01.006. [PubMed] [CrossRef] [Google Scholar]
  • Cossu M, Sampaolesi One thousand. New therapies for Duchenne muscular dystrophy: challenges, prospects and clinical trials. Trends Mol Med. 2007;thirteen(12):520–526. doi: ten.1016/j.molmed.2007.ten.003. [PubMed] [CrossRef] [Google Scholar]
  • Cossu G, Previtali SC, Napolitano S, Cicalese MP, Tedesco FS, Nicastro F, Noviello M, Roostalu U, Natali Sora MG, Scarlato M, De Pellegrin M, Godi C, Giuliani Southward, Ciotti F, Tonlorenzi R, Lorenzetti I, Rivellini C, Benedetti S, Gatti R, Marktel S, Mazzi B, Tettamanti A, Ragazzi M, Imro MA, Marano Thousand, Ambrosi A, Fiori R, Sormani MP, Bonini C, Venturini One thousand, Politi LS, Torrente Y, Ciceri F. Intra-arterial transplantation of HLA-matched donor mesoangioblasts in Duchenne muscular dystrophy. EMBO Mol Med. 2015 [PMC complimentary article] [PubMed] [Google Scholar]
  • Crane JD, Ogborn DI, Cupido C, Melov Due south, Hubbard A, Conservative JM, Tarnopolsky MA. Massage therapy attenuates inflammatory signaling after practise-induced muscle damage. Sci Transl Med. 2012;4(119):119ra113. doi: 10.1126/scitranslmed.3002882. [PubMed] [CrossRef] [Google Scholar]
  • Crisco JJ, Jokl P, Heinen GT, Connell MD, 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 key prison cell in normal and pathological tissue repair. Jail 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. Issue of VEGF on the regenerative chapters of muscle stem cells in dystrophic skeletal muscle. Mol Ther. 2009;17(10):1788–1798. doi: 10.1038/mt.2009.136. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Desmouliere A, Gabbiani Grand. Myofibroblast differentiation during fibrosis. Exp Nephrol. 1995;iii(2):134–139. [PubMed] [Google Scholar]
  • Dhawan J, Rando TA. Stem cells in postnatal myogenesis: molecular mechanisms of satellite cell quiescence, activation and replenishment. Trends Prison cell Biol. 2005;fifteen(12):666–673. doi: 10.1016/j.tcb.2005.x.007. [PubMed] [CrossRef] [Google Scholar]
  • Dumont NA, Bentzinger CF, Sincennes MC, Rudnicki MA. Satellite cells and skeletal musculus 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 function. Evolution. 2015;142(ix):1572–1581. doi: 10.1242/dev.114223. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
  • Engebretsen L, Steffen M, Alsousou J, Anitua E, Bachl N, Devilee R, Everts P, Hamilton B, Huard J, Jenoure P, Kelberine F, Kon E, Maffulli Due north, Matheson Thousand, Mei-Dan O, Menetrey J, Philippon Yard, Randelli P, Schamasch P, Schwellnus K, Vernec A, Verrall G. IOC consensus paper on the employ of platelet-rich plasma in sports medicine. Br J Sports Med. 2010;44(xv):1072–1081. doi: 10.1136/bjsm.2010.079822. [PubMed] [CrossRef] [Google Scholar]
  • Engert JC, Berglund EB, Rosenthal N. Proliferation precedes differentiation in IGF-I-stimulated myogenesis. J Cell Biol. 1996;135(ii):431–440. doi: ten.1083/jcb.135.two.431. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126(four):677–689. doi: 10.1016/j.cell.2006.06.044. [PubMed] [CrossRef] [Google Scholar]
  • Floss T, Arnold HH, Braun T. A role for FGF-6 in skeletal musculus regeneration. Genes Dev. 1997;11(16):2040–2051. doi: 10.1101/gad.eleven.16.2040. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Frey SP, Jansen H, Raschke MJ, Meffert RH, Ochman S. VEGF improves skeletal muscle regeneration later astute trauma and reconstruction of the limb in a rabbit model. Clin Orthop Relat Res. 2012;470(12):3607–3614. doi: ten.1007/s11999-012-2456-7. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • Fukushima K, Badlani N, Usas A, Riano F, Fu F, Huard J. The utilize of an antifibrosis agent to improve muscle recovery after laceration. Am J Sports Med. 2001;29(4):394–402. [PubMed] [Google Scholar]
  • Garg Thou, Corona BT, Walters TJ. Therapeutic strategies for preventing skeletal muscle fibrosis after injury. Front end Pharmacol. 2015;half-dozen:87. doi: 10.3389/fphar.2015.00087. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Garrett Nosotros, Jr, Seaber AV, Boswick J, Urbaniak JR, Goldner JL. Recovery of skeletal musculus after laceration and repair. J Hand 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 cocky-renewal in culture. Scientific discipline. 2010;329(5995):1078–1081. doi: ten.1126/science.1191035. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Grounds MD. Muscle regeneration: molecular aspects and therapeutic implications. Curr Opin Neurol. 1999;12(5):535–543. doi: x.1097/00019052-199910000-00007. [PubMed] [CrossRef] [Google Scholar]
  • Hamid MS, Yusof A, Mohamed Ali MR. Platelet-rich plasma (PRP) for acute muscle injury: a systematic review. PLoS One. 2014;9(2) doi: 10.1371/journal.pone.0090538. [PMC gratuitous commodity] [PubMed] [CrossRef] [Google Scholar]
  • Hammond JW, Hinton RY, Coil LA, Muriel JM, Lovering RM. Use of autologous platelet-rich plasma to treat muscle strain injuries. Am J Sports Med. 2009;37(6):1135–1142. doi: x.1177/0363546508330974. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hill Due east, Boontheekul T, Mooney DJ. Designing scaffolds to raise transplanted myoblast survival and migration. Tissue Eng. 2006;12(5):1295–1304. doi: 10.1089/ten.2006.12.1295. [PubMed] [CrossRef] [Google Scholar]
  • Huard J, Li Y, Fu FH. Muscle injuries and repair: current trends in research. J Bone Joint Surg Am. 2002;84-A(v):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 upshot of losartan, an angiotensin Ii type i receptor antagonist, on CCl(4)-induced skeletal muscle injury. Int J Mol Sci. 2016;17(2):227. doi: x.3390/ijms17020227. [PMC gratuitous 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: x.1007/s13346-015-0266-7. [PubMed] [CrossRef] [Google Scholar]
  • Karvinen H, Pasanen E, Rissanen TT, Korpisalo P, Vahakangas Due east, Jazwa A, Giacca K, Yla-Herttuala S. Long-term VEGF-A expression promotes aberrant angiogenesis and fibrosis in skeletal musculus. Gene Ther. 2011;eighteen(12):1166–1172. doi: 10.1038/gt.2011.66. [PubMed] [CrossRef] [Google Scholar]
  • Kasemkijwattana C, Menetrey J, Somogyl M, Moreland MS, Fu FH, Buranapanitkit B, Watkins SC, Huard J. Evolution of approaches to improve the healing following muscle contusion. Jail cell Transplant. 1998;vii(half-dozen):585–598. doi: ten.1016/S0963-6897(98)00037-2. [PubMed] [CrossRef] [Google Scholar]
  • Kasemkijwattana C, Menetrey J, Bosch P, Somogyi M, Moreland MS, Fu FH, Buranapanitkit B, Watkins SS, Huard J. Use of growth factors to improve muscle healing afterwards strain injury. Clin Orthop. 2000;370:272–285. doi: ten.1097/00003086-200001000-00028. [PubMed] [CrossRef] [Google Scholar]
  • Kastner S, 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 musculus 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 M, Fang QW, Li HJ, Kirby DS, Florendo JA, Herrod HG, Golden GS. Dystrophin production induced past myoblast transfer therapy in Duchenne muscular dystrophy. Lancet. 1990;336(8707):114–115. doi: 10.1016/0140-6736(90)91628-N. [PubMed] [CrossRef] [Google Scholar]
  • Lee C, Fukushima K, Usas A, Xin L, Pelinkovic D, Martinek V, Huard J. Biological intervention based on jail cell and gene therapy to improve musculus healing after laceration. J Musculoskelet Res. 2000;4(iv):256–277. doi: 10.1142/S0218957700000264. [CrossRef] [Google Scholar]
  • Lehto MU, Jarvinen MJ. Musculus injuries, their healing process and treatment. Ann Chir Gynaecol. 1991;eighty(2):102–108. [PubMed] [Google Scholar]
  • Lehto M, Sims TJ, Bailey AJ. Skeletal muscle injury--molecular changes in the collagen during healing. Res Exp Med. 1985;185(two):95–106. doi: 10.1007/BF01854894. [PubMed] [CrossRef] [Google Scholar]
  • Lemos DR, Babaeijandaghi F, Low Chiliad, Chang CK, Lee ST, Fiore D, Zhang RH, Natarajan A, Nedospasov SA, Rossi FM. Nilotinib reduces muscle fibrosis in chronic muscle injury past promoting TNF-mediated apoptosis of fibro/adipogenic progenitors. Nat Med. 2015;21(7):786–794. doi: ten.1038/nm.3869. [PubMed] [CrossRef] [Google Scholar]
  • Li Y, Huard J. Differentiation of muscle-derived cells into myofibroblasts in injured skeletal musculus. Am J Pathol. 2002;161(iii):895–907. doi: 10.1016/S0002-9440(10)64250-2. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Li Y, Foster W, Deasy BM, Chan Y, Prisk V, Tang Y, Cummins J, Huard J. Transforming growth factor-beta1 induces the differentiation of myogenic cells into fibrotic cells in injured skeletal musculus: a primal event in muscle fibrogenesis. Am J Pathol. 2004;164(3):1007–1019. doi: 10.1016/S0002-9440(10)63188-4. [PMC costless article] [PubMed] [CrossRef] [Google Scholar]
  • Li Y, Li J, Zhu J, Sunday B, Branca M, Tang Y, Foster W, Xiao X, Huard J. Decorin cistron transfer promotes muscle cell differentiation and musculus regeneration. Mol Ther. 2007;15(9):1616–1622. doi: x.1038/sj.mt.6300250. [PubMed] [CrossRef] [Google Scholar]
  • Li H, Hicks JJ, Wang 50, Oyster N, Philippon MJ, Hurwitz S, 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: x.1016/j.biomaterials.2016.02.017. [PubMed] [CrossRef] [Google Scholar]
  • Lipton BH, Schultz E. Developmental fate of skeletal musculus satellite cells. Scientific discipline. 1979;205(4412):1292–1294. doi: 10.1126/science.472747. [PubMed] [CrossRef] [Google Scholar]
  • Mann CJ, Perdiguero Due east, 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: x.1186/2044-5040-i-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 1):17–28. [PMC costless article] [PubMed] [Google Scholar]
  • Mendell JR, Kissel JT, Amato AA, Male monarch Due west, 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: ten.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(2):222–229. [PubMed] [Google Scholar]
  • Menetrey J, Kasemkijwattana C, Solar day CS, Bosch P, Vogt M, Fu FH, Moreland MS, Huard J. Growth factors improve musculus healing in vivo. J Bone Articulation Surg Br. 2000;82(1):131–137. doi: 10.1302/0301-620X.82B1.8954. [PubMed] [CrossRef] [Google Scholar]
  • Meng J, Chun Due south, Asfahani R, Lochmuller H, Muntoni F, Morgan J. Human skeletal muscle-derived CD133(+) cells form functional satellite cells afterward intramuscular transplantation in immunodeficient host mice. Mol Ther. 2014;22(5):1008–1017. doi: x.1038/mt.2014.26. [PMC gratis article] [PubMed] [CrossRef] [Google Scholar]
  • Messina S, Mazzeo A, Bitto A, Aguennouz Yard, Migliorato A, De Pasquale MG, Minutoli L, Altavilla D, Zentilin L, Giacca 1000, Squadrito F, Vita G. VEGF overexpression via adeno-associated virus gene transfer promotes skeletal muscle regeneration and enhances muscle function in mdx mice. FASEB J. 2007;21(13):3737–3746. doi: x.1096/fj.07-8459com. [PubMed] [CrossRef] [Google Scholar]
  • Miller KJ, Thaloor D, Matteson S, Pavlath GK. Hepatocyte growth factor affects satellite cell activation and differentiation in regenerating skeletal musculus. Am J Physiol Cell Physiol. 2000;278(one):C174–C181. [PubMed] [Google Scholar]
  • Mourkioti F, Rosenthal N. IGF-one, inflammation and stem cells: interactions during muscle regeneration. Trends Immunol. 2005;26(10):535–542. doi: 10.1016/j.it.2005.08.002. [PubMed] [CrossRef] [Google Scholar]
  • Munoz-Canoves P, Serrano AL. Macrophages decide between regeneration and fibrosis in musculus. Trends Endocrinol Metab. 2015;26(nine):449–450. doi: 10.1016/j.tem.2015.07.005. [PubMed] [CrossRef] [Google Scholar]
  • Musaro A, McCullagh KJ, Naya FJ, Olson EN, Rosenthal North. IGF-1 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 L, Dobrowolny 1000, Molinaro Grand, Barton ER, Sweeney HL, Rosenthal Northward. Localized Igf-ane transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat Genet. 2001;27(2):195–200. doi: 10.1038/84839. [PubMed] [CrossRef] [Google Scholar]
  • Musaro A, Giacinti C, Borsellino M, Dobrowolny G, Pelosi L, Cairns L, Ottolenghi S, Cossu Thou, Bernardi Yard, Battistini L, Molinaro M, Rosenthal N. Stem jail cell-mediated musculus regeneration is enhanced by local isoform of insulin-like growth factor 1. Proc Natl Acad Sci U S A. 2004;101(5):1206–1210. doi: ten.1073/pnas.0303792101. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • Ota South, Uehara K, Nozaki Chiliad, Kobayashi T, Terada South, Tobita M, 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(9):1912–1922. doi: 10.1177/0363546511415239. [PubMed] [CrossRef] [Google Scholar]
  • Park JK, Ki MR, Lee EM, Kim AY, You lot 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. Jail cell Transplant. 2012;21(11):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: x.1038/337176a0. [PubMed] [CrossRef] [Google Scholar]
  • Powell CA, Smiley BL, Mills J, Vandenburgh HH. Mechanical stimulation improves tissue-engineered homo skeletal musculus. Am J Physiol 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 Due east, Poddar K, Beckman SA, Robbins PD, Nidernhofer LJ, Imbrogno 1000, Hannigan T, Mars WM, Wang B, Huard J. NF-kappaB inhibition reveals a novel office for HGF during skeletal muscle repair. Cell Death Dis. 2015;6 doi: 10.1038/cddis.2015.66. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rantanen J, Ranne J, Hurme T, Kalimo H. Denervated segments of injured skeletal muscle fibers are reinnervated past newly formed neuromuscular junctions. J Neuropathol Exp Neurol. 1995;54(2):188–194. doi: x.1097/00005072-199503000-00005. [PubMed] [CrossRef] [Google Scholar]
  • Relaix F, Zammit PS. Satellite cells are essential for skeletal muscle regeneration: the cell on the edge returns middle stage. Development. 2012;139(16):2845–2856. doi: 10.1242/dev.069088. [PubMed] [CrossRef] [Google Scholar]
  • Reurink G, Goudswaard GJ, Moen MH, Weir A, Verhaar JA, Bierma-Zeinstra SM, Maas Thou, Tol JL, Dutch Hamstring Injection Therapy Study I Platelet-rich plasma injections in astute muscle injury. North Engl J Med. 2014;370(26):2546–2547. doi: ten.1056/NEJMc1402340. [PubMed] [CrossRef] [Google Scholar]
  • Reurink Grand, Goudswaard GJ, Moen MH, Weir A, Verhaar JA, Bierma-Zeinstra SM, Maas Yard, Tol JL, Dutch HITsI Rationale, secondary outcome scores and 1-year follow-upwardly 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: 10.1136/bjsports-2014-094250. [PubMed] [CrossRef] [Google Scholar]
  • Rocheteau P, Vinet M, Chretien F. Dormancy and quiescence of skeletal muscle stem cells. Results Probl Cell Differ. 2015;56:215–235. doi: 10.1007/978-3-662-44608-9_10. [PubMed] [CrossRef] [Google Scholar]
  • Sadtler Thou, Estrellas Grand, 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 two cells. Scientific discipline. 2016;352(6283):366–370. doi: x.1126/scientific discipline.aad9272. [PMC costless article] [PubMed] [CrossRef] [Google Scholar]
  • Saera-Vila A, Kish PE, Kahana A. Fgf regulates dedifferentiation during skeletal muscle regeneration in adult zebrafish. Prison 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 L, Paldi A, Gayraud-Morel B, Guenou H, Malissen B, Tajbakhsh Due south, Galy A. Pax7-expressing satellite cells are indispensable for adult skeletal muscle regeneration. Development. 2011;138(17):3647–3656. doi: 10.1242/dev.067587. [PubMed] [CrossRef] [Google Scholar]
  • Sampaolesi M, Blot South, D'Antona Thousand, Granger N, Tonlorenzi R, Innocenzi A, Mognol P, Thibaud JL, Galvez BG, Barthelemy I, Perani L, Mantero S, Guttinger Grand, Pansarasa O, Rinaldi C, Cusella De Angelis MG, Torrente Y, Bordignon C, Bottinelli R, Cossu Thousand. Mesoangioblast stem cells ameliorate musculus function in dystrophic dogs. Nature. 2006;444(7119):574–579. doi: 10.1038/nature05282. [PubMed] [CrossRef] [Google Scholar]
  • Schiaffino Due south, Mammucari C. Regulation of skeletal muscle growth by the IGF1-Akt/PKB pathway: insights from genetic models. Skelet Muscle. 2011;ane(i):4. doi: 10.1186/2044-5040-1-four. [PMC complimentary article] [PubMed] [CrossRef] [Google Scholar]
  • Scholz D, Thomas Due south, Sass South, Podzuweit T. Angiogenesis and myogenesis every bit two facets of inflammatory post-ischemic tissue regeneration. Mol Cell Biochem. 2003;246(1-ii):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 musculus satellite cells in vitro. Muscle Nerve. 2000;23(two):239–245. doi: 10.1002/(SICI)1097-4598(200002)23:2<239::Help-MUS15>3.0.CO;ii-U. [PubMed] [CrossRef] [Google Scholar]
  • Skuk D, Goulet M, Roy B, Piette Five, Cote CH, Chapdelaine P, Hogrel JY, Paradis M, Bouchard JP, Sylvain G, Lachance JG, Tremblay JP. Offset examination of a "high-density injection" protocol for myogenic cell transplantation throughout large volumes of muscles in a Duchenne muscular dystrophy patient: eighteen months follow-upwards. Neuromuscul Disord. 2007;17(i):38–46. doi: 10.1016/j.nmd.2006.x.003. [PubMed] [CrossRef] [Google Scholar]
  • Taniguti AP, Pertille A, Matsumura CY, Santo Neto H, Marques MJ. Prevention of musculus fibrosis and myonecrosis in mdx mice by suramin, a TGF-beta1 blocker. Musculus Nerve. 2011;43(1):82–87. doi: 10.1002/mus.21869. [PubMed] [CrossRef] [Google Scholar]
  • Tatsumi R, Sheehan SM, Iwasaki H, Hattori A, Allen RE. Mechanical stretch induces activation of skeletal musculus satellite cells in vitro. Exp Cell Res. 2001;267(1):107–114. doi: x.1006/excr.2001.5252. [PubMed] [CrossRef] [Google Scholar]
  • Tedesco FS, Cossu G. Stem jail cell therapies for musculus disorders. Curr Opin Neurol. 2012;25(5):597–603. doi: x.1097/WCO.0b013e328357f288. [PubMed] [CrossRef] [Google Scholar]
  • Tedesco FS, Dellavalle A, Diaz-Manera J, Messina G, Cossu Yard. Repairing skeletal musculus: regenerative potential of skeletal muscle stalk cells. J Clin Invest. 2010;120(1):11–19. doi: x.1172/JCI40373. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Terada Southward, Ota S, Kobayashi M, Kobayashi T, Mifune Y, Takayama K, Witt M, Vadala Thou, Oyster Due north, Otsuka T, Fu FH, Huard J. Utilize of an antifibrotic agent improves the issue of platelet-rich plasma on muscle healing after injury. J Bone Articulation Surg Am. 2013;95(xi):980–988. doi: ten.2106/JBJS.L.00266. [PubMed] [CrossRef] [Google Scholar]
  • Tidball JG. Inflammatory cell response to acute musculus injury. Med Sci Sports Exerc. 1995;27(7):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(ii):R345–R353. doi: 10.1152/ajpregu.00454.2004. [PubMed] [CrossRef] [Google Scholar]
  • Tidball JG. Mechanisms of muscle injury, repair, and regeneration. Compr Physiol. 2011;1(4):2029–2062. [PubMed] [Google Scholar]
  • Tidball JG, Wehling-Henricks M. Macrophages promote muscle 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 gratis article] [PubMed] [CrossRef] [Google Scholar]
  • Tidball JG, Welc SS. Macrophage-Derived IGF-1 Is a Potent Coordinator of Myogenesis and Inflammation in Regenerating Muscle. Mol Ther. 2015;23(seven):1134–1135. doi: 10.1038/mt.2015.97. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • Toumi H, Best TM. The inflammatory response: friend or enemy for muscle injury? Br J Sports Med. 2003;37(4):284–286. doi: 10.1136/bjsm.37.iv.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 musculus. J Neuropathol Exp Neurol. 2001;60(6):588–597. doi: ten.1093/jnen/threescore.6.588. [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 L. To build a synapse: signaling pathways in neuromuscular junction assembly. Development. 2010;137(7):1017–1033. doi: ten.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 adult mouse skeletal musculus by specific isoforms of PDGF. J Cell Biol. 1990;111(four):1623–1629. doi: 10.1083/jcb.111.iv.1623. [PMC costless article] [PubMed] [CrossRef] [Google Scholar]
  • Yin H, Price F, Rudnicki MA. Satellite cells and the muscle stem cell niche. Physiol Rev. 2013;93(1):23–67. doi: x.1152/physrev.00043.2011. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
  • Zhao West, Lu H, Wang X, Ransohoff RM, Zhou L. CX3CR1 deficiency delays acute skeletal musculus injury repair past impairing macrophage functions. FASEB J. 2016;30(1):380–393. doi: x.1096/fj.xiv-270090. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zheng B, Cao B, Crisan M, Sun B, Li One thousand, Logar A, Yap S, Pollett JB, Drowley L, Cassino T, Gharaibeh B, Deasy BM, Huard J, Peault B. Prospective identification of myogenic endothelial cells in man skeletal muscle. Nat Biotechnol. 2007;25(9):1025–1034. doi: x.1038/nbt1334. [PubMed] [CrossRef] [Google Scholar]

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


How Long Does It Take Muscles To Grow Back Together After Being Repaired,

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

Posted by: meyerdencen1959.blogspot.com

0 Response to "How Long Does It Take Muscles To Grow Back Together After Being Repaired"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel