Comprehensive Physiology Wiley Online Library

Mechanisms of Muscle Injury, Repair, and Regeneration

Full Article on Wiley Online Library



Abstract

Skeletal muscle continuously adapts to changes in its mechanical environment through modifications in gene expression and protein stability that affect its physiological function and mass. However, mechanical stresses commonly exceed the parameters that induce adaptations, producing instead acute injury. Furthermore, the relatively superficial location of many muscles in the body leaves them further vulnerable to acute injuries by exposure to extreme temperatures, contusions, lacerations or toxins. In this article, the molecular, cellular, and mechanical factors that underlie muscle injury and the capacity of muscle to repair and regenerate are presented. Evidence shows that muscle injuries that are caused by eccentric contractions result from direct mechanical damage to myofibrils. However, muscle pathology following other acute injuries is largely attributable to damage to the muscle cell membrane. Many feaures in the injury‐repair‐regeneration cascade relate to the unregulated influx of calcium through membrane lesions, including: (i) activation of proteases and hydrolases that contribute muscle damage, (ii) activation of enzymes that drive the production of mitogens and motogens for muscle and immune cells involved in injury and repair, and (iii) enabling protein‐protein interactions that promote membrane repair. Evidence is also presented to show that the myogenic program that is activated by acute muscle injury and the inflammatory process that follows are highly coordinated, with myeloid cells playing a central role in modulating repair and regeneration. The early‐invading, proinflammatory M1 macrophages remove debris caused by injury and express Th1 cytokines that play key roles in regulating the proliferation, migration, and differentiation of satellite cells. The subsequent invasion by anti‐inflammatory, M2 macrophages promotes tissue repair and attenuates inflammation. Although this system provides an effective mechanism for muscle repair and regeneration following acute injury, it is dysregulated in chronic injuries. In this article, the process of muscle injury, repair and regeneration that occurs in muscular dystrophy is used as an example of chronic muscle injury, to highlight similarities and differences between the injury and repair processes that occur in acutely and chronically injured muscle. © 2011 American Physiological Society. Compr Physiol 1:2029‐2062, 2011.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1.

Lecture announcement for a presentation by Professor D.R. Wilkie given at the Institution of Electrical Engineers in London, UK on the subject of muscle physiology.

Figure 2. Figure 2.

Scanning electron micrograph of two skeletal muscle fibers terminating at their myotendinous junctions (MTJs), where they are mechanically coupled to tendon collagen fibers. Bundles of collagen fibers pass from the tendon in the bottom third of the micrograph to bind to the ends of the muscle fibers at the MTJ (between brackets). During muscle strain injuries, lesions occur at or near the MTJ depending on the state of activation of the fiber and the muscle experiencing the strain injury. Bar = 100 μm.

[Reproduced, with permission, from reference 238].
Figure 3. Figure 3.

Transmission electron micrographs of longitudinal sections through the myotendinous junction (MTJ) region of frog skeletal muscle that was strained to failure. (A) The upper portion of the micrograph shows tendon collagen fibers. The lower portion of the micrograph shows the MTJ of a muscle fiber that terminates with digit‐like extensions of the muscle cell into the tendon. This muscle was unstimulated and shows tissue tears occurred within the connective tissue, near the surface of the muscle fiber (outlined with arrowheads). Bar = 2.0 μm. (B) Complete separations (S) occurred in the connective tissue near the MTJs in unstimulated muscles strained to failure. Other cells in the same preparations show complete lesions within the muscle fiber near the MTJ. Bar = 2.5 μm. (C) Muscles that were strained to failure while stimulated at 20 Hz show that separation in the tissue occurred at the MTJ, along the external surface of the muscle fibers, leaving the digit‐like extensions of the cell (arrowheads) protruding into the lesion. Bar = 2.0 μm.

[Reproduced, with permission, from reference 242].
Figure 4. Figure 4.

Rabbit tibialis anterior muscle subjected to a strain injury. (A) The muscle on the left was photographed immediately after the strain injury and shows a hematoma (bracket) in the area of the distal, myotendinous junction (MTJ). The muscle on the right is an unstrained control. (B) A longitudinal section of a strained muscle preserved immediately following injury showing the distal MTJ region of muscle fibers (red). The dark, vertical band on the left of the micrograph is tendon. Note that the morphology of the muscle fibers near the tendon is disrupted. Bar = 80 μm.

[Reproduced, with permission, from reference 174].
Figure 5. Figure 5.

Rabbit tibialis anterior muscle subjected to a strain injury. (A) and (B) myotendinous junction (MTJ) region of strain‐injured muscle at 24‐h postinjury. Note the separation tissue separation in the MTJ region of the injured muscle between tendon (blue) and muscle fibers (red) (panel A) and the large number of red cells and leukocytes (arrow heads) in the injured MTJ region (panel B). (C) and (D) MTJ region of strain‐injured muscle at 48‐h postinjury. Increased numbers of leukocytes have entered the sites of tissue separation in the MTJ region (panels C and D) and necrotic fibers (brown) invaded by leukocytes are apparent at the sites of tissue damage (panel D). Bars in upper panels = 100 μm. Bars in lower panels = 50 μm.

[Reproduced, with permission, from reference 174.]
Figure 6. Figure 6.

Transmission electron micrograph of human skeletal muscle sampled after injury caused by eccentric contractions. (A) Focal disruption of myofibrillar structure in which Z‐disks are no longer aligned, Z‐disk streaming is present (arrowhead) and in some cases (*) normal myofibril structure is degraded. Bar = 3.5 μm. (B) Region of an injured muscle fiber that shows extreme degradation of myofibrillar structure, including extensive Z‐disk streaming (arrowheads). Bar = 4.5 μm.

[Reproduced, with permission, from reference 125].
Figure 7. Figure 7.

Simplified schematic representing some of the steps of the complement cascade that have been implicated in skeletal muscle injury. Current experimental data support a role for the classical pathway, a component of humoral immunity, and the alternative pathway, a component of innate immunity, in causing muscle damage. The potential involvement of the lectin pathway has not been explored. Each pathway progresses through multiple steps to lead to the proteolysis of C3. The products of C3 cleavage, C3a and C3b, can contribute to muscle inflammation and injury through multiple effects. C3a released into the extracellular space can increase the expression of adhesion molecules on the surface of leukocytes and endothelial cells that enables the more rapid extravasation of leukocytes into injured tissue. C3a can also act as a chemoattractant to bring more myeloid cells to the site of injury. C3b can amplify muscle inflammation and damage by participating in the cleavage of C5. C5a that is generated by C5 cleavage can increase histamine release by mast cells, which drives edema and further inflammation that can promote muscle damage. C5a can also increase the production of potentially cytotoxic free radicals by neutrophils and elevate the expression of Th1 cytokines, which could further elevate inflammation and inflammatory cell‐mediated damage to muscle cells. C5b contributes to muscle cell damage by associating with other proteins in the complement system to form a transmembrane, membrane‐attack complex that allows the unregulated flux of ions and small molecules across the target cell membrane, leading to cell lysis. Experimental data show that muscle cell damage during IR is less in CR2 null mice, which would have impaired activation of the classical pathway 7. Treatment of mice subjected to IR with sCR1 to block C3b or genetic ablation of C5 120 also reduced muscle damage, confirming involvement of the complement system in the injury, although not discriminating between specific pathways. Similarly, treatments with sCR1 reduce muscle damage in unloading/reloading 69.

Figure 8. Figure 8.

Schematic representation of the cycle of satellite cell activation, proliferation, and differentiation following muscle injury. Acute trauma of muscle fibers induces activation of satellite cells that normally reside in a quiescent state between the exterior surface of the muscle fiber's plasma membrane and the enveloping basal lamina. Activated satellite cells can then enter the interstitium between muscle fibers and proliferate to expand their numbers. Arrows marked “A” represent events that occur during the activation/proliferation stage of regeneration. Some of the activated cells then exit from the cell cycle and return to their niche as quiescent satellite cells to renew and maintain the satellite cell population. Other satellite cells migrate to the site where muscle repair and regeneration proceed, where they withdraw from the cell cycle to undergo differentiation. Arrows marked “B” represent events that occur during the early differentiation stage of regeneration. As part of their program of differentiation, they can fuse with existing fibers to contribute new nuclei to the fibers or fuse with other mononucleated myogenic cells to form new fibers. During these later stages of muscle regeneration, muscle nuclei align along the longitudinal axis of the muscle fiber, becoming central nuclei which is a characteristic of myotubes or muscle fibers experiencing repair after injury. Arrows marked “C” represent events that occur during the terminal differentiation stage of regeneration. Upon completion of terminal differentiation, the central nuclei migrate to the surface of the muscle fiber. [Adapted and modified, with permission, from reference 93].

Figure 9. Figure 9.

Schematic representation of a potential mechanism through which muscle injury could lead to satellite cell activation. Muscle membrane lesions or muscle stretching increases the calcium ion entry into muscle, producing an increase in nNOS activation. The elevated release of NO from muscle fibers can increase activation of MMP‐2 present in the extracellular space. MMP‐mediated cleavage of HSP core protein permits the release of HGF and FGF2 from their bound, inactive state to become active. HGF can then bind its receptor c‐met that is present on the surface of quiescent satellite cells to induce their activation, proliferation, and chemotaxis. Similarly, FGF2 can binds its receptor present on the satellite cell surface to drive proliferation and chemotaxis.

Figure 10. Figure 10.

Diagrammatic representation of the temporal relationship between the changes in the populations of inflammatory cells in muscle following acute injury and the changes in gene expression in muscle following injury. The early stages of muscle repair following injury are characterized by a contemporaneous invasion of neutrophils (PMN) and M1 macrophages with the activation of expression of transcription factors associated with the proliferative stage of myogenesis (e.g., MyoD and Myf5). Subsequent declines in the myeloid cells associated with Th1 inflammatory response and elevated numbers of leukocytes characteristic of a Th2 response, especially M2 macrophages, occurs at the time muscle elevates expression of transcription factors associated with the early differentiation stage of myogenesis (e.g., myogenin and MEF2). As inflammation further resolves, muscle cells undergo terminal differentiation and express elevated levels of muscle‐specific enzymes and structural proteins. [Adapted and modified, with permission, from reference 244].

Figure 11. Figure 11.

Diagram of alternative fates of macrophages activated to the M1 phenotype by Th1 cytokines or to the M2 phenotype by Th2 cytokines. In addition to promoting the M2 phenotype, IL‐10 can also deactivate the M1 phenotype. Following muscle injury, M1 macrophages dominate the inflammatory infiltrate between 1 and 2 days postinjury, when they can promote further analysis of muscle membranes by free radical production and remove cellular debris by phagocytosis. The M1 macrophages are they superceded by the M2 population between days 2 and 4 postinjury. M2 macrophages attenuate inflammation by deactivating M1 macrophages. They also promote muscle repair, growth, differentiation, and fibrosis.

Figure 12. Figure 12.

Simplified summary diagram of the competing roles played by IFNγ in muscle injury and repair. IFNγ can be expressed and secreted by M1 macrophages, further driving monocytes to the M1 macrophage phenotype and also activating neutrophils. IFNγ stimulation of neutrophils and M1 macrophages elevates their expression of constitutively active iNOS, increasing the production of NO to levels that can be cytolytic. Activation of neutrophils by IFNγ can also increase their production and release of MPO, which can also promote cytotoxicity and muscle damage. These functions of IFNγ appear to be most important during the Th1 inflammatory response. IFNγ can also act directly on muscle cells to increase their proliferation, attract them to sites of tissue damage and inhibit apoptosis. These functions of IFNγ appear to be most important during the Th2 inflammatory response while muscle repair and regeneration proceed.

Figure 13. Figure 13.

Simplified summary diagram of the competing roles played by tumor necrosis factor‐alpha (TNFα) in muscle injury and repair. TNFα secreted by M1 macrophages can activate NFκB in monocytes and macrophage to drive them to an M1 macrophage phenotype. NFκB activation promotes the expression of iNOS in macrophages, thereby increasing the production of NO to levels that can be cytotoxic. TNFα also function as a chemoattractant to myogenic cells, drawing them to sites of muscle injury. These functions of TNFα appear to be most important during the Th1 inflammatory response. TNFα can also activate NFκB in proliferative, satellite cells to increase their proliferation and suppress their differentiation. TNFα can also activate signaling via p38 in satellite cells to promote their differentiation and fusion. These latter functions of TNFα are expected to be most important during the Th2 inflammatory response when muscle repair and regeneration occur.

Figure 14. Figure 14.

Immunohistochemistry of an inflammatory lesion in muscle in the mdx mouse model of Duchenne muscular dystrophy. This lesion contains pro‐inflammatory, M1 macrophages (red) that can promote muscle damage, as well as anti‐inflammatory, M2 macrophages (orange), that can affect regeneration and fibrosis. The lesion also contains muscle satellite cells (green) whose proliferation and differentiation can be modulated by the neighboring macrophages. The antibodies used for labeling were anti‐F4/80 (red fluorophore) which binds all macrophages and anti‐CD206 (green fluorophore) that binds M2 macrophages and satellite cells. Hoechts labeling (blue) shows nuclei. Bar = 50 μm.

[Reproduced, with permission, from reference 244].
Figure 15. Figure 15.

Diagram of pathways for arginine metabolism that contribute to muscle injury, repair, and fibrosis. Arginine is a conditionally essential amino acid that is responsible for substrate limitation of NOS activity and arginase activity under conditions of rapid growth during development or during tissue repair following injury. Thus, NOS and arginase can compete for substrate in inflamed tissues following injury. Arginine metabolism by iNOS drives the production of NO to high levels that contribute to the formation of more toxic free radicals such as peroxynitrite (ONOO) that may contribute to the formation of membrane lesions and to denaturing of proteins, lipids, and nucleic acids. Arginine metabolism by this pathway would be amplified during the Th1 inflammatory response to injury. However, arginine is also metabolized by arginase present in M2 macrophage to yield its hydrolysates L‐ornithine and urea. L‐ornithine is then further metabolized to yield L‐proline that is necessary for synthesis of collagen and to yield other metabolites that drive the proliferation of cells, including fibroblasts. Thus, the amplification of this pathway of arginine metabolism in M2 macrophage during the Th2 inflammatory response would contribute to fibrosis and would healing.



Figure 1.

Lecture announcement for a presentation by Professor D.R. Wilkie given at the Institution of Electrical Engineers in London, UK on the subject of muscle physiology.



Figure 2.

Scanning electron micrograph of two skeletal muscle fibers terminating at their myotendinous junctions (MTJs), where they are mechanically coupled to tendon collagen fibers. Bundles of collagen fibers pass from the tendon in the bottom third of the micrograph to bind to the ends of the muscle fibers at the MTJ (between brackets). During muscle strain injuries, lesions occur at or near the MTJ depending on the state of activation of the fiber and the muscle experiencing the strain injury. Bar = 100 μm.

[Reproduced, with permission, from reference 238].


Figure 3.

Transmission electron micrographs of longitudinal sections through the myotendinous junction (MTJ) region of frog skeletal muscle that was strained to failure. (A) The upper portion of the micrograph shows tendon collagen fibers. The lower portion of the micrograph shows the MTJ of a muscle fiber that terminates with digit‐like extensions of the muscle cell into the tendon. This muscle was unstimulated and shows tissue tears occurred within the connective tissue, near the surface of the muscle fiber (outlined with arrowheads). Bar = 2.0 μm. (B) Complete separations (S) occurred in the connective tissue near the MTJs in unstimulated muscles strained to failure. Other cells in the same preparations show complete lesions within the muscle fiber near the MTJ. Bar = 2.5 μm. (C) Muscles that were strained to failure while stimulated at 20 Hz show that separation in the tissue occurred at the MTJ, along the external surface of the muscle fibers, leaving the digit‐like extensions of the cell (arrowheads) protruding into the lesion. Bar = 2.0 μm.

[Reproduced, with permission, from reference 242].


Figure 4.

Rabbit tibialis anterior muscle subjected to a strain injury. (A) The muscle on the left was photographed immediately after the strain injury and shows a hematoma (bracket) in the area of the distal, myotendinous junction (MTJ). The muscle on the right is an unstrained control. (B) A longitudinal section of a strained muscle preserved immediately following injury showing the distal MTJ region of muscle fibers (red). The dark, vertical band on the left of the micrograph is tendon. Note that the morphology of the muscle fibers near the tendon is disrupted. Bar = 80 μm.

[Reproduced, with permission, from reference 174].


Figure 5.

Rabbit tibialis anterior muscle subjected to a strain injury. (A) and (B) myotendinous junction (MTJ) region of strain‐injured muscle at 24‐h postinjury. Note the separation tissue separation in the MTJ region of the injured muscle between tendon (blue) and muscle fibers (red) (panel A) and the large number of red cells and leukocytes (arrow heads) in the injured MTJ region (panel B). (C) and (D) MTJ region of strain‐injured muscle at 48‐h postinjury. Increased numbers of leukocytes have entered the sites of tissue separation in the MTJ region (panels C and D) and necrotic fibers (brown) invaded by leukocytes are apparent at the sites of tissue damage (panel D). Bars in upper panels = 100 μm. Bars in lower panels = 50 μm.

[Reproduced, with permission, from reference 174.]


Figure 6.

Transmission electron micrograph of human skeletal muscle sampled after injury caused by eccentric contractions. (A) Focal disruption of myofibrillar structure in which Z‐disks are no longer aligned, Z‐disk streaming is present (arrowhead) and in some cases (*) normal myofibril structure is degraded. Bar = 3.5 μm. (B) Region of an injured muscle fiber that shows extreme degradation of myofibrillar structure, including extensive Z‐disk streaming (arrowheads). Bar = 4.5 μm.

[Reproduced, with permission, from reference 125].


Figure 7.

Simplified schematic representing some of the steps of the complement cascade that have been implicated in skeletal muscle injury. Current experimental data support a role for the classical pathway, a component of humoral immunity, and the alternative pathway, a component of innate immunity, in causing muscle damage. The potential involvement of the lectin pathway has not been explored. Each pathway progresses through multiple steps to lead to the proteolysis of C3. The products of C3 cleavage, C3a and C3b, can contribute to muscle inflammation and injury through multiple effects. C3a released into the extracellular space can increase the expression of adhesion molecules on the surface of leukocytes and endothelial cells that enables the more rapid extravasation of leukocytes into injured tissue. C3a can also act as a chemoattractant to bring more myeloid cells to the site of injury. C3b can amplify muscle inflammation and damage by participating in the cleavage of C5. C5a that is generated by C5 cleavage can increase histamine release by mast cells, which drives edema and further inflammation that can promote muscle damage. C5a can also increase the production of potentially cytotoxic free radicals by neutrophils and elevate the expression of Th1 cytokines, which could further elevate inflammation and inflammatory cell‐mediated damage to muscle cells. C5b contributes to muscle cell damage by associating with other proteins in the complement system to form a transmembrane, membrane‐attack complex that allows the unregulated flux of ions and small molecules across the target cell membrane, leading to cell lysis. Experimental data show that muscle cell damage during IR is less in CR2 null mice, which would have impaired activation of the classical pathway 7. Treatment of mice subjected to IR with sCR1 to block C3b or genetic ablation of C5 120 also reduced muscle damage, confirming involvement of the complement system in the injury, although not discriminating between specific pathways. Similarly, treatments with sCR1 reduce muscle damage in unloading/reloading 69.



Figure 8.

Schematic representation of the cycle of satellite cell activation, proliferation, and differentiation following muscle injury. Acute trauma of muscle fibers induces activation of satellite cells that normally reside in a quiescent state between the exterior surface of the muscle fiber's plasma membrane and the enveloping basal lamina. Activated satellite cells can then enter the interstitium between muscle fibers and proliferate to expand their numbers. Arrows marked “A” represent events that occur during the activation/proliferation stage of regeneration. Some of the activated cells then exit from the cell cycle and return to their niche as quiescent satellite cells to renew and maintain the satellite cell population. Other satellite cells migrate to the site where muscle repair and regeneration proceed, where they withdraw from the cell cycle to undergo differentiation. Arrows marked “B” represent events that occur during the early differentiation stage of regeneration. As part of their program of differentiation, they can fuse with existing fibers to contribute new nuclei to the fibers or fuse with other mononucleated myogenic cells to form new fibers. During these later stages of muscle regeneration, muscle nuclei align along the longitudinal axis of the muscle fiber, becoming central nuclei which is a characteristic of myotubes or muscle fibers experiencing repair after injury. Arrows marked “C” represent events that occur during the terminal differentiation stage of regeneration. Upon completion of terminal differentiation, the central nuclei migrate to the surface of the muscle fiber. [Adapted and modified, with permission, from reference 93].



Figure 9.

Schematic representation of a potential mechanism through which muscle injury could lead to satellite cell activation. Muscle membrane lesions or muscle stretching increases the calcium ion entry into muscle, producing an increase in nNOS activation. The elevated release of NO from muscle fibers can increase activation of MMP‐2 present in the extracellular space. MMP‐mediated cleavage of HSP core protein permits the release of HGF and FGF2 from their bound, inactive state to become active. HGF can then bind its receptor c‐met that is present on the surface of quiescent satellite cells to induce their activation, proliferation, and chemotaxis. Similarly, FGF2 can binds its receptor present on the satellite cell surface to drive proliferation and chemotaxis.



Figure 10.

Diagrammatic representation of the temporal relationship between the changes in the populations of inflammatory cells in muscle following acute injury and the changes in gene expression in muscle following injury. The early stages of muscle repair following injury are characterized by a contemporaneous invasion of neutrophils (PMN) and M1 macrophages with the activation of expression of transcription factors associated with the proliferative stage of myogenesis (e.g., MyoD and Myf5). Subsequent declines in the myeloid cells associated with Th1 inflammatory response and elevated numbers of leukocytes characteristic of a Th2 response, especially M2 macrophages, occurs at the time muscle elevates expression of transcription factors associated with the early differentiation stage of myogenesis (e.g., myogenin and MEF2). As inflammation further resolves, muscle cells undergo terminal differentiation and express elevated levels of muscle‐specific enzymes and structural proteins. [Adapted and modified, with permission, from reference 244].



Figure 11.

Diagram of alternative fates of macrophages activated to the M1 phenotype by Th1 cytokines or to the M2 phenotype by Th2 cytokines. In addition to promoting the M2 phenotype, IL‐10 can also deactivate the M1 phenotype. Following muscle injury, M1 macrophages dominate the inflammatory infiltrate between 1 and 2 days postinjury, when they can promote further analysis of muscle membranes by free radical production and remove cellular debris by phagocytosis. The M1 macrophages are they superceded by the M2 population between days 2 and 4 postinjury. M2 macrophages attenuate inflammation by deactivating M1 macrophages. They also promote muscle repair, growth, differentiation, and fibrosis.



Figure 12.

Simplified summary diagram of the competing roles played by IFNγ in muscle injury and repair. IFNγ can be expressed and secreted by M1 macrophages, further driving monocytes to the M1 macrophage phenotype and also activating neutrophils. IFNγ stimulation of neutrophils and M1 macrophages elevates their expression of constitutively active iNOS, increasing the production of NO to levels that can be cytolytic. Activation of neutrophils by IFNγ can also increase their production and release of MPO, which can also promote cytotoxicity and muscle damage. These functions of IFNγ appear to be most important during the Th1 inflammatory response. IFNγ can also act directly on muscle cells to increase their proliferation, attract them to sites of tissue damage and inhibit apoptosis. These functions of IFNγ appear to be most important during the Th2 inflammatory response while muscle repair and regeneration proceed.



Figure 13.

Simplified summary diagram of the competing roles played by tumor necrosis factor‐alpha (TNFα) in muscle injury and repair. TNFα secreted by M1 macrophages can activate NFκB in monocytes and macrophage to drive them to an M1 macrophage phenotype. NFκB activation promotes the expression of iNOS in macrophages, thereby increasing the production of NO to levels that can be cytotoxic. TNFα also function as a chemoattractant to myogenic cells, drawing them to sites of muscle injury. These functions of TNFα appear to be most important during the Th1 inflammatory response. TNFα can also activate NFκB in proliferative, satellite cells to increase their proliferation and suppress their differentiation. TNFα can also activate signaling via p38 in satellite cells to promote their differentiation and fusion. These latter functions of TNFα are expected to be most important during the Th2 inflammatory response when muscle repair and regeneration occur.



Figure 14.

Immunohistochemistry of an inflammatory lesion in muscle in the mdx mouse model of Duchenne muscular dystrophy. This lesion contains pro‐inflammatory, M1 macrophages (red) that can promote muscle damage, as well as anti‐inflammatory, M2 macrophages (orange), that can affect regeneration and fibrosis. The lesion also contains muscle satellite cells (green) whose proliferation and differentiation can be modulated by the neighboring macrophages. The antibodies used for labeling were anti‐F4/80 (red fluorophore) which binds all macrophages and anti‐CD206 (green fluorophore) that binds M2 macrophages and satellite cells. Hoechts labeling (blue) shows nuclei. Bar = 50 μm.

[Reproduced, with permission, from reference 244].


Figure 15.

Diagram of pathways for arginine metabolism that contribute to muscle injury, repair, and fibrosis. Arginine is a conditionally essential amino acid that is responsible for substrate limitation of NOS activity and arginase activity under conditions of rapid growth during development or during tissue repair following injury. Thus, NOS and arginase can compete for substrate in inflamed tissues following injury. Arginine metabolism by iNOS drives the production of NO to high levels that contribute to the formation of more toxic free radicals such as peroxynitrite (ONOO) that may contribute to the formation of membrane lesions and to denaturing of proteins, lipids, and nucleic acids. Arginine metabolism by this pathway would be amplified during the Th1 inflammatory response to injury. However, arginine is also metabolized by arginase present in M2 macrophage to yield its hydrolysates L‐ornithine and urea. L‐ornithine is then further metabolized to yield L‐proline that is necessary for synthesis of collagen and to yield other metabolites that drive the proliferation of cells, including fibroblasts. Thus, the amplification of this pathway of arginine metabolism in M2 macrophage during the Th2 inflammatory response would contribute to fibrosis and would healing.

References
 1. Acharyya S, Villalta SA, Bakkar N, Bupha‐Intr T, Janssen PM, Carathers M, Li ZW, Beg AA, Ghosh S, Sahenk Z, Weinstein M, Gardner KL, Rafael‐Fortney JA, Karin M, Tidball JG, Baldwin AS, Guttridge DC. Interplay of IKK/NF‐kappaB signaling in macrophages and myofibers promotes muscle degeneration in Duchenne muscular dystrophy. J Clin Invest 117: 889‐901, 2007.
 2. Allen RE, Boxhorn LK. Inhibition of skeletal muscle satellite cell differentiation by transforming growth factor‐beta. J Cell Physiol 133: 567‐572, 1987.
 3. Allen RE, Boxhorn LK. Regulation of skeletal muscle satellite cell proliferation and differentiation by transforming growth factor‐beta, insulin‐like growth factor I, and fibroblast growth factor. J Cell Physiol 138: 311‐315, 1989.
 4. Anderson JE, Bressler BH, Ovalle WK. Functional regeneration in the hindlimb skeletal muscle of the mdx mouse. J Muscle Res Cell Motil 9: 499‐515, 1988.
 5. Armstrong RB, Duan C, Delp MD, Hayes DA, Glenn GM, Allen GD. Elevations in rat soleus muscle [Ca2+] with passive stretch. J Appl Physiol 74: 2990‐2997, 1993.
 6. Arnold L, Henry A, Poron F, Baba‐Amer Y, van Rooijen N, Plonquet A, Gherardi RK, Chazaud B. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exper Med 204: 1057‐1069, 2007.
 7. Austen WG Jr., Zhang M, Chan R, Friend D, Hechtman HB, Carroll MC, Moore FD, Jr. Murine hindlimb reperfusion injury can be initiated by a self‐reactive monoclonal IgM. Surgery 136: 401‐406, 2004.
 8. Bakkar N, Wang J, Ladner KJ, Wang H, Dahlman JM, Carathers M, Acharyya S, Rudnicki MA, Hollenbach AD, Guttridge DC. IKK/NF‐kappaB regulates skeletal myogenesis via a signaling switch to inhibit differentiation and promote mitochondrial biogenesis. J Cell Biol 180: 787‐802, 2008.
 9. Balon TW, Nadler JL. Nitric oxide release is present from incubated skeletal muscle preparations. J Appl Physiol 77: 2519‐2521, 1994.
 10. Balnave CD, Davey DF, Allen DG. Distribution of sarcomere length and intracellular calcium in mouse skeletal muscle following stretch‐induced injury. J Physiol 502: 649‐659, 1997.
 11. Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, Williamson R, McNeil PL, Campbell KP. Defective membrane repair in dysferlin‐deficient muscular dystrophy. Nature 423: 168‐172, 2003.
 12. Bansal V, Ochoa JB. Arginine availability, arginase, and the immune response. Curr Opinion Clin Nutrition Metab Care 6: 223‐228, 2003.
 13. Barbul A. Proline precursors to sustain Mammalian collagen synthesis. J Nutrition 138: 2021S‐2024S, 2008.
 14. Barker JE, Knight KR, Romeo R, Hurley JV, Morrison WA, Stewart AG. Targeted disruption of the nitric oxide synthase 2 gene protects against ischaemia/reperfusion injury to skeletal muscle. J Pathol 194: 109‐115, 2001.
 15. Belcastro AN, Arthur GD, Albisser TA, Raj DA. Heart, liver, and skeletal muscle myeloperoxidase activity during exercise. J Appl Physiol 80: 1331‐1335, 1996.
 16. Bernasconi P, Torchiana E, Confalonieri P, Brugnoni R, Barresi R, Mora M, Cornelio F, Morandi L, 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 96: 1137‐1144, 1995.
 17. Bischoff R. A satellite cell mitogen from crushed adult muscle. Devel Biol 115: 140‐147, 1986.
 18. Bischoff R. Chemotaxis of skeletal muscle satellite cells. Dev Dyn 208: 505‐515, 1997.
 19. Blau HM, Webster C, Pavlath GK. Defective myoblasts identified in Duchenne muscular dystrophy. Proc Nat Acad Sci USA 80: 4856‐4860, 1983.
 20. Bockhold KJ, Rosenblatt JD, Partridge TA. Aging normal and dystrophic mouse muscle: Analysis of myogenicity in cultures of living single fibers. Muscle Nerve 21: 173‐183, 1998.
 21. Bonecchi R, Polentarutti N, Luini W, Borsatti A, Bernasconi S, Locati M, Power C, Proudfoot A, Wells TN, Mackay C, Mantovani A, Sozzani S. Up‐regulation of CCR1 and CCR3 and induction of chemotaxis to CC chemokines by IFN‐gamma in human neutrophils. J Immunol 162: 474‐479, 1999.
 22. Boriek AM, Capetanaki Y, Hwang W, Officer T, Badshah M, Rodarte J, Tidball JG. Desmin integrates the three‐dimensional mechanical properties of muscles. Am J Physiol 280: C46‐52, 2001.
 23. Bortolotto SK, Morrison WA, Han X, Messina A. Mast cells play a pivotal role in ischaemia reperfusion injury to skeletal muscles. Lab Invest 84: 1103‐1111, 2004.
 24. Bredt DS, Snyder SH. Isolation of nitric oxide synthetase, a calmodulin‐requiring enzyme. Proc Nat Acad Sci USA 87: 682‐685, 1990.
 25. Brenman JE, Chao DS, Xia H, Aldape K, Bredt DS. Nitric oxide synthase complexed with dystrophin and absent from skeletal muscle sarcolemma in Duchenne muscular dystrophy. Cell 82: 743‐752, 1995.
 26. Brickson S, Ji LL, Schell K, Olabisi R, St Pierre Schneider B, Best TM. M1/70 attenuates blood‐borne neutrophil oxidants, activation, and myofiber damage following stretch injury. J Appl Physiol 95: 969‐976, 2003.
 27. Brooks SV, Zerba E, Faulkner JA. Injury to muscle fibres after single stretches of passive and maximally stimulated muscles in mice. J Physiol 488: 459‐469, 1995.
 28. Buechler C, Ritter M, Orso E, Langmann T, Klucken J, Schmitz G. Regulation of scavenger receptor CD163 expression in human monocytes and macrophages by pro‐ and antiinflammatory stimuli. J Leukocyte Biol 67: 97‐103, 2000.
 29. Bury TB, Pirnay F. Effect of prolonged exercise on neutrophil myeloperoxidase secretion. Intern J Sports Med 16: 410‐412, 1995.
 30. Cavaillon JM, Fitting C, Haeffner‐Cavaillon N. Recombinant C5a enhances interleukin 1 and tumor necrosis factor release by lipopolysaccharide‐stimulated monocytes and macrophages. Eur J Immunol 20(2): 253‐7, 1990.
 31. Cenacchi G, Fanin M, De Giorgi LB, Angelini C. Ultrastructural changes in dysferlinopathy support defective membrane repair mechanism. J Clin Pathol 58: 190‐195, 2005.
 32. Chabot B, Stephenson DA, Chapman VM, Besmer P, Bernstein A. The proto‐oncogene c‐kit encoding a transmembrane tyrosine kinase receptor maps to the mouse W locus. Nature 335: 88‐89, 1988.
 33. Cheeseman KH, Slater TF. An introduction to free radical biochemistry. Brit Med Bull 49: 481‐493, 1993.
 34. Chen SE, Gerken E, Zhang Y, Zhan M, Mohan RK, Li AS, Reid MB, Li YP. Role of TNF‐α signaling in regeneration of cardiotoxin‐injured muscle. Am J Physiol 289: C1179‐1187, 2005.
 35. Cheng M, Nguyen MH, Fantuzzi G, Koh TJ. Endogenous interferon‐gamma is required for efficient skeletal muscle regeneration. Am J Physiol 294: C1183‐1191, 2008.
 36. Chenoweth DE, Hugli TE. Demonstration of specific C5a receptor on intact human polymorphonuclear leukocytes. Proc Nat Acad Sci USA 75: 3943‐3947, 1978.
 37. Chiu YH, Hornsey MA, Klinge L, Jorgensen LH, Laval SH, Charlton R, Barresi R, Straub V, Lochmuller H, Bushby K. Attenuated muscle regeneration is a key factor in dysferlin‐deficient muscular dystrophy. Hum Molec Genetics 18: 1976‐1989, 2009.
 38. Clarke MS, Feeback DL. Mechanical load induces sarcoplasmic wounding and FGF release in differentiated human skeletal muscle cultures. FASEB J 10: 502‐509, 1996.
 39. Clarke MS, Khakee R, McNeil PL. Loss of cytoplasmic basic fibroblast growth factor from physiologically wounded myofibers of normal and dystrophic muscle. J Cell Sci 106: 121‐133, 1993.
 40. Clarkson PM, Apple FS, Byrnes WC, McCormick KM, Triffletti P. Creatine kinase isoforms following isometric exercise. Muscle Nerve 10: 41‐44, 1987.
 41. Collins CA, Partridge TA. Self‐renewal of the adult skeletal muscle satellite cell. Cell Cycle 4: 1338‐1341, 2005.
 42. Contreras‐Shannon V, Ochoa O, Reyes‐Reyna SM, Sun D, Michalek JE, Kuziel WA, McManus LM, Shireman PK. Fat accumulation with altered inflammation and regeneration in skeletal muscle of CCR2−/− mice following ischemic injury. Am J Physiol 292: C953‐967, 2007.
 43. Cornelison DD, Olwin BB, Rudnicki MA, Wold BJ. MyoD(−/−) satellite cells in single‐fiber culture are differentiation defective and MRF4 deficient. Develop Biol 224: 122‐137, 2000.
 44. Cornelison DD, Wold BJ. Single‐cell analysis of regulatory gene expression in quiescent and activated mouse skeletal muscle satellite cells. Develop Biol 191: 270‐283, 1997.
 45. Coulton GR, Curtin NA, Morgan JE, Partridge TA. The mdx mouse skeletal muscle myopathy: II. Contractile properties. Neuropathol Appl Neurobiol 14: 299‐314, 1988.
 46. Csete M, Walikonis J, Slawny N, Wei Y, Korsnes S, Doyle JC, Wold B. Oxygen‐mediated regulation of skeletal muscle satellite cell proliferation and adipogenesis in culture. J Cell Physiol 189: 189‐196, 2001.
 47. Cullen MJ, Jaros E. Ultrastructure of the skeletal muscle in the X chromosome‐linked dystrophic (mdx) mouse. Comparison with Duchenne muscular dystrophy. Acta Neuropathol 77: 69‐81, 1988.
 48. Curran JN, Winter DC, Bouchier‐Hayes D. Biological fate and clinical implications of arginine metabolism in tissue healing. Wound Repair Regen 14: 376‐386, 2006.
 49. Davis DB, Doherty KR, Delmonte AJ, McNally EM. Calcium‐sensitive phospholipid binding properties of normal and mutant ferlin C2 domains. J Biol Chem 277: 22883‐22888, 2002.
 50. Decary S, Mouly V, Hamida CB, Sautet A, Barbet JP, Butler‐Browne GS. Replicative potential and telomere length in human skeletal muscle: Implications for satellite cell‐mediated gene therapy. Human Gene Ther 8: 1429‐1438, 1997.
 51. Deconinck N, Tinsley J, De Backer F, Fisher R, Kahn D, Phelps S, Davies K, Gillis JM. Expression of truncated utrophin leads to major functional improvements in dystrophin‐deficient muscles of mice. Nature Med 3: 1216‐1221, 1997.
 52. Dellorusso C, Crawford RW, Chamberlain JS, Brooks SV. Tibialis anterior muscles in mdx mice are highly susceptible to contraction‐induced injury. J Muscle Res Cell Motil 22: 467‐475, 2001.
 53. Dennis EA. Diversity of group types, regulation, and function of phospholipase A2. J Biol Chem 269: 13057‐13060, 1994.
 54. DiCorleto PE Bowen‐Pope DF. Cultured endothelial cells produce a platelet‐derived growth factor‐like protein. Proc Nat Acad Sci USA 80: 1919‐1923, 1983.
 55. DiMario J, Buffinger N, Yamada S, Strohman RC. Fibroblast growth factor in the extracellular matrix of dystrophic (mdx) mouse muscle. Science 244: 688‐690, 1989.
 56. Disatnik MH, Dhawan J, Yu Y, Beal MF, Whirl MM, Franco AA, Rando TA. Evidence of oxidative stress in mdx mouse muscle: Studies of the pre‐necrotic state. J Neurol Sci 161: 77‐84, 1998.
 57. Dudeck A, Leist M, Rubant S, Zimmermann A, Dudeck J, Boehncke WH, Maurer M. Immature mast cells exhibit rolling and adhesion to endothelial cells and subsequent diapedesis triggered by E‐ and P‐selectin, VCAM‐1 and PECAM‐1. Exper Dermatol 19: 424‐434, 2010.
 58. Dufaux B, Order U. Complement activation after prolonged exercise. Clin Chim Acta 179: 45‐49, 1989.
 59. Duncan CJ. The role of phospholipase A2 in calcium‐induced damage in cardiac and skeletal muscle. Cell Tiss Res 253: 457‐462, 1988.
 60. Duncan CJ, Jackson MJ. Different mechanisms mediate structural changes and intracellular enzyme efflux following damage to skeletal muscle. J Cell Sci 87: 183‐188, 1987.
 61. Ember JA, Sanderson SD, Hugli TE, Morgan EL. Induction of interleukin‐8 synthesis from monocytes by human C5a anaphylatoxin. Am J Pathol 144: 393‐403, 1994.
 62. Evers B, Odemis V, Gerngross H. Intramuscular oxygen partial pressure in patients with chronic exertional compartment syndrome (CECS). Adv Exper Med Biol 428: 311‐316, 1997.
 63. Fadok VA, Bratton DL, Guthrie L, Henson PM. Differential effects of apoptotic versus lysed cells on macrophage production of cytokines: Role of proteases. J Immunol 166: 6847‐6854, 2001.
 64. Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF‐beta, PGE2, and PAF. J Clinic Invest 101: 890‐898, 1998.
 65. Fielding RA, Manfredi TJ, Ding W, Fiatarone MA, Evans WJ, Cannon JG. Acute phase response in exercise. III. Neutrophil and IL‐1 beta accumulation in skeletal muscle. Am J Physiol 265: R166‐172, 1993.
 66. Fleming SD, Shea‐Donohue T, Guthridge JM, Kulik L, Waldschmidt TJ, Gipson MG, Tsokos GC, Holers VM. Mice deficient in complement receptors 1 and 2 lack a tissue injury‐inducing subset of the natural antibody repertoire. J Immunol 169: 2126‐2133, 2002.
 67. Formigli L, Lombardo LD, Adembri C, Brunelleschi S, Ferrari E, Novelli GP. Neutrophils as mediators of human skeletal muscle ischemia‐reperfusion syndrome. Human Pathol 23: 627‐634, 1992.
 68. Fredsted A, Gissel H, Madsen K, Clausen T. Causes of excitation‐induced muscle cell damage in isometric contractions: Mechanical stress or calcium overload? Am J Physiol 292: R2249‐2258, 2007.
 69. Frenette J, Cai B, Tidball JG. Complement activation promotes muscle inflammation during modified muscle use. Am J Pathol 156: 2103‐2110, 2000.
 70. Friden J. Changes in human skeletal muscle induced by long‐term eccentric exercise. Cell Tiss Res 236: 365‐372, 1984.
 71. Friden J, Lieber RL. Segmental muscle fiber lesions after repetitive eccentric contractions. Cell Tiss Res 293: 165‐171, 1998.
 72. Friden J, Seger J, Sjostrom M, Ekblom B. Adaptive response in human skeletal muscle subjected to prolonged eccentric training. Internat J Sports Med 4: 177‐183, 1983.
 73. Friden J, Sjostrom M, Ekblom B. A morphological study of delayed muscle soreness. Experientia 37: 506‐507, 1981.
 74. Fuchtbauer EM, Westphal H. MyoD and myogenin are coexpressed in regenerating skeletal muscle of the mouse. Dev Dyn 193: 34‐39, 1992.
 75. Gabriel A, Porrino ML, Stephenson LL, Zamboni WA. Effect of l‐arginine on leukocyte adhesion in ischemia‐reperfusion injury. Plastic Reconstruct Surg 113: 1698‐1702, 2004.
 76. Garrett WE Jr., Nikolaou PK, Ribbeck BM, Glisson RR, Seaber AV. The effect of muscle architecture on the biomechanical failure properties of skeletal muscle under passive extension. Am J Sports Med 16: 7‐12, 1988.
 77. Geissler EN, Russell ES. Analysis of the hematopoietic effects of new dominant spotting (W) mutations of the mouse. II. Effects on mast cell development. Exper Hematol 11: 461‐466, 1983.
 78. Glover L, Brown RH Jr. Dysferlin in membrane trafficking and patch repair. Traffic 8: 785‐794, 2007.
 79. Goldstein IM, Roos D, Kaplan HB, Weissmann G. Complement and immunoglobulins stimulate superoxide production by human leukocytes independently of phagocytosis. J Clin Invest 56: 1155‐1163, 1975.
 80. Goodman MG, Chenoweth DE, Weigle WO. Induction of interleukin 1 secretion and enhancement of humoral immunity by binding of human C5a to macrophage surface C5a receptors. J Exp Med Sep 1: 156(3): 912‐917, 1982.
 81. Gopalakrishnakone P, Dempster DW, Hawgood BJ, Elder HY. Cellular and mitochondrial changes induced in the structure of murine skeletal muscle by crotoxin, a neurotoxic phospholipase A2 complex. Toxicon 22: 85‐98, 1984.
 82. Gordon AM, Huxley AF, Julian FJ. The variation in isometric tension with sarcomere length in vertebrate muscle fibres. J Physiol 184: 170‐192, 1966.
 83. Gordon S. Alternative activation of macrophages. Nature Rev 3: 23‐35, 2003.
 84. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nature Rev 5: 953‐964, 2005.
 85. Graham IL, Gresham HD, Brown EJ. An immobile subset of plasma membrane CD11b/CD18 (Mac‐1) is involved in phagocytosis of targets recognized by multiple receptors. J Immunol 142: 2352‐2358, 1989.
 86. Granger BL, Lazarides E. Desmin and vimentin coexist at the periphery of the myofibril Z disc. Cell 18: 1053‐1063, 1979.
 87. Grounds MD, Garrett KL, Lai MC, Wright WE, Beilharz MW. Identification of skeletal muscle precursor cells in vivo by use of MyoD1 and myogenin probes. Cell Tiss Res 267: 99‐104, 1992.
 88. Gutierrez JM, Arce V, Brenes F, Chaves F. Changes in myofibrillar components after skeletal muscle necrosis induced by a myotoxin isolated from the venom of the snake Bothrops asper. Exper Molec Pathol 52: 25‐36, 1990.
 89. Gutierrez JM, Ownby CL. Skeletal muscle degeneration induced by venom phospholipases A2: Insights into the mechanisms of local and systemic myotoxicity. Toxicon 42: 915‐931, 2003.
 90. Guttridge DC, Albanese C, Reuther JY, Pestell RG, Baldwin AS Jr. NF‐kappaB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Mol Cell Biol 19: 5785‐5799, 1999.
 91. Han J, Jiang Y, Li Z, Kravchenko VV, Ulevitch RJ. Activation of the transcription factor MEF2C by the MAP kinase p38 in inflammation. Nature 386: 296‐299, 1997.
 92. Harris JB. Myotoxic phospholipases A2 and the regeneration of skeletal muscles. Toxicon 42: 933‐945, 2003.
 93. Hawke TJ, Garry DJ. Myogenic satellite cells: Physiology to molecular biology. J Appl Physiol 91: 534‐551, 2001.
 94. Hayes A, Williams DA. Beneficial effects of voluntary wheel running on the properties of dystrophic mouse muscle. J Appl Physiol 80: 670‐679, 1996.
 95. Heinrich R, Gunderoth‐Palmowski M, Grauer W, Machac N, Dette S, Egberts EH. [Tissue pO2 in the anterior tibial muscle of healthy probands in normovolemic hemodilution with 10% HES 200]. VASA Zeitschrift fur Gefasskrankheiten 16: 318‐323, 1987.
 96. Hesselink MK, Kuipers H, Geurten P, Van Straaten H. Structural muscle damage and muscle strength after incremental number of isometric and forced lengthening contractions. J Muscle Res Cell Motil 17: 335‐341, 1996.
 97. Hinz M, Krappmann D, Eichten A, Heder A, Scheidereit C, Strauss M. NF‐kappaB function in growth control: Regulation of cyclin D1 expression and G0/G1‐to‐S‐phase transition. Mol Cell Biol 19: 2690‐2698, 1999.
 98. Hoffman EP, Brown RH Jr, Kunkel LM. Dystrophin: The protein product of the Duchenne muscular dystrophy locus. Cell 51: 919‐928, 1987.
 99. Horsley V, Jansen KM, Mills ST, Pavlath GK. IL‐4 acts as a myoblast recruitment factor during mammalian muscle growth. Cell 113: 483‐494, 2003.
 100. Huk I, Nanobashvili J, Neumayer C, Punz A, Mueller M, Afkhampour K, Mittlboeck M, Losert U, Polterauer P, Roth E, Patton S, Malinski T. L‐arginine treatment alters the kinetics of nitric oxide and superoxide release and reduces ischemia/reperfusion injury in skeletal muscle. Circ 96: 667‐675, 1997.
 101. Ishimoto S, Goto I, Ohta M, Kuroiwa Y. A quantitative study of the muscle satellite cells in various neuromuscular disorders. J Neurol Sci 62: 303‐314, 1983.
 102. Itoh N, Mima T, Mikawa T. Loss of fibroblast growth factor receptors is necessary for terminal differentiation of embryonic limb muscle. Develop 122: 291‐300, 1996.
 103. Jin P, Sejersen T, Ringertz NR. Recombinant platelet‐derived growth factor‐BB stimulates growth and inhibits differentiation of rat L6 myoblasts. J Biol Chem 266: 1245‐1249, 1991.
 104. Jones DA, Newham DJ, Round JM, Tolfree SE. Experimental human muscle damage: Morphological changes in relation to other indices of damage. J Physiol 375: 435‐448, 1986.
 105. Kajita T, Hugli TE. C5a‐induced neutrophilia. A primary humoral mechanism for recruitment of neutrophils. Am J Pathol 137: 467‐477, 1990.
 106. Kasper CE. Sarcolemmal disruption in reloaded atrophic skeletal muscle. J Appl Physiol 79: 607‐614, 1995.
 107. Kawasaki S, Sugiyama S, Ishiguro N, Ozawa T, Miura T. Implication of superoxide radicals on ischemia‐reperfusion‐induced skeletal muscle injury in rats. Eur Surg Res 25: 129‐136, 1993.
 108. Kelic S, Olsson T, Kristensson K. Interferon‐gamma promotes proliferation of rat skeletal muscle cells in vitro and alters their AChR distribution. J Neurol Sci 114: 62‐67, 1993.
 109. Kherif S, Lafuma C, Dehaupas M, Lachkar S, Fournier JG, Verdiere‐Sahuque M, Fardeau M, Alameddine HS. Expression of matrix metalloproteinases 2 and 9 in regenerating skeletal muscle: A study in experimentally injured and mdx muscles. Develop Biol 205: 158‐170, 1999.
 110. Kissel JT, Halterman RK, Rammohan KW, Mendell JR. The relationship of complement‐mediated microvasculopathy to the histologic features and clinical duration of disease in dermatomyositis. Arch Neurol 48: 26‐30, 1991.
 111. Klein‐Ogus C, Harris JB. Preliminary observations of satellite cells in undamaged fibres of the rat soleus muscle assaulted by a snake‐venom toxin. Cell Tiss Res 230: 671‐676, 1983.
 112. Knight KR, Zhang B, Morrison WA, Stewart AG. Ischaemia‐reperfusion injury in mouse skeletal muscle is reduced by N omega‐nitro‐L‐arginine methyl ester and dexamethasone. Eur J Pharmacol 332: 273‐278, 1997.
 113. Kobzik L, Reid MB, Bredt DS, Stamler JS. Nitric oxide in skeletal muscle. Nature 372: 546‐548, 1994.
 114. Komulainen J, Kytola J, Vihko V. Running‐induced muscle injury and myocellular enzyme release in rats. J Appl Physiol 77: 2299‐2304, 1994.
 115. Korthuis RJ, Grisham MB, Granger DN. Leukocyte depletion attenuates vascular injury in postischemic skeletal muscle. Am J Physiol 254: H823‐827, 1988.
 116. Krippendorf BB, Riley DA. Distinguishing unloading‐ versus reloading‐induced changes in rat soleus muscle. Muscle Nerve 16: 99‐108, 1993.
 117. Kristiansen M, Graversen JH, Jacobsen C, Sonne O, Hoffman HJ, Law SK, Moestrup SK. Identification of the haemoglobin scavenger receptor. Nature 409: 198‐201, 2001.
 118. Kumar A, Bhatnagar S, Kumar A. Matrix metalloproteinase inhibitor batimastat alleviates pathology and improves skeletal muscle function in dystrophin‐deficient mdx mice. Am J Physiol 177: 248‐260, 2010.
 119. Kunze K. Spontaneous oscillations of PO2 in muscle tissue. Advan Exper Med Biol 75: 631‐637, 1976.
 120. Kyriakides C, Austen W Jr, Wang Y, Favuzza J, Kobzik L, Moore FD Jr, Hechtman HB. Skeletal muscle reperfusion injury is mediated by neutrophils and the complement membrane attack complex. Am J Physiol 277: C1263‐1268, 1999.
 121. Langen RC, Schols AM, Kelders MC, Van Der Velden JL, Wouters EF, Janssen‐Heininger YM. Muscle wasting and impaired muscle regeneration in a murine model of chronic pulmonary inflammation. Am J Resp Cell Mol Biol 35: 689‐696, 2006.
 122. Langen RC, Schols AM, Kelders MC, Wouters EF, Janssen‐Heininger YM. Inflammatory cytokines inhibit myogenic differentiation through activation of nuclear factor‐kappaB. FASEB J 15: 1169‐1180, 2001.
 123. Langen RC, Van Der Velden JL, Schols AM, Kelders MC, Wouters EF, Janssen‐Heininger YM. Tumor necrosis factor‐alpha inhibits myogenic differentiation through MyoD protein destabilization. FASEB J 18: 227‐237, 2004.
 124. Launay T, Armand AS, Charbonnier F, Mira JC, Donsez E, Gallien CL, Chanoine C. Expression and neural control of myogenic regulatory factor genes during regeneration of mouse soleus. J Histochem Cytochem 49: 887‐899, 2001.
 125. Lauritzen F, Paulsen G, Raastad T, Bergersen LH, Owe SG. Gross ultrastructural changes and necrotic fiber segments in elbow flexor muscles after maximal voluntary eccentric action in humans. J Appl Physiol 107: 1923‐1934, 2009.
 126. Law DJ, Allen DL, Tidball JG. Talin, vinculin and DRP (utrophin) concentrations are increased at mdx myotendinous junctions following onset of necrosis. J Cell Sci 107: 1477‐1483, 1994.
 127. Lazarus B, Messina A, Barker JE, Hurley JV, Romeo R, Morrison WA, Knight KR. The role of mast cells in ischaemia‐reperfusion injury in murine skeletal muscle. J Path 191: 443‐448, 2000.
 128. Le Grand F, Rudnicki MA. Skeletal muscle satellite cells and adult myogenesis. Curr Opin Cell Biol 19: 628‐633, 2007.
 129. Lee KK, Wong CC, Webb SE, Tang MK, Leung AK, Kwok PF, Cai DQ, Chan KM. Hepatocyte growth factor stimulates chemotactic response in mouse embryonic limb myogenic cells in vitro. J Exper Zool 283: 170‐180, 1999.
 130. Lennon NJ, Kho A, Bacskai BJ, Perlmutter SL, Hyman BT, Brown RH Jr. Dysferlin interacts with annexins A1 and A2 and mediates sarcolemmal wound‐healing. J Biol Chem 278: 50466‐50473, 2003.
 131. Leslie CC. Properties and regulation of cytosolic phospholipase A2. J Biol Chem 272: 16709‐16712, 1997.
 132. Li G, Feng X, Wang S. Effects of Cu/Zn superoxide dismutase on strain injury‐induced oxidative damage to skeletal muscle in rats. Physiol Res. 54: 193‐199, 2005.
 133. Li H, Mittal A, Makonchuk DY, Bhatnagar S, Kumar A. Matrix metalloproteinase‐9 inhibition ameliorates pathogenesis and improves skeletal muscle regeneration in muscular dystrophy. Human Molec Genetics 18: 2584‐2598, 2009.
 134. Li YP. TNF‐alpha is a mitogen in skeletal muscle. Am J Physiol 285: C370‐C376, 2003.
 135. Lieber RL, Friden J. Muscle damage is not a function of muscle force but active muscle strain. J Appl Physiol 74: 520‐526, 1993.
 136. Lieber RL, Leonard ME, Brown CG, Trestik CL. Frog semitendinosis tendon load‐strain and stress‐strain properties during passive loading. Am J Physiol 261: C86‐92, 1991.
 137. Lieber RL, Thornell LE, Friden J. Muscle cytoskeletal disruption occurs within the first 15 min of cyclic eccentric contraction. J Appl Physiol 80: 278‐284, 1996.
 138. Liu J, Aoki M, Illa I, Wu C, Fardeau M, Angelini C, Serrano C, Urtizberea JA, Hentati F, Hamida MB, Bohlega S, Culper EJ, Amato AA, Bossie K, Oeltjen J, Bejaoui K, McKenna‐Yasek D, Hosler BA, Schurr E, Arahata K, de Jong PJ, Brown RH Jr. Dysferlin, a novel skeletal muscle gene, is mutated in Miyoshi myopathy and limb girdle muscular dystrophy. Nature Genetics 20: 31‐36, 1998.
 139. Lluís F, Perdiguero E, Nebreda AR, Muñoz‐Cánoves P. Regulation of skeletal muscle gene expression by p38 MAP kinases. Trends Cell Biol. 16: 36‐44, 2006.
 140. Lolmede K, Campana L, Vezzoli M, Bosurgi L, Tonlorenzi R, Clementi E, Bianchi ME, Cossu G, Manfredi AA, Brunelli S, Rovere‐Querini P. Inflammatory and alternatively activated human macrophages attract vessel‐associated stem cells, relying on separate HMGB1‐ and MMP‐9‐dependent pathways. J Leukocyte Biol 85: 779‐787, 2009.
 141. Lowe DA, Warren GL, Hayes DA, Farmer MA, Armstrong RB. Eccentric contraction‐induced injury of mouse soleus muscle: Effect of varying [Ca2+]o. J Appl Physiol 76: 1445‐1453, 1994.
 142. Lowe DA, Warren GL, Ingalls CP, Boorstein DB, Armstrong RB. Muscle function and protein metabolism after initiation of eccentric contraction‐induced injury. J Appl Physiol 79: 1260‐1270, 1995.
 143. Lynch GS, Faulkner JA. Contraction‐induced injury to single muscle fibers: Velocity of stretch does not influence the force deficit. Am J Physiol 275: C1548‐C1554, 1998.
 144. Ma XL, Tsao PS, Lefer AM. Antibody to CD‐18 exerts endothelial and cardiac protective effects in myocardial ischemia and reperfusion. J Clin Invest 88(4): 1237‐1243, 1991.
 145. Macpherson PC, Schork MA, Faulkner JA. Contraction‐induced injury to single fiber segments from fast and slow muscles of rats by single stretches. Am J Physiol 271: C1438‐C1446, 1996.
 146. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 25: 677‐686, 2004.
 147. Matsuda R, Nishikawa A, Tanaka H. Visualization of dystrophic muscle fibers in mdx mouse by vital staining with Evans blue: Evidence of apoptosis in dystrophin‐deficient muscle. J Biochem 118: 959‐964, 1995.
 148. Mauro A. Satellite cell of skeletal muscle fibers. J Biophys Biochem Cytol 9: 493‐495, 1961.
 149. McBride JM, Kraemer WJ, Triplett‐McBride T, Sebastianelli W. Effect of resistance exercise on free radical production. Med Sci Sports Exer 30: 67‐72, 1998.
 150. McCully KK, Faulkner JA. Injury to skeletal muscle fibers of mice following lengthening contractions. J Appl Physiol 59: 119‐126, 1985.
 151. McCully KK, Faulkner JA. Characteristics of lengthening contractions associated with injury to skeletal muscle fibers. J Appl Physiol 61: 293‐299, 1986.
 152. McLoon LK, Nguyen LT, Wirtschafter J. Time course of the regenerative response in bupivacaine injured orbicularis oculi muscle. Cell Tissue Res 294: 439‐447, 1998.
 153. Meldrum DG, Stephenson LL, Zamboni WA. Effects of L‐NAME and L‐arginine on ischemia‐reperfusion injury in rat skeletal muscle. Plastic Reconstruct Surg 103: 935‐940, 1999.
 154. Mentzer SJ, Herrmann SH, Crimmins MA, Burakoff SJ, Faller DV. The role of CD18 in phorbol ester‐induced human monocyte‐mediated cytotoxicity. Cellular Immunol 115: 66‐77, 1988.
 155. Messina A, Knight KR, Dowsing BJ, Zhang B, Phan LH, Hurley JV, Morrison WA, Stewart AG. Localization of inducible nitric oxide synthase to mast cells during ischemia/reperfusion injury of skeletal muscle. Laboratory Invest 80: 423‐431, 2000.
 156. Meuer S, Ecker U, Hadding U, Bitter‐Suermann D. Platelet‐serotonin release by C3a and C5a: Two independent pathways of activation. J Immunol 126: 1506‐1509, 1981.
 157. Miura P, Chakkalakal JV, Boudreault L, Belanger G, Hebert RL, Renaud JM, Jasmin BJ. Pharmacological activation of PPARbeta/delta stimulates utrophin A expression in skeletal muscle fibers and restores sarcolemmal integrity in mature mdx mice. Human Molec Genetics 18: 4640‐4649, 2009.
 158. Molkentin JD, Black BL, Martin JF, Olson EN. Cooperative activation of muscle gene expression by MEF2 and myogenic bHLH proteins. Cell 83: 1125‐1136, 1995.
 159. Morgan JE, Coulton GR, Partridge TA. Muscle precursor cells invade and repopulate freeze‐killed muscles. J Muscle Res Cell Motil 8: 386‐396, 1987.
 160. Morrison J, Palmer DB, Cobbold S, Partridge T, Bou‐Gharios G. Effects of T‐lymphocyte depletion on muscle fibrosis in the mdx mouse. Am J Pathol 166: 1701‐1710, 2005.
 161. Mourkioti F, Rosenthal N. NF‐kappaB signaling in skeletal muscle: Prospects for intervention in muscle diseases. J Mol Med 86: 747‐759, 2008.
 162. Muller‐Eberhard HJ. The membrane attack complex of complement. Ann Rev Immunol 4: 503‐528, 1986.
 163. Munder M, Mallo M, Eichmann K, Modolell M. Murine macrophages secrete interferon gamma upon combined stimulation with interleukin (IL)‐12 and IL‐18: A novel pathway of autocrine macrophage activation. J Exper Med 187: 2103‐2108, 1998.
 164. Murachi T, Tanaka K, Hatanaka M, Murakami T. Intracellular Ca2+‐dependent protease (calpain) and its high‐molecular‐weight endogenous inhibitor (calpastatin). Adv Enzyme Regul. 19: 407‐24, 1980.
 165. Nakane M, Schmidt HH, Pollock JS, Forstermann U, Murad F. Cloned human brain nitric oxide synthase is highly expressed in skeletal muscle. FEBS Lett 316: 175‐180, 1993.
 166. Naldini L, Weidner KM, Vigna E, Gaudino G, Bardelli A, Ponzetto C, Narsimhan RP, Hartmann G, Zarnegar R, Michalopoulos GK, Birchmeier W, Comoglio PM. Scatter factor and hepatocyte growth factor are indistinguishable ligands for the MET receptor. EMBO J 10: 2867‐2878, 1991.
 167. Nelson WJ, Traub P. Proteolysis of vimentin and desmin by the Ca2+‐activated proteinase specific for the intermediate filament proteins. Mol Cell Biol. 3(6): 1146‐56, 1983.
 168. Nethery D, Stofan D, Callahan L, DiMarco A, Supinski G. Formation of reactive oxygen species by the contracting diaphragm is PLA(2) dependent. J Appl Physiol 87: 792‐800, 1999.
 169. Newham DJ, McPhail G, Mills KR, Edwards RH. Ultrastructural changes after concentric and eccentric contractions of human muscle. J Neurol Sci 61: 109‐122, 1983.
 170. Nguyen HX, Lusis AJ, Tidball JG. Null mutation of myeloperoxidase in mice prevents mechanical activation of neutrophil lysis of muscle cell membranes in vitro and in vivo. J Physiol 565: 403‐413, 2005.
 171. Nguyen HX, Tidball JG. Interactions between neutrophils and macrophages promote macrophage killing of rat muscle cells in vitro. J Physiol 547: 125‐132, 2003.
 172. Nguyen HX, Tidball JG. Null mutation of gp91phox reduces muscle membrane lysis during muscle inflammation in mice. J Physiol 553: 833‐841, 2003.
 173. Nguyen TM, Ellis JM, Love DR, Davies KE, Gatter KC, Dickson G, Morris GE. Localization of the DMDL gene‐encoded dystrophin‐related protein using a panel of nineteen monoclonal antibodies: Presence at neuromuscular junctions, in the sarcolemma of dystrophic skeletal muscle, in vascular and other smooth muscles, and in proliferating brain cell lines. J Cell Biol 115: 1695‐1700, 1991.
 174. Nikolaou PK, MacDonald BL, Glisson RR, Seaber AV, Garrett WE Jr. Biomechanical and hisological evaluation of muscle after controlled strain injury. Am J Sports Med 15, 9‐14, 1987.
 175. Ochoa O, Sun D, Reyes‐Reyna SM, Waite LL, Michalek JE, McManus LM, Shireman PK. Delayed angiogenesis and VEGF production in CCR2‐/‐ mice during impaired skeletal muscle regeneration. Am J Physiol 293: R651‐661, 2007.
 176. Ohlendieck K, Campbell KP. Dystrophin‐associated proteins are greatly reduced in skeletal muscle from mdx mice. J Cell Biol 115: 1685‐1694, 1991.
 177. Ohlendieck K, Ervasti JM, Snook JB, Campbell KP. Dystrophin‐glycoprotein complex is highly enriched in isolated skeletal muscle sarcolemma. J Cell Biol 112: 135‐148, 1991.
 178. Olwin BB, Hauschka SD. Cell surface fibroblast growth factor and epidermal growth factor receptors are permanently lost during skeletal muscle terminal differentiation in culture. J Cell Biol 107: 761‐769, 1988.
 179. Ottnad E, Parthasarathy S, Sambrano GR, Ramprasad MP, Quehenberger O, Kondratenko N, Green S, Steinberg D. A macrophage receptor for oxidized low density lipoprotein distinct from the receptor for acetyl low density lipoprotein: Partial purification and role in recognition of oxidatively damaged cells. Proc Nat Acad Sci USA 92: 1391‐1395, 1995.
 180. Ozaki M, Kawashima S, Hirase T, Yamashita T, Namiki M, Inoue N, Hirata Ki K, Yokoyama M. Overexpression of endothelial nitric oxide synthase in endothelial cells is protective against ischemia‐reperfusion injury in mouse skeletal muscle. Am J Pathol 160: 1335‐1344, 2002.
 181. Passerini L, Bernasconi P, Baggi F, Confalonieri P, Cozzi F, Cornelio F, Mantegazza R. Fibrogenic cytokines and extent of fibrosis in muscle of dogs with X‐linked golden retriever muscular dystrophy. Neuromuscul Disord 12: 828‐835, 2002.
 182. Pastoret C, Sebille A. mdx mice show progressive weakness and muscle deterioration with age. J Neurol Sci 129: 97‐105, 1995.
 183. Pattwell D, McArdle A, Griffiths RD, Jackson MJ. Measurement of free radical production by in vivo microdialysis during ischemia/reperfusion injury to skeletal muscle. Free Radic Biol Med 30: 979‐985, 2001.
 184. Pattwell DM, McArdle A, Morgan JE, Partridge TA, Jackson MJ. Release of reactive oxygen and nitrogen species from contracting skeletal muscle cells. Free Radic Biol Med 37: 1064‐1072, 2004.
 185. Pavlath GK, Dominov JA, Kegley KM, Miller JB. Regeneration of transgenic skeletal muscles with altered timing of expression of the basic helix‐loop‐helix muscle regulatory factor MRF4. Am J Pathol 162: 1685‐1691, 2003.
 186. Pemberton M, Anderson G, Vetvicka V, Justus DE, Ross GD. Microvascular effects of complement blockade with soluble recombinant CR1 on ischemia/reperfusion injury of skeletal muscle. J Immunol 150: 5104‐5113, 1993.
 187. Petrasek PF, Homer‐Vanniasinkam S, Walker PM. Determinants of ischemic injury to skeletal muscle. J Vasc Surg 19: 623‐631, 1994.
 188. Petrasek PF, Liauw S, Romaschin AD, Walker PM. Salvage of postischemic skeletal muscle by monoclonal antibody blockade of neutrophil adhesion molecule CD18. J Surg Res 56: 5‐12, 1994.
 189. Petrof BJ, Shrager JB, Stedman HH, Kelly AM, Sweeney HL. Dystrophin protects the sarcolemma from stresses developed during muscle contraction. Proc Nat Acad Sci USA 90: 3710‐3714, 1993.
 190. Pizza FX, Peterson JM, Baas JH, Koh TJ. Neutrophils contribute to muscle injury and impair its resolution after lengthening contractions in mice. J Physiol 562: 899‐913, 2005.
 191. Price M, Sanger JW. Intermediate filaments connect z‐discs in adult chicken muscle. J Exper Zool 208: 263‐269, 1979.
 192. Price MG, Sanger JW. Intermediate filaments in striated muscle. A review of structural studies in embryonic and adult skeletal and cardiac muscle. Cell Muscle Motil 3: 1‐40, 1983.
 193. Rader EP, Song W, Van Remmen H, Richardson A, Faulkner JA. Raising the antioxidant levels within mouse muscle fibres does not affect contraction‐induced injury. Exper Physiol 91: 781‐789, 2006.
 194. Rafael JA, Tinsley JM, Potter AC, Deconinck AE, Davies KE. Skeletal muscle‐specific expression of a utrophin transgene rescues utrophin‐dystrophin deficient mice. Nature Genetics 19: 79‐82, 1998.
 195. Raingeaud J, Gupta S, Rogers JS, Dickens M, Han J, Ulevitch RJ, Davis RJ. Pro‐inflammatory cytokines and environmental stress cause p38 mitogen‐activated protein kinase activation by dual phosphorylation on tyrosine and threonine. J Biol Chem 270: 7420‐7426, 1995.
 196. Ramprasad MP, Fischer W, Witztum JL, Sambrano GR, Quehenberger O, Steinberg D. The 94‐ to 97‐kDa mouse macrophage membrane protein that recognizes oxidized low density lipoprotein and phosphatidylserine‐rich liposomes is identical to macrosialin, the mouse homologue of human CD68. Proc Nat Acad Sci USA 92: 9580‐9584, 1995.
 197. Ramsey RW, Street SF. The isometric length‐tension diagram of isolated skeletal muscle fibers of the frog. J. Cell Comp Physiol 15: 11‐34, 1940.
 198. Ratajczak MZ, Majka M, Kucia M, Drukala J, Pietrzkowski Z, Peiper S, Janowska‐Wieczorek A. Expression of functional CXCR4 by muscle satellite cells and secretion of SDF‐1 by muscle‐derived fibroblasts is associated with the presence of both muscle progenitors in bone marrow and hematopoietic stem/progenitor cells in muscles. Stem Cells 21: 363‐371, 2003.
 199. Raynal P, Pollard HB. Annexins: The problem of assessing the biological role for a gene family of multifunctional calcium‐ and phospholipid‐binding proteins. Biochim Biophy Acta 1197: 63‐93, 1994.
 200. Reddy MK, Rabinowitz M, Zak R. Stringent requirement for Ca2+ in the removal of Z‐lines and alpha‐actinin from isolated myofibrils by Ca2+ = activated neutral proteinase. Biochem J 209(3): 635‐41, 1983.
 201. Reid MB, Shoji T, Moody MR, Entman ML. Reactive oxygen in skeletal muscle. II. Extracellular release of free radicals. J Appl Physiol 73: 1805‐1809, 1992.
 202. Reid RR, Woodcock S, Shimabukuro‐Vornhagen A, Austen WG Jr, Kobzik L, Zhang M, Hechtman HB, Moore FD Jr, Carroll MC. Functional activity of natural antibody is altered in Cr2‐deficient mice. J Immunol 169: 5433‐5440, 2002.
 203. Renault V, Piron‐Hamelin G, Forestier C, DiDonna S, Decary S, Hentati F, Saillant G, Butler‐Browne GS, Mouly V. Skeletal muscle regeneration and the mitotic clock. Exper Gerontol 35: 711‐719, 2000.
 204. Riley DA, Ellis S, Giometti CS, Hoh JF, Ilyina‐Kakueva EI, Oganov VS, Slocum GR, Bain JL, Sedlak FR. Muscle sarcomere lesions and thrombosis after spaceflight and suspension unloading. J Appl Physiol 73: 33S‐43S, 1992.
 205. Robertson TA, Maley MA, Grounds MD, Papadimitriou JM. The role of macrophages in skeletal muscle regeneration with particular reference to chemotaxis. Exper Cell Res 207: 321‐331, 1993.
 206. Roche JA, Lovering RM, Bloch RJ. Impaired recovery of dysferlin‐null skeletal muscle after contraction‐induced injury in vivo. Neuroreport 19: 1579‐1584, 2008.
 207. Rubin BB, Smith A, Liauw S, Isenman D, Romaschin AD, Walker PM. Complement activation and white cell sequestration in postischemic skeletal muscle. Am J Physiol 259: H525‐531, 1990.
 208. Sam M, Shah S, Fridén J, Milner DJ, Capetanaki Y, Lieber RL. Desmin knockout muscles generate lower stress and are less vulnerable to injury compared with wild‐type muscles. Amer J Physiol 279: C1116‐1122, 2000.
 209. Schaer DJ, Boretti FS, Hongegger A, Poehler D, Linnscheid P, Staege H, Muller C, Schoedon G, Schaffner A. Molecular cloning and characterization of the mouse CD163 homologue, a highly glucocorticoid‐inducible member of the scavenger receptor cysteine‐rich family. Immunogenet 53: 170‐177, 2001.
 210. Schafer H, Mathey D, Hugo F, Bhakdi S. Deposition of the terminal C5b‐9 complement complex in infarcted areas of human myocardium. J Immunol 137: 1945‐1949, 1986.
 211. Schultz E, Albright DJ, Jaryszak DL, David TL. Survival of satellite cells in whole muscle transplants. Anat Rec 222: 12‐17, 1988.
 212. Schultz E, Jaryszak DL, Valliere CR. Response of satellite cells to focal skeletal muscle injury. Muscle Nerve 8: 217‐222, 1985.
 213. Schultz E, Lipton BH. Skeletal muscle satellite cells: Changes in proliferation potential as a function of age. Mech Age Develop 20: 377‐383, 1982.
 214. Seed J, Olwin BB, Hauschka SD. Fibroblast growth factor levels in the whole embryo and limb bud during chick development. Develop Biol 128: 50‐57, 1988.
 215. Seekamp A, Till GO, Mulligan MS, Paulson JC, Anderson DC, Miyasaka M, Ward PA. Role of selectins in local and remote tissue injury following ischemia and reperfusion. Am J Pathol 144: 592‐598, 1994.
 216. Selcen D, Stilling G, Engel AG. The earliest pathologic alterations in dysferlinopathy. Neurol 56: 1472‐1481, 2001.
 217. Shireman PK, Contreras‐Shannon V, Ochoa O, Karia BP, Michalek JE, McManus LM. MCP‐1 deficiency causes altered inflammation with impaired skeletal muscle regeneration. J Leukocyte Biol 81: 775‐785, 2007.
 218. Siegel AL, Atchison K, Fisher KE, Davis GE, Cornelison DD. 3D timelapse analysis of muscle satellite cell motility. Stem Cells 27: 2527‐2538, 2009.
 219. Soltow QA, Lira VA, Betters JL, Long JH, Sellman JE, Zeanah EH, Criswell DS. Nitric oxide regulates stretch‐induced proliferation in C2C12 myoblasts. J Muscle Res Cell Motil 31: 215‐225, 2010.
 220. Spuler S, Engel AG. Unexpected sarcolemmal complement membrane attack complex deposits on nonnecrotic muscle fibers in muscular dystrophies. Neurol 50: 41‐46, 1998.
 221. Stedman HH, Sweeney HL, Shrager JB, Maguire HC, Panettieri RA, Petrof B, Narusawa M, Leferovich JM, Sladky JT, Kelly AM. The mdx mouse diaphragm reproduces the degenerative changes of Duchenne muscular dystrophy. Nature 352: 536‐539, 1991.
 222. Steegmaier M, Blanks JE, Borges E, Vestweber D. P‐selectin glycoprotein ligand‐1 mediates rolling of mouse bone marrow‐derived mast cells on P‐selectin but not efficiently on E‐selectin. Eur J Immunol 27: 1339‐1345, 1997.
 223. Stockdale FE. Myogenic cell lineages. Develop Biol 154: 284‐298, 1992.
 224. Stoker M, Gherardi E, Perryman M, Gray J. Scatter factor is a fibroblast‐derived modulator of epithelial cell mobility. Nature 327: 239‐242, 1987.
 225. Street SF, Ramsey RW. Sarcolemma: Transmitter of active tension in frog skeletal muscle. Science 149: 1379‐1380, 1965.
 226. St Pierre BA, Tidball JG. Differential response of macrophage subpopulations to soleus muscle reloading after rat hindlimb suspension. J Appl Physiol 77: 290‐297, 1994.
 227. St Pierre Schneider B, Brickson S, Corr DT, Best T. CD11b+ neutrophils predominate over RAM11 +macrophages in stretch‐injured muscle. Muscle Nerve 25: 837‐844, 2002.
 228. Suelves M, Lluis F, Ruiz V, Nebreda AR, Munoz‐Canoves P. Phosphorylation of MRF4 transactivation domain by p38 mediates repression of specific myogenic genes. EMBO J 23: 365‐375, 2004.
 229. Summan M, Warren GL, Mercer RR, Chapman R, Hulderman T, Van Rooijen N, Simeonova PP. Macrophages and skeletal muscle regeneration: A clodronate‐containing liposome depletion study. Am J Physiol 290: R1488‐1495, 2006.
 230. Sun D, Martinez CO, Ochoa O, Ruiz‐Willhite L, Bonilla JR, Centonze VE, Waite LL, Michalek JE, McManus LM, Shireman PK. Bone marrow‐derived cell regulation of skeletal muscle regeneration. FASEB J 23: 382‐395, 2009.
 231. Suzuki K, Sato H, Kikuchi T, Abe T, Nakaji S, Sugawara K, Totsuka M, Sato K, Yamaya K. Capacity of circulating neutrophils to produce reactive oxygen species after exhaustive exercise. J Appl Physiol 81: 1213‐1222, 1996.
 232. Takayama H, La Rochelle WJ, Anver M, Bockman DE, Merlino G. Scatter factor/hepatocyte growth factor as a regulator of skeletal muscle and neural crest development. Proc Nat Acad Sci USA 93: 5866‐5871, 1996.
 233. Tatsumi R, Anderson JE, Nevoret CJ, Halevy O, Allen RE. HGF/SF is present in normal adult skeletal muscle and is capable of activating satellite cells. Develop Biol 194: 114‐128, 1998.
 234. Tatsumi R, Hattori A, Ikeuchi Y, Anderson JE, Allen RE. Release of hepatocyte growth factor from mechanically stretched skeletal muscle satellite cells and role of pH and nitric oxide. Molec Biol Cell 13: 2909‐2918, 2002.
 235. Tatsumi R, Liu X, Pulido A, Morales M, Sakata T, Dial S, Hattori A, Ikeuchi Y, Allen RE. Satellite cell activation in stretched skeletal muscle and the role of nitric oxide and hepatocyte growth factor. Am J Physiol 290: C1487‐1494, 2006.
 236. Taylor ML, Brummet ME, Hudson SA, Miura K, Bochner BS. Expression and function of P‐selectin glycoprotein ligand 1 (CD162) on human basophils. J Allergy Clinical Immunol 106: 918‐924, 2000.
 237. Teixeira CF, Zamuner SR, Zuliani JP, Fernandes CM, Cruz‐Hofling MA, Fernandes I, Chaves F, Gutierrez JM. Neutrophils do not contribute to local tissue damage, but play a key role in skeletal muscle regeneration, in mice injected with Bothrops asper snake venom. Muscle Nerve 28: 449‐459, 2003.
 238. Tidball JG. Myotendinous junctions: Morphological changes and mechanical failure associated with muscle cell atrophy. Exp Molec Pathol 40: 1‐12, 1984.
 239. Tidball JG. Desmin at myotendinous junctions. Exper Cell Res 199: 206‐212, 1992.
 240. Tidball JG, Berchenko E, Frenette J. Macrophage invasion does not contribute to muscle membrane injury during inflammation. J Leukocyte Biol 65: 492‐498, 1999.
 241. Tidball JG, Lavergne E, Lau KS, Spencer MJ, Stull JT, Wehling M. Mechanical loading regulates NOS expression and activity in developing and adult skeletal muscle. Am J Physiol 275: C260‐266, 1998.
 242. Tidball JG, Salem G, Zernicke R. Site and mechanical conditions for failure of skeletal muscle in experimental strain injuries. J Appl Physiol 74: 1280‐1286, 1993.
 243. Tidball JG, St Pierre BA. Apoptosis of macrophages during the resulution of muscle inflammation. J Leukocyte Biol 59: 380‐388, 1996.
 244. Tidball JG, Villalta SA. Regulatory interactions between muscle and the immune system during muscle regeneration. Am J Physiol 298: R1173‐1187, 2010.
 245. 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 578: 327‐336, 2007.
 246. Tinsley JM, Potter AC, Phelps SR, Fisher R, Trickett JI, Davies KE. Amelioration of the dystrophic phenotype of mdx mice using a truncated utrophin transgene. Nature 384: 349‐353, 1996.
 247. Tokuyasu KT, Dutton AH, Singer SJ. Immunoelectron microscopic studies of desmin (skeletin) localization and intermediate filament organization in chicken skeletal muscle. J Cell Biol 96: 1727‐1735, 1983.
 248. Tolosa L, Morla M, Iglesias A, Busquets X, Llado J, Olmos G. IFN‐gamma prevents TNF‐alpha‐induced apoptosis in C2C12 myotubes through down‐regulation of TNF‐R2 and increased NF‐kappaB activity. Cell Signal 17: 1333‐1342, 2005.
 249. Torrente Y, El Fahime E, Caron NJ, Del Bo R, Belicchi M, Pisati F, Tremblay JP, Bresolin N. Tumor necrosis factor‐alpha (TNF‐alpha) stimulates chemotactic response in mouse myogenic cells. Cell Transplant 12: 91‐100, 2003.
 250. Trezzini C, Schuepp B, Maly FE, Jungi TW. Evidence that exposure to fibrinogen or to antibodies directed against Mac‐1 (CD11b/CD18; CR3) modulates human monocyte effector functions. Brit J Haematol 77: 16‐24, 1991.
 251. Tsujihata M, Yoshimura T, Satoh A, Kinoshita I, Matsuo H, Mori M, Nagataki S. Diagnostic significance of IgG, C3, and C9 at the limb muscle motor end‐plate in minimal myasthenia gravis. Neurology 39: 1359‐1363, 1989.
 252. Tsujimura T, Koshimizu U, Katoh H, Isozaki K, Kanakura Y, Tono T, Adachi S, Kasugai T, Tei H, Nishimune Y, Nomura S, Kitamura Y. Mast cell number in the skin of heterozygotes reflects the molecular nature of c‐kit mutation. Blood 81: 2530‐2538, 1993.
 253. Vandenburgh H, Kaufman S. In vitro model for stretch‐induced hypertrophy of skeletal muscle. Science 203: 265‐268, 1979.
 254. VanSaun M, Werle MJ. Matrix metalloproteinase‐3 removes agrin from synaptic basal lamina. J Neurobiol 44: 369, 2000.
 255. Van Velzen AG, Da Silva RP, Gordon S, Van Berkel TJ. Characterization of a receptor for oxidized low‐density lipoproteins on rat Kupffer cells: Similarity to macrosialin. Biochem J 322(Pt 2): 411‐415, 1997.
 256. Venkatasubramanian K, Solursh M. Chemotactic behavior of myoblasts. Develop Biol 104: 428‐433, 1984.
 257. Vetrone SA, Montecino‐Rodriguez E, Kudryashova E, Kramerova I, Hoffman EP, Liu SD, Miceli MC, Spencer MJ. Osteopontin promotes fibrosis in dystrophic mouse muscle by modulating immune cell subsets and intramuscular TGF‐beta. J Clin Invest 119: 1583‐1594, 2009.
 258. Villalta SA, Nguyen HX, Deng B, Gotoh T, Tidball JG. Shifts in macrophage phenotypes and macrophage competition for arginine metabolism affect the severity of muscle pathology in muscular dystrophy. Human Molec Genetics 18: 482‐496, 2009.
 259. Villalta SA, Rinaldi C, Deng B, Liu G, Fedor B, Tidball JG. Interleukin‐10 reduces the pathology of mdx muscular dystrophy by deactivating M1 macrophages and modulating macrophage phenotype. Human Molec Genetics 20: 790‐805, 2011.
 260. Wang H, Hertlein E, Bakkar N, Sun H, Acharyya S, Wang J, Carathers M, Davuluri R, Guttridge DC. NF‐kappaB regulation of YY1 inhibits skeletal myogenesis through transcriptional silencing of myofibrillar genes. Molec Cell Biol 27: 4374‐4387, 2007.
 261. Wang K. Sarcomere‐associated cytoskeletal lattices in striated muscle. Review and hypothesis. Cell Muscle Motil 6: 315‐369, 1985.
 262. Warren GL, Hayes DA, Lowe DA, Armstrong RB. Mechanical factors in the initiation of eccentric contraction‐induced injury in rat soleus muscle. J Physiol 464: 457‐475, 1993.
 263. Warren GL, Hulderman T, Jensen N, McKinstry M, Mishra M, Luster MI, Simeonova PP. Physiological role of tumor necrosis factor alpha in traumatic muscle injury. FASEB J 16: 1630‐1632, 2002.
 264. Warren GL, Hulderman T, Mishra D, Gao X, Millecchia L, O'Farrell L, Kuziel WA, Simeonova PP. Chemokine receptor CCR2 involvement in skeletal muscle regeneration. FASEB J 19: 413‐415, 2005.
 265. Warren GL, Lowe DA, Hayes DA, Farmer MA, Armstrong RB. Redistribution of cell membrane probes following contraction‐induced injury of mouse soleus muscle. Cell Tiss Res 282: 311‐320, 1995.
 266. Warren GL, O'Farrell L, Summan M, Hulderman T, Mishra D, Luster MI, Kuziel WA, Simeonova PP. Role of CC chemokines in skeletal muscle functional restoration after injury. Am J Physiol 286: C1031‐1036, 2004.
 267. Watt DJ, Karasinski J, Moss J, England MA. Migration of muscle cells. Nature 368: 406‐407, 1994.
 268. Webb SE, Lee KK, Tang MK, Ede DA. Fibroblast growth factors 2 and 4 stimulate migration of mouse embryonic limb myogenic cells. Dev Dyn 209: 206‐216, 1997.
 269. Webster C, Blau HM. Accelerated age‐related decline in replicative life‐span of Duchenne muscular dystrophy myoblasts: Implications for cell and gene therapy. Som Cell Molec Genetics 16: 557‐565, 1990.
 270. Wehling M, Spencer MJ, Tidball JG. A nitric oxide synthase transgene ameliorates muscular dystrophy in mdx mice. J Cell Biol 155: 123‐131, 2001.
 271. Wehling‐Henricks M, Jordan MC, Gotoh T, Grody WW, Roos KP, Tidball JG. Arginine metabolism by macrophages promotes cardiac and muscle fibrosis in mdx muscular dystrophy. PLoS One 5: e10763, 2010.
 272. Wehling‐Henricks M, Lee JJ, Tidball JG. Prednisolone decreases cellular adhesion molecules required for inflammatory cell infiltration in dystrophin‐deficient skeletal muscle. Neuromuscul Disord 14: 483‐490, 2004.
 273. Weiser MR, Gibbs SA, Valeri CR, Shepro D, Hechtman HB. Anti‐selectin therapy modifies skeletal muscle ischemia and reperfusion injury. Shock 5: 402‐407, 1996.
 274. Welbourn CR, Goldman G, Paterson IS, Valeri CR, Shepro D, Hechtman HB. Pathophysiology of ischaemia reperfusion injury: Central role of the neutrophil. Br J Surg. 78: 651‐655, 1991.
 275. Whitehead NP, Pham C, Gervasio OL, Allen DG. N‐Acetylcysteine ameliorates skeletal muscle pathophysiology in mdx mice. J Physiol 586: 2003‐2014, 2008.
 276. Witte MB, Barbul A. Arginine physiology and its implication for wound healing. Wound Repair Regen 11: 419‐423, 2003.
 277. Witte MB, Vogt N, Stuelten C, Gotoh T, Mori M, Becker HD. Arginase acts as an alternative pathway of L‐arginine metabolism in experimental colon anastomosis. J Gastrointest Surg 7: 378‐385, 2003.
 278. Wu G, Morris SM Jr. Arginine metabolism: Nitric oxide and beyond. The Biochem J 336: 1‐17, 1998.
 279. Yablonka‐Reuveni Z, Balestreri TM, Bowen‐Pope DF. Regulation of proliferation and differentiation of myoblasts derived from adult mouse skeletal muscle by specific isoforms of PDGF. J Cell Biol 111: 1623‐1629, 1990.
 280. Yablonka‐Reuveni Z, Rivera AJ. Temporal expression of regulatory and structural muscle proteins during myogenesis of satellite cells on isolated adult rat fibers. Develop Biol 164: 588‐603, 1994.
 281. Yahiaoui L, Gvozdic D, Danialou G, Mack M, Petrof BJ. CC family chemokines directly regulate myoblast responses to skeletal muscle injury. J Physiol 586: 3991‐4004, 2008.
 282. Yamada M, Sankoda Y, Tatsumi R, Mizunoya W, Ikeuchi Y, Sunagawa K, Allen RE. Matrix metalloproteinase‐2 mediates stretch‐induced activation of skeletal muscle satellite cells in a nitric oxide‐dependent manner. Intern J Biochem Cell Biol 40: 2183‐2191, 2008.
 283. Yan Z, Choi S, Liu X, Zhang M, Schageman JJ, Lee SY, Hart R, Lin L, Thurmond FA, Williams RS. Highly coordinated gene regulation in mouse skeletal muscle regeneration. J Biol Chem 278: 8826‐8836, 2003.
 284. Yokota J, Minei JP, Fantini GA, Shires GT. Role of leukocytes in reperfusion injury of skeletal muscle after partial ischemia. Am J Physiol 257: H1068‐1075, 1989.
 285. Zerria K, Jerbi E, Hammami S, Maaroufi A, Boubaker S, Xiong JP, Arnaout MA, Fathallah DM. Recombinant integrin CD11b A‐domain blocks polymorphonuclear cells recruitment and protects against skeletal muscle inflammatory injury in the rat. Immunology 119: 431‐440, 2006.
 286. Zetser A, Gredinger E, Bengal E. p38 mitogen‐activated protein kinase pathway promotes skeletal muscle differentiation. Participation of the Mef2c transcription factor. J Biol Chem 274: 5193‐5200, 1999.
 287. Zhang Y, Pilon G, Marette A, Baracos VE. Cytokines and endotoxin induce cytokine receptors in skeletal muscle. Am J Physiol 279: E196‐E205, 2000.
 288. Zouaoui Boudjeltia K, Moguilevsky N, Legssyer I, Babar S, Guillaume M, Delree P, Vanhaeverbeek M, Brohee D, Ducobu J, Remacle C. Oxidation of low density lipoproteins by myeloperoxidase at the surface of endothelial cells: An additional mechanism to subendothelium oxidation. Biochem Biophys Res Comm 325: 434‐438, 2004.

Related Articles:

Alterations in Myofibril Size and Structure During Growth, Exercise, and Changes in Environmental Temperature
Diseases of Skeletal Muscle
Surface and Internal Morphology of Skeletal Muscle
Molecular Mechanisms of Muscle Plasticity with Exercise
Reactive Oxygen Species: Impact on Skeletal Muscle
Immunity and Inflammation

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

James G. Tidball. Mechanisms of Muscle Injury, Repair, and Regeneration. Compr Physiol 2011, 1: 2029-2062. doi: 10.1002/cphy.c100092