Comprehensive Physiology Wiley Online Library

Satellite Cells and Skeletal Muscle Regeneration

Full Article on Wiley Online Library



ABSTRACT

Skeletal muscles are essential for vital functions such as movement, postural support, breathing, and thermogenesis. Muscle tissue is largely composed of long, postmitotic multinucleated fibers. The life‐long maintenance of muscle tissue is mediated by satellite cells, lying in close proximity to the muscle fibers. Muscle satellite cells are a heterogeneous population with a small subset of muscle stem cells, termed satellite stem cells. Under homeostatic conditions all satellite cells are poised for activation by stimuli such as physical trauma or growth signals. After activation, satellite stem cells undergo symmetric divisions to expand their number or asymmetric divisions to give rise to cohorts of committed satellite cells and thus progenitors. Myogenic progenitors proliferate, and eventually differentiate through fusion with each other or to damaged fibers to reconstitute fiber integrity and function. In the recent years, research has begun to unravel the intrinsic and extrinsic mechanisms controlling satellite cell behavior. Nonetheless, an understanding of the complex cellular and molecular interactions of satellite cells with their dynamic microenvironment remains a major challenge, especially in pathological conditions. The goal of this review is to comprehensively summarize the current knowledge on satellite cell characteristics, functions, and behavior in muscle regeneration and in pathological conditions. © 2015 American Physiological Society. Compr Physiol 5:1027‐1059, 2015.

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. Skeletal muscle anatomy. Schematic illustrating the structure of skeletal muscle. Skeletal muscles are divided into bundles containing numerous myofibers. Myofibers are multinucleated cylindrical cells that are surrounded by a vast network of blood vessels and nerves. Satellite cells are small mononuclear cells located between the plasmalemma of the myofibers and the basal membrane.
Figure 2. Figure 2. Satellite cell markers. Images of single extensor digitorium longus (EDL) myofiber labeled for different markers of satellite cells. The left image shows a multinucleated myofiber with two satellite cells stained by Pax7 antibody (red) and a nuclear dye (DAPI, blue). The right picture displays a satellite cell stained with M‐cadherin (red) on the apical side and α7‐integrin (white) on the basal side.
Figure 3. Figure 3. The satellite cell niche. Cross‐section of a tibialis anterior (TA) muscle immunostained with laminin (white), M‐cadherin (green), Pax7 (red), and a nuclear dye (DAPI, blue). All muscle fibers are surrounded by laminin. The insert in the top right corner shows a magnification of a satellite cell (Pax7‐positive) in its niche.
Figure 4. Figure 4. Embryonic muscle development. The image shows a mouse embryo transgenic for a fluorescent expression reporter (TdTomato) of the myogenic transcription factor Myf5 at 10.5 days after fertilization. At this developmental stage Myf5 is highly expressed in the somites. Somites develop antero‐posteriorly, with the anterior somites being the most mature ones and the posterior somites the least developed.
Figure 5. Figure 5. Genetic programme regulating skeletal muscle stem cell fate. Hierarchical network of the transcription factors controlling embryonic progenitor cell fate during limb and trunk muscle development. Pax3 and Pax7 control the myogenic specification of embryonic progenitors. Pax3/7 can directly target Myf5 and MyoD expression and thus myogenic determination. Myf5 also acts partially upstream of MyoD and can trigger its expression. Upregulation of myogenin and MRF4 induces myogenic differentiation. A population of Pax3/7+ progenitor cells does not express MRFs and gives rise to satellite stem cells during late fetal myogenesis.
Figure 6. Figure 6. Skeletal muscle regeneration. H&E staining (nuclei stained in dark blue and cytosolic proteins stained in red) illustrating the different stages of muscle regeneration following cardiotoxin injury. Two days following injury, myofiber structure is severely impaired and a multitude of mononuclear cells (myoblasts, inflammatory cells, fibroblasts, etc.) are present in the damaged area. At five days postinjury, there is the formation of small new myofibers with centralized nuclei and many cells remain in the exudate. Two months after the injury, muscle architecture is similar to uninjured muscle.
Figure 7. Figure 7. Myogenic lineage progression. Following muscle injury quiescent satellite cells (Pax7+ and Myf5+/−) are activated and differentiate to myoblasts (Pax7+, Myf5+, and MyoD+). After several rounds of proliferation, myoblasts exit the cell cycle and become myocytes (Pax7−, MyoD+, myogenin+, and MRF4+). Myocytes can undergo a fusion process to form multinucleated myotubes (myogenin+, MRF4+, and MyHC+) that eventually mature into myofibers. The satellite stem cell subpopulation (Pax7+ and Myf5−) can also self‐renew to replenish the satellite cell pool.
Figure 8. Figure 8. Myogenic regulatory factors in proliferating myoblasts. Images from single EDL myofibers cultured ex vivo for 60 h (dashed line delineates the myofiber membrane). Staining for Pax7 (green) and MyoD (red) shows that at this stage cells express either only Pax7 (arrowhead), only MyoD, or both (arrow).
Figure 9. Figure 9. Myogenic regulatory factors in differentiating myoblasts. Pictures from EDL isolated myofibers cultured ex vivo for 72 h (dashed line delineates the myofiber membrane). Staining for Pax7 (green) and myogenin (red) shows that cells that acquire the differentiation marker myogenin lose expression of Pax7.
Figure 10. Figure 10. Asymmetric and stochastic self‐renewal. Satellite cells can be divided in two subpopulations: committed satellite cells and satellite stem cells. Satellite stem cells can perform asymmetric division in an apical‐basal orientation or symmetric division in a planar orientation (relative to the myofiber). Asymmetric division allows for the formation of one committed daughter cell that will participate in myogenesis, and for the maintenance of the original satellite stem cell. Symmetric division of satellite stem cells produces two daughter stem cells and favors satellite stem cell expansion. Committed satellite cells can divide to form two committed daughter cells that participate in the myogenesis process.
Figure 11. Figure 11. Symmetric versus asymmetric divisions. Images show myofibers isolated from EDL muscles of Myf5‐cre R26R‐YFP stained with Pax7 (red), YFP (for Myf5, green), and a nuclear dye (DAPI, blue). The myofiber membrane is outlined by a dashed line. In these mice, expression of Myf5 leads to permanent yellow‐fluorescent protein (YFP) staining. Most of the satellite cells express Myf5 (YFP+) during development, but a subpopulation of satellite stem cells never expressed Myf5 (YFP−). YFP− satellite stem cells can perform asymmetric division (A) and give rise to a committed progenitor (YFP+), or perform symmetric division (B) and produce two satellite stem cells (YFP−). Asymmetric divisions occur mostly in an apical‐basal orientation (A), while symmetric divisions occur with planar orientation (B and C).
Figure 12. Figure 12. Wnt7a‐activated pathways in myogenic cells. In myogenic cells, Wnt7a binds to Fzd7 and induces various responses depending on the myogenic stage. In satellite stem cells, Wnt7a forms a complex with the Fzd7 receptor, Scd4 coreceptor and fibronectin to activate the planar cell polarity pathway. This noncanonical pathway leads to the symmetric stem cell division. In myogenic progenitors, Wnt7a leads to rearrangements of the cytoskeleton and increases directional cell migration. Lastly, in muscle fibers, Wnt7a activates the Akt/mTOR pathway, in an IGF‐1‐independent manner, and promotes myofiber hypertrophy.
Figure 13. Figure 13. Satellite cell migration. Schematic representing a migrating satellite cell. Satellite cells receive many guidance cues from their microenvironment that promote chemotaxis toward the damaged area (ex: HGF) or repulse the cell away from uninjured region (ex: Ephrins).
Figure 14. Figure 14. Myotube formation. Pictures from primary myoblasts cultured in vitro in differentiation medium (low‐serum medium). Cells are stained with a nuclear dye (DAPI, blue) and myosin heavy chain (MyHC, in green). Almost no myoblasts express MyHC before the addition of differentiation medium. After 1 day in differentiation medium, the cells start to express MyHC. At three days, myoblasts fuse to form small multinucleated myotubes (few myonuclei per myotube). After 5 days of differentiation, myotubes become larger and contain more nuclei.
Figure 15. Figure 15. Epigenetic control of stemness and lineage determination genes during myogenic differentiation. In undifferentiated satellite cells and myoblasts, lineage determination gene regulatory regions such as the myogenin promoter are repressed through a combination of epigenetic mechanisms. MyoD is repressed by its association with SIRT1 and HDACs. MEF2 also recruits HDACs as well as SUV39H1, the histone methyltransferase responsible for H3K9 methylation. The Polycomb Repressive complex 2 (PRC2) containing the histone methyltransferase EZH2 is also recruited on the myogenin promoter by the transcription factor YY1. Together, these protein complexes are responsible to establish heterochromatin formation that is repressive for transcription. In undifferentiated satellite cells and myoblasts, stemness genes like Pax7 are active and are thus occupied by histone acetyltransferases (HAT) as well as the SWI/SNF chromatin remodeling complex and the MLL/Trithorax complex, responsible for H3K4me3 deposition. These proteins promote the formation of euchromatin, which is permissive for transcription. When cells receive differentiation cues, these epigenetic complexes are relocalized to different regulatory regions. Repressive complexes leave the promoters of lineage determination genes, which become active, and could be recruited to the promoters of stemness genes that need to be silenced. In contrast, proteins involved in active chromatin configuration leave the promoters of stemness genes and are relocalized at muscle specific promoters (myogenin). The myogenin promoter is thus occupied by the MLL/Trithorax complex, by HATs, by the SWI/SNF complex as well as by the demethylase UTX.
Figure 16. Figure 16. The satellite cell microenvironment. The satellite cell niche is composed of various ECM proteins and cell types. Cell types such as inflammatory cells and stromal cells can physically interact with satellite cells or release various cytokines, growth factors and ECM components that will influence satellite cell behavior.
Figure 17. Figure 17. Inflammation and myogenesis. Inflammation is characterized by the activation of mast cells (MC), followed by the early recruitment of granulocytes (polymorphonuclear cells, PMN), and the accumulation of monocytes/macrophages. Macrophage accumulation is classically composed of an initial proinflammatory phase (M1 macrophages) followed by an anti‐inflammatory phase (M2 macrophages). These different leukocytes secrete factors that directly influence the function of myogenic cells. Initial burst of leukocytes has been shown to stimulate satellite cell activation and proliferation. M1 macrophages promote myoblast proliferation and repress early differentiation, while M2 macrophages stimulate differentiation and myofiber growth.
Figure 18. Figure 18. Degeneration of dystrophic muscles. Images from dystrophic tibialis anterior muscles. H&E staining showing centronucleated fibers that indicate muscle regeneration. Sirius red (fibrotic tissue in red) and trichrome masson (fibrotic tissue in blue) illustrate the massive fibrosis response in the dystrophic muscles.
Figure 19. Figure 19. Satellite cell therapy. Schematic illustrating the different steps of cell therapy. Healthy satellite cells are isolated from donor muscle tissue and are cultured in vitro or directly transplanted into diseased host muscle. Healthy satellite cells will then fuse to host myofibers to form hybrid fibers. Addition of new healthy nuclei to host myofibers can partially correct genetic disorders such as muscular dystrophies. However, this therapeutic avenue has major technical limitations such as short‐term engraftment, limited availability of donor cells, and poor cell migration and survival.


Figure 1. Skeletal muscle anatomy. Schematic illustrating the structure of skeletal muscle. Skeletal muscles are divided into bundles containing numerous myofibers. Myofibers are multinucleated cylindrical cells that are surrounded by a vast network of blood vessels and nerves. Satellite cells are small mononuclear cells located between the plasmalemma of the myofibers and the basal membrane.


Figure 2. Satellite cell markers. Images of single extensor digitorium longus (EDL) myofiber labeled for different markers of satellite cells. The left image shows a multinucleated myofiber with two satellite cells stained by Pax7 antibody (red) and a nuclear dye (DAPI, blue). The right picture displays a satellite cell stained with M‐cadherin (red) on the apical side and α7‐integrin (white) on the basal side.


Figure 3. The satellite cell niche. Cross‐section of a tibialis anterior (TA) muscle immunostained with laminin (white), M‐cadherin (green), Pax7 (red), and a nuclear dye (DAPI, blue). All muscle fibers are surrounded by laminin. The insert in the top right corner shows a magnification of a satellite cell (Pax7‐positive) in its niche.


Figure 4. Embryonic muscle development. The image shows a mouse embryo transgenic for a fluorescent expression reporter (TdTomato) of the myogenic transcription factor Myf5 at 10.5 days after fertilization. At this developmental stage Myf5 is highly expressed in the somites. Somites develop antero‐posteriorly, with the anterior somites being the most mature ones and the posterior somites the least developed.


Figure 5. Genetic programme regulating skeletal muscle stem cell fate. Hierarchical network of the transcription factors controlling embryonic progenitor cell fate during limb and trunk muscle development. Pax3 and Pax7 control the myogenic specification of embryonic progenitors. Pax3/7 can directly target Myf5 and MyoD expression and thus myogenic determination. Myf5 also acts partially upstream of MyoD and can trigger its expression. Upregulation of myogenin and MRF4 induces myogenic differentiation. A population of Pax3/7+ progenitor cells does not express MRFs and gives rise to satellite stem cells during late fetal myogenesis.


Figure 6. Skeletal muscle regeneration. H&E staining (nuclei stained in dark blue and cytosolic proteins stained in red) illustrating the different stages of muscle regeneration following cardiotoxin injury. Two days following injury, myofiber structure is severely impaired and a multitude of mononuclear cells (myoblasts, inflammatory cells, fibroblasts, etc.) are present in the damaged area. At five days postinjury, there is the formation of small new myofibers with centralized nuclei and many cells remain in the exudate. Two months after the injury, muscle architecture is similar to uninjured muscle.


Figure 7. Myogenic lineage progression. Following muscle injury quiescent satellite cells (Pax7+ and Myf5+/−) are activated and differentiate to myoblasts (Pax7+, Myf5+, and MyoD+). After several rounds of proliferation, myoblasts exit the cell cycle and become myocytes (Pax7−, MyoD+, myogenin+, and MRF4+). Myocytes can undergo a fusion process to form multinucleated myotubes (myogenin+, MRF4+, and MyHC+) that eventually mature into myofibers. The satellite stem cell subpopulation (Pax7+ and Myf5−) can also self‐renew to replenish the satellite cell pool.


Figure 8. Myogenic regulatory factors in proliferating myoblasts. Images from single EDL myofibers cultured ex vivo for 60 h (dashed line delineates the myofiber membrane). Staining for Pax7 (green) and MyoD (red) shows that at this stage cells express either only Pax7 (arrowhead), only MyoD, or both (arrow).


Figure 9. Myogenic regulatory factors in differentiating myoblasts. Pictures from EDL isolated myofibers cultured ex vivo for 72 h (dashed line delineates the myofiber membrane). Staining for Pax7 (green) and myogenin (red) shows that cells that acquire the differentiation marker myogenin lose expression of Pax7.


Figure 10. Asymmetric and stochastic self‐renewal. Satellite cells can be divided in two subpopulations: committed satellite cells and satellite stem cells. Satellite stem cells can perform asymmetric division in an apical‐basal orientation or symmetric division in a planar orientation (relative to the myofiber). Asymmetric division allows for the formation of one committed daughter cell that will participate in myogenesis, and for the maintenance of the original satellite stem cell. Symmetric division of satellite stem cells produces two daughter stem cells and favors satellite stem cell expansion. Committed satellite cells can divide to form two committed daughter cells that participate in the myogenesis process.


Figure 11. Symmetric versus asymmetric divisions. Images show myofibers isolated from EDL muscles of Myf5‐cre R26R‐YFP stained with Pax7 (red), YFP (for Myf5, green), and a nuclear dye (DAPI, blue). The myofiber membrane is outlined by a dashed line. In these mice, expression of Myf5 leads to permanent yellow‐fluorescent protein (YFP) staining. Most of the satellite cells express Myf5 (YFP+) during development, but a subpopulation of satellite stem cells never expressed Myf5 (YFP−). YFP− satellite stem cells can perform asymmetric division (A) and give rise to a committed progenitor (YFP+), or perform symmetric division (B) and produce two satellite stem cells (YFP−). Asymmetric divisions occur mostly in an apical‐basal orientation (A), while symmetric divisions occur with planar orientation (B and C).


Figure 12. Wnt7a‐activated pathways in myogenic cells. In myogenic cells, Wnt7a binds to Fzd7 and induces various responses depending on the myogenic stage. In satellite stem cells, Wnt7a forms a complex with the Fzd7 receptor, Scd4 coreceptor and fibronectin to activate the planar cell polarity pathway. This noncanonical pathway leads to the symmetric stem cell division. In myogenic progenitors, Wnt7a leads to rearrangements of the cytoskeleton and increases directional cell migration. Lastly, in muscle fibers, Wnt7a activates the Akt/mTOR pathway, in an IGF‐1‐independent manner, and promotes myofiber hypertrophy.


Figure 13. Satellite cell migration. Schematic representing a migrating satellite cell. Satellite cells receive many guidance cues from their microenvironment that promote chemotaxis toward the damaged area (ex: HGF) or repulse the cell away from uninjured region (ex: Ephrins).


Figure 14. Myotube formation. Pictures from primary myoblasts cultured in vitro in differentiation medium (low‐serum medium). Cells are stained with a nuclear dye (DAPI, blue) and myosin heavy chain (MyHC, in green). Almost no myoblasts express MyHC before the addition of differentiation medium. After 1 day in differentiation medium, the cells start to express MyHC. At three days, myoblasts fuse to form small multinucleated myotubes (few myonuclei per myotube). After 5 days of differentiation, myotubes become larger and contain more nuclei.


Figure 15. Epigenetic control of stemness and lineage determination genes during myogenic differentiation. In undifferentiated satellite cells and myoblasts, lineage determination gene regulatory regions such as the myogenin promoter are repressed through a combination of epigenetic mechanisms. MyoD is repressed by its association with SIRT1 and HDACs. MEF2 also recruits HDACs as well as SUV39H1, the histone methyltransferase responsible for H3K9 methylation. The Polycomb Repressive complex 2 (PRC2) containing the histone methyltransferase EZH2 is also recruited on the myogenin promoter by the transcription factor YY1. Together, these protein complexes are responsible to establish heterochromatin formation that is repressive for transcription. In undifferentiated satellite cells and myoblasts, stemness genes like Pax7 are active and are thus occupied by histone acetyltransferases (HAT) as well as the SWI/SNF chromatin remodeling complex and the MLL/Trithorax complex, responsible for H3K4me3 deposition. These proteins promote the formation of euchromatin, which is permissive for transcription. When cells receive differentiation cues, these epigenetic complexes are relocalized to different regulatory regions. Repressive complexes leave the promoters of lineage determination genes, which become active, and could be recruited to the promoters of stemness genes that need to be silenced. In contrast, proteins involved in active chromatin configuration leave the promoters of stemness genes and are relocalized at muscle specific promoters (myogenin). The myogenin promoter is thus occupied by the MLL/Trithorax complex, by HATs, by the SWI/SNF complex as well as by the demethylase UTX.


Figure 16. The satellite cell microenvironment. The satellite cell niche is composed of various ECM proteins and cell types. Cell types such as inflammatory cells and stromal cells can physically interact with satellite cells or release various cytokines, growth factors and ECM components that will influence satellite cell behavior.


Figure 17. Inflammation and myogenesis. Inflammation is characterized by the activation of mast cells (MC), followed by the early recruitment of granulocytes (polymorphonuclear cells, PMN), and the accumulation of monocytes/macrophages. Macrophage accumulation is classically composed of an initial proinflammatory phase (M1 macrophages) followed by an anti‐inflammatory phase (M2 macrophages). These different leukocytes secrete factors that directly influence the function of myogenic cells. Initial burst of leukocytes has been shown to stimulate satellite cell activation and proliferation. M1 macrophages promote myoblast proliferation and repress early differentiation, while M2 macrophages stimulate differentiation and myofiber growth.


Figure 18. Degeneration of dystrophic muscles. Images from dystrophic tibialis anterior muscles. H&E staining showing centronucleated fibers that indicate muscle regeneration. Sirius red (fibrotic tissue in red) and trichrome masson (fibrotic tissue in blue) illustrate the massive fibrosis response in the dystrophic muscles.


Figure 19. Satellite cell therapy. Schematic illustrating the different steps of cell therapy. Healthy satellite cells are isolated from donor muscle tissue and are cultured in vitro or directly transplanted into diseased host muscle. Healthy satellite cells will then fuse to host myofibers to form hybrid fibers. Addition of new healthy nuclei to host myofibers can partially correct genetic disorders such as muscular dystrophies. However, this therapeutic avenue has major technical limitations such as short‐term engraftment, limited availability of donor cells, and poor cell migration and survival.
References
 1. Abmayr SM , Pavlath GK . Myoblast fusion: Lessons from flies and mice. Development 139: 641‐656, 2012.
 2. Abou‐Khalil R , Le Grand F , Pallafacchina G , Valable S , Authier FJ , Rudnicki MA , Gherardi RK , Germain S , Chretien F , Sotiropoulos A , Lafuste P , Montarras D , Chazaud B . Autocrine and paracrine angiopoietin 1/Tie‐2 signaling promotes muscle satellite cell self‐renewal. Cell Stem Cell 5: 298‐309, 2009.
 3. Acharyya S , Sharma SM , Cheng AS , Ladner KJ , He W , Kline W , Wang H , Ostrowski MC , Huang TH , Guttridge DC . TNF inhibits Notch‐1 in skeletal muscle cells by Ezh2 and DNA methylation mediated repression: Implications in duchenne muscular dystrophy. PloS One 5: e12479, 2010.
 4. Adli M , Bernstein BE . Whole‐genome chromatin profiling from limited numbers of cells using nano‐ChIP‐seq. Nat Protoc 6: 1656‐1668, 2011.
 5. Alfaro LA , Dick SA , Siegel AL , Anonuevo AS , McNagny KM , Megeney LA , Cornelison DD , Rossi FM . CD34 promotes satellite cell motility and entry into proliferation to facilitate efficient skeletal muscle regeneration. Stem Cells 29: 2030‐2041, 2011.
 6. Allen RE , Sheehan SM , Taylor RG , Kendall TL , Rice GM . Hepatocyte growth factor activates quiescent skeletal muscle satellite cells in vitro. J Cell Physiol 165: 307‐312, 1995.
 7. Alter J , Rozentzweig D , Bengal E . Inhibition of myoblast differentiation by tumor necrosis factor alpha is mediated by c‐Jun N‐terminal kinase 1 and leukemia inhibitory factor. J Biol Chem 283: 23224‐23234, 2008.
 8. Anderson JE . A role for nitric oxide in muscle repair: Nitric oxide‐mediated activation of muscle satellite cells. Mol Biol Cell 11: 1859‐1874, 2000.
 9. Anderson MS , Kunkel LM . The molecular and biochemical basis of Duchenne muscular dystrophy. Trends Biochem Sci 17: 289‐292, 1992.
 10. Argiles JM , Busquets S , Felipe A , Lopez‐Soriano FJ . Muscle wasting in cancer and ageing: Cachexia versus sarcopenia. Adv Gerontol 18: 39‐54, 2006.
 11. 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 Exp Med 204: 1057‐1069, 2007.
 12. Asakura A , Seale P , Girgis‐Gabardo A , Rudnicki MA . Myogenic specification of side population cells in skeletal muscle. J Cell Biol 159: 123‐134, 2002.
 13. Asp P , Blum R , Vethantham V , Parisi F , Micsinai M , Cheng J , Bowman C , Kluger Y , Dynlacht BD . Genome‐wide remodeling of the epigenetic landscape during myogenic differentiation. Proc Natl Acad Sci U S A 108: E149‐E158, 2011.
 14. Auffray C , Fogg D , Garfa M , Elain G , Join‐Lambert O , Kayal S , Sarnacki S , Cumano A , Lauvau G , Geissmann F . Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science 317: 666‐670, 2007.
 15. Bachrach E , Li S , Perez AL , Schienda J , Liadaki K , Volinski J , Flint A , Chamberlain J , Kunkel LM . Systemic delivery of human microdystrophin to regenerating mouse dystrophic muscle by muscle progenitor cells. Proc Natl Acad Sci U S A 101: 3581‐3586, 2004.
 16. Barani AE , Durieux AC , Sabido O , Freyssenet D . Age‐related changes in the mitotic and metabolic characteristics of muscle‐derived cells. J Appl Physiol (1985) 95: 2089‐2098, 2003.
 17. Barjot C , Cotten ML , Goblet C , Whalen RG , Bacou F . Expression of myosin heavy chain and of myogenic regulatory factor genes in fast or slow rabbit muscle satellite cell cultures. J Muscle Res Cell Motil 16: 619‐628, 1995.
 18. Bassel‐Duby R , Olson EN . Signaling pathways in skeletal muscle remodeling. Annu Rev Biochem 75: 19‐37, 2006.
 19. Beauchamp JR , Heslop L , Yu DS , Tajbakhsh S , Kelly RG , Wernig A , Buckingham ME , Partridge TA , Zammit PS . Expression of CD34 and Myf5 defines the majority of quiescent adult skeletal muscle satellite cells. J Cell Biol 151: 1221‐1234, 2000.
 20. Bencze M , Negroni E , Vallese D , Yacoub‐Youssef H , Chaouch S , Wolff A , Aamiri A , Di Santo JP , Chazaud B , Butler‐Browne G , Savino W , Mouly V , Riederer I . Proinflammatory macrophages enhance the regenerative capacity of human myoblasts by modifying their kinetics of proliferation and differentiation. Mol Ther 20: 2168‐2179, 2012.
 21. Bentzinger CF , Lin S , Romanino K , Castets P , Guridi M , Summermatter S , Handschin C , Tintignac LA , Hall MN , Ruegg MA . Differential response of skeletal muscles to mTORC1 signaling during atrophy and hypertrophy. Skelet Muscle 3: 6, 2013.
 22. Bentzinger CF , Rudnicki MA . Rejuvenating aged muscle stem cells. Nat Med 20: 234‐235, 2014.
 23. Bentzinger CF , von Maltzahn J , Dumont NA , Stark DA , Wang YX , Nhan K , Frenette J , Cornelison DD , Rudnicki MA . Wnt7a stimulates myogenic stem cell motility and engraftment resulting in improved muscle strength. J Cell Biol 205: 97‐111, 2014.
 24. Bentzinger CF , Wang YX , Dumont NA , Rudnicki MA . Cellular dynamics in the muscle satellite cell niche. EMBO Rep 14: 1062‐1072, 2013.
 25. Bentzinger CF , Wang YX , Rudnicki MA . Building muscle: Molecular regulation of myogenesis. Cold Spring Harb Perspect Biol 4: pii: a008342, 2012.
 26. Bentzinger CF , Wang YX , von Maltzahn J , Soleimani VD , Yin H , Rudnicki MA . Fibronectin regulates Wnt7a signaling and satellite cell expansion. Cell Stem Cell 12: 75‐87, 2013.
 27. Berkes CA , Bergstrom DA , Penn BH , Seaver KJ , Knoepfler PS , Tapscott SJ . Pbx marks genes for activation by MyoD indicating a role for a homeodomain protein in establishing myogenic potential. Mol Cell 14: 465‐477, 2004.
 28. Bernet JD , Doles JD , Hall JK , Kelly Tanaka K , Carter TA , Olwin BB . p38 MAPK signaling underlies a cell‐autonomous loss of stem cell self‐renewal in skeletal muscle of aged mice. Nat Med 20: 265‐271, 2014.
 29. Bintliff S , Walker BE . Radioautographic study of skeletal muscle regeneration. Am J Anat 106: 233‐245, 1960.
 30. Biressi S , Molinaro M , Cossu G . Cellular heterogeneity during vertebrate skeletal muscle development. Dev Biol 308: 281‐293, 2007.
 31. Bischoff R . Regeneration of single skeletal muscle fibers in vitro. Anat Rec 182: 215‐235, 1975.
 32. Bischoff R . Chemotaxis of skeletal muscle satellite cells. Dev Dyn 208: 505‐515, 1997.
 33. Bjornson CR , Cheung TH , Liu L , Tripathi PV , Steeper KM , Rando TA . Notch signaling is necessary to maintain quiescence in adult muscle stem cells. Stem Cells 30: 232‐242, 2012.
 34. Blaauw B , Schiaffino S , Reggiani C . Mechanisms modulating skeletal muscle phenotype. Compr Physiol 3: 1645‐1687, 2013.
 35. Bober E , Franz T , Arnold HH , Gruss P , Tremblay P . Pax‐3 is required for the development of limb muscles: A possible role for the migration of dermomyotomal muscle progenitor cells. Development 120: 603‐612, 1994.
 36. Bonavaud S , Agbulut O , Nizard R , D'Honneur G , Mouly V , Butler‐Browne G . A discrepancy resolved: Human satellite cells are not preprogrammed to fast and slow lineages. Neuromuscul Disord 11: 747‐752, 2001.
 37. Bosnakovski D , Xu Z , Li W , Thet S , Cleaver O , Perlingeiro RC , Kyba M . Prospective isolation of skeletal muscle stem cells with a Pax7 reporter. Stem Cells 26: 3194‐3204, 2008.
 38. Bouchentouf M , Benabdallah BF , Bigey P , Yau TM , Scherman D , Tremblay JP . Vascular endothelial growth factor reduced hypoxia‐induced death of human myoblasts and improved their engraftment in mouse muscles. Gene Ther 15: 404‐414, 2008.
 39. Brack AS , Conboy IM , Conboy MJ , Shen J , Rando TA . A temporal switch from notch to Wnt signaling in muscle stem cells is necessary for normal adult myogenesis. Cell Stem Cell 2: 50‐59, 2008.
 40. Brack AS , Rando TA . Intrinsic changes and extrinsic influences of myogenic stem cell function during aging. Stem Cell Rev 3: 226‐237, 2007.
 41. Braun T , Rudnicki MA , Arnold HH , Jaenisch R . Targeted inactivation of the muscle regulatory gene Myf‐5 results in abnormal rib development and perinatal death. Cell 71: 369‐382, 1992.
 42. Buckingham M , Rigby PW . Gene regulatory networks and transcriptional mechanisms that control myogenesis. Dev Cell 28: 225‐238, 2014.
 43. Campisi J , Kim SH , Lim CS , Rubio M . Cellular senescence, cancer and aging: The telomere connection. Exp Gerontol 36: 1619‐1637, 2001.
 44. Cantini M , Massimino ML , Rapizzi E , Rossini K , Catani C , Dalla Libera L , Carraro U . Human satellite cell proliferation in vitro is regulated by autocrine secretion of IL‐6 stimulated by a soluble factor(s) released by activated monocytes. Biochem Biophys Res Commun 216: 49‐53, 1995.
 45. Capers CR . Multinucleation of skeletal muscle in vitro. J Biophy Biochem Cytol 7: 559‐565, 1960.
 46. Cardinali B , Castellani L , Fasanaro P , Basso A , Alema S , Martelli F , Falcone G . Microrna‐221 and microrna‐222 modulate differentiation and maturation of skeletal muscle cells. PloS One 4: e7607, 2009.
 47. Caretti G , Di Padova M , Micales B , Lyons GE , Sartorelli V . The Polycomb Ezh2 methyltransferase regulates muscle gene expression and skeletal muscle differentiation. Genes Dev 18: 2627‐2638, 2004.
 48. Carlson BM , Faulkner JA . Muscle transplantation between young and old rats: Age of host determines recovery. Am J Physiol 256: C1262‐C1266, 1989.
 49. Carlson ME , Conboy IM . Loss of stem cell regenerative capacity within aged niches. Aging Cell 6: 371‐382, 2007.
 50. Carlson ME , Hsu M , Conboy IM . Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature 454: 528‐532, 2008.
 51. Cerletti M , Jurga S , Witczak CA , Hirshman MF , Shadrach JL , Goodyear LJ , Wagers AJ . Highly efficient, functional engraftment of skeletal muscle stem cells in dystrophic muscles. Cell 134: 37‐47, 2008.
 52. Chakkalakal JV , Christensen J , Xiang W , Tierney MT , Boscolo FS , Sacco A , Brack AS . Early forming label‐retaining muscle stem cells require p27kip1 for maintenance of the primitive state. Development 141: 1649‐1659, 2014.
 53. Chakkalakal JV , Jones KM , Basson MA , Brack AS . The aged niche disrupts muscle stem cell quiescence. Nature 490: 355‐360, 2012.
 54. Chakravarthy MV , Abraha TW , Schwartz RJ , Fiorotto ML , Booth FW . Insulin‐like growth factor‐I extends in vitro replicative life span of skeletal muscle satellite cells by enhancing G1/S cell cycle progression via the activation of phosphatidylinositol 3'‐kinase/Akt signaling pathway. J Biol Chem 275: 35942‐35952, 2000.
 55. Charge SB , Brack AS , Hughes SM . Aging‐related satellite cell differentiation defect occurs prematurely after Ski‐induced muscle hypertrophy. Am J Physiol Cell Physiol 283: C1228‐C1241, 2002.
 56. Chen JF , Mandel EM , Thomson JM , Wu Q , Callis TE , Hammond SM , Conlon FL , Wang DZ . The role of microRNA‐1 and microRNA‐133 in skeletal muscle proliferation and differentiation. Nat Genet 38: 228‐233, 2006.
 57. Chen JF , Tao Y , Li J , Deng Z , Yan Z , Xiao X , Wang DZ . microRNA‐1 and microRNA‐206 regulate skeletal muscle satellite cell proliferation and differentiation by repressing Pax7. J Cell Biol 190: 867‐879, 2010.
 58. Chen SE , Jin B , Li YP . TNF‐alpha regulates myogenesis and muscle regeneration by activating p38 MAPK. Am J Physiol Cell Physiol 292: C1660‐C1671, 2007.
 59. Cheung TH , Quach NL , Charville GW , Liu L , Park L , Edalati A , Yoo B , Hoang P , Rando TA . Maintenance of muscle stem‐cell quiescence by microRNA‐489. Nature 482: 524‐528, 2012.
 60. Cheung TH , Rando TA . Molecular regulation of stem cell quiescence. Nat Rev Mol Cell Biol 14: 329‐340, 2013.
 61. Christov C , Chretien F , Abou‐Khalil R , Bassez G , Vallet G , Authier FJ , Bassaglia Y , Shinin V , Tajbakhsh S , Chazaud B , Gherardi RK . Muscle satellite cells and endothelial cells: Close neighbors and privileged partners. Mol Biol Cell 18: 1397‐1409, 2007.
 62. Cirillo LA , Lin FR , Cuesta I , Friedman D , Jarnik M , Zaret KS . Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA‐4. Mol Cell 9: 279‐289, 2002.
 63. Clark P , Coles D , Peckham M . Preferential adhesion to and survival on patterned laminin organizes myogenesis in vitro. Exp Cell Res 230: 275‐283, 1997.
 64. Clark WE , Blomfield LB . The efficiency of intramuscular anastomoses, with observations on the regeneration of devascularized muscle. J Anat 79: 15‐32.4, 1945.
 65. Cohn RD , Henry MD , Michele DE , Barresi R , Saito F , Moore SA , Flanagan JD , Skwarchuk MW , Robbins ME , Mendell JR , Williamson RA , Campbell KP . Disruption of DAG1 in differentiated skeletal muscle reveals a role for dystroglycan in muscle regeneration. Cell 110: 639‐648, 2002.
 66. Cohn RD , van Erp C , Habashi JP , Soleimani AA , Klein EC , Lisi MT , Gamradt M , ap Rhys CM , Holm TM , Loeys BL , Ramirez F , Judge DP , Ward CW , Dietz HC . Angiotensin II type 1 receptor blockade attenuates TGF‐beta‐induced failure of muscle regeneration in multiple myopathic states. Nat Med 13: 204‐210, 2007.
 67. Collins CA , Olsen I , Zammit PS , Heslop L , Petrie A , Partridge TA , Morgan JE . Stem cell function, self‐renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell 122: 289‐301, 2005.
 68. Conboy IM , Conboy MJ , Smythe GM , Rando TA . Notch‐mediated restoration of regenerative potential to aged muscle. Science 302: 1575‐1577, 2003.
 69. Conboy IM , Conboy MJ , Wagers AJ , Girma ER , Weissman IL , Rando TA . Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433: 760‐764, 2005.
 70. Conboy IM , Rando TA . The regulation of Notch signaling controls satellite cell activation and cell fate determination in postnatal myogenesis. Dev Cell 3: 397‐409, 2002.
 71. Corbeil HB , Whyte P , Branton PE . Characterization of transcription factor E2F complexes during muscle and neuronal differentiation. Oncogene 11: 909‐920, 1995.
 72. Cornelison DD , Olwin BB , Rudnicki MA , Wold BJ . MyoD(‐/‐) satellite cells in single‐fiber culture are differentiation defective and MRF4 deficient. Dev Biol 224: 122‐137, 2000.
 73. Cornelison DD , Wilcox‐Adelman SA , Goetinck PF , Rauvala H , Rapraeger AC , Olwin BB . Essential and separable roles for Syndecan‐3 and Syndecan‐4 in skeletal muscle development and regeneration. Genes Dev 18: 2231‐2236, 2004.
 74. Cosgrove BD , Gilbert PM , Porpiglia E , Mourkioti F , Lee SP , Corbel SY , Llewellyn ME , Delp SL , Blau HM . Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat Med 20: 255‐264, 2014.
 75. Crist CG , Montarras D , Buckingham M . Muscle satellite cells are primed for myogenesis but maintain quiescence with sequestration of Myf5 mRNA targeted by microRNA‐31 in mRNP granules. Cell Stem Cell 11: 118‐126, 2012.
 76. Cserjesi P , Olson EN . Myogenin induces the myocyte‐specific enhancer binding factor MEF‐2 independently of other muscle‐specific gene products. Mol Cell Biol 11: 4854‐4862, 1991.
 77. Dacwag CS , Ohkawa Y , Pal S , Sif S , Imbalzano AN . The protein arginine methyltransferase Prmt5 is required for myogenesis because it facilitates ATP‐dependent chromatin remodeling. Mol Cell Biol 27: 384‐394, 2007.
 78. Dalkilic I , Schienda J , Thompson TG , Kunkel LM . Loss of FilaminC (FLNc) results in severe defects in myogenesis and myotube structure. Mol Cell Biol 26: 6522‐6534, 2006.
 79. Darabi R , Arpke RW , Irion S , Dimos JT , Grskovic M , Kyba M , Perlingeiro RC . Human ES‐ and iPS‐derived myogenic progenitors restore DYSTROPHIN and improve contractility upon transplantation in dystrophic mice. Cell Stem Cell 10: 610‐619, 2012.
 80. Davie JK , Cho JH , Meadows E , Flynn JM , Knapp JR , Klein WH . Target gene selectivity of the myogenic basic helix‐loop‐helix transcription factor myogenin in embryonic muscle. Dev Biol 311: 650‐664, 2007.
 81. de la Serna IL , Carlson KA , Imbalzano AN . Mammalian SWI/SNF complexes promote MyoD‐mediated muscle differentiation. Nat Genet 27: 187‐190, 2001.
 82. de la Serna IL , Ohkawa Y , Berkes CA , Bergstrom DA , Dacwag CS , Tapscott SJ , Imbalzano AN . MyoD targets chromatin remodeling complexes to the myogenin locus prior to forming a stable DNA‐bound complex. Mol Cell Biol 25: 3997‐4009, 2005.
 83. Decary S , Hamida CB , Mouly V , Barbet JP , Hentati F , Butler‐Browne GS . Shorter telomeres in dystrophic muscle consistent with extensive regeneration in young children. Neuromuscul Disord 10: 113‐120, 2000.
 84. 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. Hum Gene Ther 8: 1429‐1438, 1997.
 85. Dellavalle A , Sampaolesi M , Tonlorenzi R , Tagliafico E , Sacchetti B , Perani L , Innocenzi A , Galvez BG , Messina G , Morosetti R , Li S , Belicchi M , Peretti G , Chamberlain JS , Wright WE , Torrente Y , Ferrari S , Bianco P , Cossu G . Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nat Cell Biol 9: 255‐267, 2007.
 86. Deng B , Wehling‐Henricks M , Villalta SA , Wang Y , Tidball JG . IL‐10 triggers changes in macrophage phenotype that promote muscle growth and regeneration. J Immunol 189: 3669‐3680, 2012.
 87. Dey BK , Gagan J , Dutta A . miR‐206 and ‐486 induce myoblast differentiation by downregulating Pax7. Mol Cell Biol 31: 203‐214, 2011.
 88. 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.
 89. Doyle MJ , Zhou S , Tanaka KK , Pisconti A , Farina NH , Sorrentino BP , Olwin BB . Abcg2 labels multiple cell types in skeletal muscle and participates in muscle regeneration. J Cell Biol 195: 147‐163, 2011.
 90. Duchesne E , Bouchard P , Roussel MP , Cote CH . Mast cells can regulate skeletal muscle cell proliferation by multiple mechanisms. Muscle Nerve 48: 403‐414, 2013.
 91. Duchesne E , Tremblay MH , Cote CH . Mast cell tryptase stimulates myoblast proliferation; A mechanism relying on protease‐activated receptor‐2 and cyclooxygenase‐2. BMC Musculoskelet Disord 12: 235, 2011.
 92. Dumont N , Bouchard P , Frenette J . Neutrophil‐induced skeletal muscle damage: A calculated and controlled response following hindlimb unloading and reloading. Am J Physiol Regul Integr Comp Physiol 295: R1831‐R1838, 2008.
 93. Dumont N , Lepage K , Cote CH , Frenette J . Mast cells can modulate leukocyte accumulation and skeletal muscle function following hindlimb unloading. J Appl Physiol (1985) 103: 97‐104, 2007.
 94. Dumont NA , Frenette J . Macrophage colony‐stimulating factor‐induced macrophage differentiation promotes regrowth in atrophied skeletal muscles and C2C12 myotubes. Am J Pathol 182: 505‐515, 2013.
 95. Edom F , Mouly V , Barbet JP , Fiszman MY , Butler‐Browne GS . Clones of human satellite cells can express in vitro both fast and slow myosin heavy chains. Dev Biol 164: 219‐229, 1994.
 96. Epstein JA , Shapiro DN , Cheng J , Lam PY , Maas RL . Pax3 modulates expression of the c‐Met receptor during limb muscle development. Proc Natl Acad Sci U S A 93: 4213‐4218, 1996.
 97. Feldman JL , Stockdale FE . Skeletal muscle satellite cell diversity: Satellite cells form fibers of different types in cell culture. Dev Biol 143: 320‐334, 1991.
 98. Fitts RH , Riley DR , Widrick JJ . Physiology of a microgravity environment invited review: Microgravity and skeletal muscle. J Appl Physiol (1985) 89: 823‐839, 2000.
 99. Floss T , Arnold HH , Braun T . A role for FGF‐6 in skeletal muscle regeneration. Genes Dev 11: 2040‐2051, 1997.
 100. Friday BB , Horsley V , Pavlath GK . Calcineurin activity is required for the initiation of skeletal muscle differentiation. J Cell Biol 149: 657‐666, 2000.
 101. Friday BB , Mitchell PO , Kegley KM , Pavlath GK . Calcineurin initiates skeletal muscle differentiation by activating MEF2 and MyoD. Differentiation 71: 217‐227, 2003.
 102. Fuchtbauer EM , Westphal H . MyoD and myogenin are coexpressed in regenerating skeletal muscle of the mouse. Dev Dyn 193: 34‐39, 1992.
 103. Fukada S , Higuchi S , Segawa M , Koda K , Yamamoto Y , Tsujikawa K , Kohama Y , Uezumi A , Imamura M , Miyagoe‐Suzuki Y , Takeda S , Yamamoto H . Purification and cell‐surface marker characterization of quiescent satellite cells from murine skeletal muscle by a novel monoclonal antibody. Exp Cell Res 296: 245‐255, 2004.
 104. Fukada S , Uezumi A , Ikemoto M , Masuda S , Segawa M , Tanimura N , Yamamoto H , Miyagoe‐Suzuki Y , Takeda S . Molecular signature of quiescent satellite cells in adult skeletal muscle. Stem Cells 25: 2448‐2459, 2007.
 105. Fukada S , Yamaguchi M , Kokubo H , Ogawa R , Uezumi A , Yoneda T , Matev MM , Motohashi N , Ito T , Zolkiewska A , Johnson RL , Saga Y , Miyagoe‐Suzuki Y , Tsujikawa K , Takeda S , Yamamoto H . Hesr1 and Hesr3 are essential to generate undifferentiated quiescent satellite cells and to maintain satellite cell numbers. Development 138: 4609‐4619, 2011.
 106. Fulco M , Schiltz RL , Iezzi S , King MT , Zhao P , Kashiwaya Y , Hoffman E , Veech RL , Sartorelli V . Sir2 regulates skeletal muscle differentiation as a potential sensor of the redox state. Mol Cell 12: 51‐62, 2003.
 107. Galli SJ , Borregaard N , Wynn TA . Phenotypic and functional plasticity of cells of innate immunity: Macrophages, mast cells and neutrophils. Nat Immunol 12: 1035‐1044, 2011.
 108. Germani A , Di Carlo A , Mangoni A , Straino S , Giacinti C , Turrini P , Biglioli P , Capogrossi MC . Vascular endothelial growth factor modulates skeletal myoblast function. Am J Pathol 163: 1417‐1428, 2003.
 109. Gibson MC , Schultz E . Age‐related differences in absolute numbers of skeletal muscle satellite cells. Muscle Nerve 6: 574‐580, 1983.
 110. 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 stem cell self‐renewal in culture. Science 329: 1078‐1081, 2010.
 111. Gildor B , Schejter ED , Shilo BZ . Bidirectional Notch activation represses fusion competence in swarming adult Drosophila myoblasts. Development 139: 4040‐4050, 2012.
 112. Gillespie MA , Le Grand F , Scime A , Kuang S , von Maltzahn J , Seale V , Cuenda A , Ranish JA , Rudnicki MA . p38‐{gamma}‐dependent gene silencing restricts entry into the myogenic differentiation program. J Cell Biol 187: 991‐1005, 2009.
 113. Gnocchi VF , White RB , Ono Y , Ellis JA , Zammit PS . Further characterisation of the molecular signature of quiescent and activated mouse muscle satellite cells. PloS One 4: e5205, 2009.
 114. Gomez‐Gaviro MV , Lovell‐Badge R , Fernandez‐Aviles F , Lara‐Pezzi E . The vascular stem cell niche. J Cardiovasc Transl Res 5: 618‐630, 2012.
 115. Gopinath SD , Webb AE , Brunet A , Rando TA . FOXO3 promotes quiescence in adult muscle stem cells during the process of self‐renewal. Stem Cell Reports 2: 414‐426, 2014.
 116. Gordon S . Alternative activation of macrophages. Nat Rev Immunol 3: 23‐35, 2003.
 117. Gratchev A , Guillot P , Hakiy N , Politz O , Orfanos CE , Schledzewski K , Goerdt S . Alternatively activated macrophages differentially express fibronectin and its splice variants and the extracellular matrix protein betaIG‐H3. Scand J Immunol 53: 386‐392, 2001.
 118. Gros J , Serralbo O , Marcelle C . WNT11 acts as a directional cue to organize the elongation of early muscle fibres. Nature 457: 589‐593, 2009.
 119. Grounds MD . Age‐associated changes in the response of skeletal muscle cells to exercise and regeneration. Ann N Y Acad Sci 854: 78‐91, 1998.
 120. 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 Tissue Res 267: 99‐104, 1992.
 121. Gundersen K , Bruusgaard JC . Nuclear domains during muscle atrophy: Nuclei lost or paradigm lost? J Physiol 586: 2675‐2681, 2008.
 122. Gunther S , Kim J , Kostin S , Lepper C , Fan CM , Braun T . Myf5‐positive satellite cells contribute to Pax7‐dependent long‐term maintenance of adult muscle stem cells. Cell Stem Cell 13: 590‐601, 2013.
 123. Gussoni E , Soneoka Y , Strickland CD , Buzney EA , Khan MK , Flint AF , Kunkel LM , Mulligan RC . Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature 401: 390‐394, 1999.
 124. 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.
 125. Halevy O , Novitch BG , Spicer DB , Skapek SX , Rhee J , Hannon GJ , Beach D , Lassar AB . Correlation of terminal cell cycle arrest of skeletal muscle with induction of p21 by MyoD. Science 267: 1018‐1021, 1995.
 126. 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.
 127. Hasty P , Bradley A , Morris JH , Edmondson DG , Venuti JM , Olson EN , Klein WH . Muscle deficiency and neonatal death in mice with a targeted mutation in the myogenin gene. Nature 364: 501‐506, 1993.
 128. Hawke TJ , Garry DJ . Myogenic satellite cells: Physiology to molecular biology. J Appl Physiol (1985) 91: 534‐551, 2001.
 129. Heredia JE , Mukundan L , Chen FM , Mueller AA , Deo RC , Locksley RM , Rando TA , Chawla A . Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell 153: 376‐388, 2013.
 130. Hernandez‐Hernandez JM , Mallappa C , Nasipak BT , Oesterreich S , Imbalzano AN . The Scaffold attachment factor b1 (Safb1) regulates myogenic differentiation by facilitating the transition of myogenic gene chromatin from a repressed to an activated state. Nucleic Acids Res 41: 5704‐5716, 2013.
 131. Heron MI , Richmond FJ . In‐series fiber architecture in long human muscles. J Morphol 216: 35‐45, 1993.
 132. Hollenberg SM , Cheng PF , Weintraub H . Use of a conditional MyoD transcription factor in studies of MyoD trans‐activation and muscle determination. Proc Natl Acad Sci U S A 90: 8028‐8032, 1993.
 133. 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.
 134. Hosoyama T , Nishijo K , Prajapati SI , Li G , Keller C . Rb1 gene inactivation expands satellite cell and postnatal myoblast pools. J Biol Chem 286: 19556‐19564, 2011.
 135. Huang YC , Dennis RG , Baar K . Cultured slow vs. fast skeletal muscle cells differ in physiology and responsiveness to stimulation. Am J Physiol Cell Physiol 291: C11‐C17, 2006.
 136. Huard J , Bouchard JP , Roy R , Malouin F , Dansereau G , Labrecque C , Albert N , Richards CL , Lemieux B , Tremblay JP . Human myoblast transplantation: Preliminary results of 4 cases. Muscle Nerve 15: 550‐560, 1992.
 137. Hutcheson DA , Zhao J , Merrell A , Haldar M , Kardon G . Embryonic and fetal limb myogenic cells are derived from developmentally distinct progenitors and have different requirements for beta‐catenin. Genes Dev 23: 997‐1013, 2009.
 138. Iezzi S , Cossu G , Nervi C , Sartorelli V , Puri PL . Stage‐specific modulation of skeletal myogenesis by inhibitors of nuclear deacetylases. Proc Natl Acad Sci U S A 99: 7757‐7762, 2002.
 139. Irintchev A , Zeschnigk M , Starzinski‐Powitz A , Wernig A . Expression pattern of M‐cadherin in normal, denervated, and regenerating mouse muscles. Dev Dyn 199: 326‐337, 1994.
 140. Ishikawa H . The fine structure of myo‐tendon junction in some mammalian skeletal muscles. Arch Histol Jpn 25: 275‐296, 1965.
 141. Ito N , Ruegg UT , Kudo A , Miyagoe‐Suzuki Y , Takeda S . Activation of calcium signaling through Trpv1 by nNOS and peroxynitrite as a key trigger of skeletal muscle hypertrophy. Nat Med 19: 101‐106, 2013.
 142. Jockusch H , Voigt S . Migration of adult myogenic precursor cells as revealed by GFP/nLacZ labelling of mouse transplantation chimeras. J Cell Sci 116: 1611‐1616, 2003.
 143. Joe AW , Yi L , Natarajan A , Le Grand F , So L , Wang J , Rudnicki MA , Rossi FM . Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nat Cell Biol 12: 153‐163, 2010.
 144. Jones NC , Tyner KJ , Nibarger L , Stanley HM , Cornelison DD , Fedorov YV , Olwin BB . The p38alpha/beta MAPK functions as a molecular switch to activate the quiescent satellite cell. J Cell Biol 169: 105‐116, 2005.
 145. Juan AH , Derfoul A , Feng X , Ryall JG , Dell'Orso S , Pasut A , Zare H , Simone JM , Rudnicki MA , Sartorelli V . Polycomb EZH2 controls self‐renewal and safeguards the transcriptional identity of skeletal muscle stem cells. Genes Dev 25: 789‐794, 2011.
 146. Juan AH , Kumar RM , Marx JG , Young RA , Sartorelli V . Mir‐214‐dependent regulation of the polycomb protein Ezh2 in skeletal muscle and embryonic stem cells. Mol Cell 36: 61‐74, 2009.
 147. Kaar JL , Li Y , Blair HC , Asche G , Koepsel RR , Huard J , Russell AJ . Matrix metalloproteinase‐1 treatment of muscle fibrosis. Acta Biomater 4: 1411‐1420, 2008.
 148. Kalhovde JM , Jerkovic R , Sefland I , Cordonnier C , Calabria E , Schiaffino S , Lomo T . “Fast” and “slow” muscle fibres in hindlimb muscles of adult rats regenerate from intrinsically different satellite cells. J Physiol 562: 847‐857, 2005.
 149. Kanisicak O , Mendez JJ , Yamamoto S , Yamamoto M , Goldhamer DJ . Progenitors of skeletal muscle satellite cells express the muscle determination gene, MyoD. Dev Biol 332: 131‐141, 2009.
 150. Karalaki M , Fili S , Philippou A , Koutsilieris M . Muscle regeneration: Cellular and molecular events. In Vivo 23: 779‐796, 2009.
 151. Kassar‐Duchossoy L , Gayraud‐Morel B , Gomes D , Rocancourt D , Buckingham M , Shinin V , Tajbakhsh S . Mrf4 determines skeletal muscle identity in Myf5:Myod double‐mutant mice. Nature 431: 466‐471, 2004.
 152. Kassar‐Duchossoy L , Giacone E , Gayraud‐Morel B , Jory A , Gomes D , Tajbakhsh S . Pax3/Pax7 mark a novel population of primitive myogenic cells during development. Genes Dev 19: 1426‐1431, 2005.
 153. 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 48: 1079‐1096, 2000.
 154. Kaushal S , Schneider JW , Nadal‐Ginard B , Mahdavi V . Activation of the myogenic lineage by MEF2A, a factor that induces and cooperates with MyoD. Science 266: 1236‐1240, 1994.
 155. Kawabe Y , Wang YX , McKinnell IW , Bedford MT , Rudnicki MA . Carm1 regulates Pax7 transcriptional activity through MLL1/2 recruitment during asymmetric satellite stem cell divisions. Cell Stem Cell 11: 333‐345, 2012.
 156. 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. Dev Biol 205: 158‐170, 1999.
 157. Kim S , Shilagardi K , Zhang S , Hong SN , Sens KL , Bo J , Gonzalez GA , Chen EH . A critical function for the actin cytoskeleton in targeted exocytosis of prefusion vesicles during myoblast fusion. Dev Cell 12: 571‐586, 2007.
 158. Komiya Y , Habas R . Wnt signal transduction pathways. Organogenesis 4: 68‐75, 2008.
 159. Koning M , Werker PM , van der Schaft DW , Bank RA , Harmsen MC . MicroRNA‐1 and microRNA‐206 improve differentiation potential of human satellite cells: A novel approach for tissue engineering of skeletal muscle. Tissue Eng Part A 18: 889‐898, 2012.
 160. Koning M , Werker PM , van Luyn MJ , Krenning G , Harmsen MC . A global downregulation of microRNAs occurs in human quiescent satellite cells during myogenesis. Differentiation 84: 314‐321, 2012.
 161. Kottlors M , Kirschner J . Elevated satellite cell number in Duchenne muscular dystrophy. Cell Tissue Res 340: 541‐548, 2010.
 162. Kuang S , Charge SB , Seale P , Huh M , Rudnicki MA . Distinct roles for Pax7 and Pax3 in adult regenerative myogenesis. J Cell Biol 172: 103‐113, 2006.
 163. Kuang S , Kuroda K , Le Grand F , Rudnicki MA . Asymmetric self‐renewal and commitment of satellite stem cells in muscle. Cell 129: 999‐1010, 2007.
 164. Lafreniere JF , Caron MC , Skuk D , Goulet M , Cheikh AR , Tremblay JP . Growth factor coinjection improves the migration potential of monkey myogenic precursors without affecting cell transplantation success. Cell Transplant 18: 719‐730, 2009.
 165. Lafreniere JF , Mills P , Bouchentouf M , Tremblay JP . Interleukin‐4 improves the migration of human myogenic precursor cells in vitro and in vivo. Exp Cell Res 312: 1127‐1141, 2006.
 166. Lansdorp PM . Immortal strands? Give me a break. Cell 129: 1244‐1247, 2007.
 167. Laurie GW , Leblond CP , Martin GR . Localization of type IV collagen, laminin, heparan sulfate proteoglycan, and fibronectin to the basal lamina of basement membranes. J Cell Biol 95: 340‐344, 1982.
 168.Le Grand F , Grifone R , Mourikis P , Houbron C , Gigaud C , Pujol J , Maillet M , Pages G , Rudnicki M , Tajbakhsh S , Maire P . Six1 regulates stem cell repair potential and self‐renewal during skeletal muscle regeneration. J Cell Biol 198: 815‐832, 2012.
 169. Le Grand F , Jones AE , Seale V , Scime A , Rudnicki MA . Wnt7a activates the planar cell polarity pathway to drive the symmetric expansion of satellite stem cells. Cell Stem Cell 4: 535‐547, 2009.
 170. Lee H , Habas R , Abate‐Shen C . MSX1 cooperates with histone H1b for inhibition of transcription and myogenesis. Science 304: 1675‐1678, 2004.
 171. Lee TI , Jenner RG , Boyer LA , Guenther MG , Levine SS , Kumar RM , Chevalier B , Johnstone SE , Cole MF , Isono K , Koseki H , Fuchikami T , Abe K , Murray HL , Zucker JP , Yuan B , Bell GW , Herbolsheimer E , Hannett NM , Sun K , Odom DT , Otte AP , Volkert TL , Bartel DP , Melton DA , Gifford DK , Jaenisch R , Young RA . Control of developmental regulators by Polycomb in human embryonic stem cells. Cell 125: 301‐313, 2006.
 172. Lepper C , Partridge TA , Fan CM . An absolute requirement for Pax7‐positive satellite cells in acute injury‐induced skeletal muscle regeneration. Development 138: 3639‐3646, 2011.
 173. Lesault PF , Theret M , Magnan M , Cuvellier S , Niu Y , Gherardi RK , Tremblay JP , Hittinger L , Chazaud B . Macrophages improve survival, proliferation and migration of engrafted myogenic precursor cells into MDX skeletal muscle. PloS One 7: e46698, 2012.
 174. Lesley J , Hyman R , Kincade PW . CD44 and its interaction with extracellular matrix. Adv Immunol 54: 271‐335, 1993.
 175. Lewis WH , Lewis MR . Behavior of cross striated muscle in tissue cultures. Am J Anat 22: 169‐194, 1917.
 176. Li YP . TNF‐alpha is a mitogen in skeletal muscle. Am J Physiol Cell Physiol 285: C370‐C376, 2003.
 177. Liadaki K , Casar JC , Wessen M , Luth ES , Jun S , Gussoni E , Kunkel LM . beta4 integrin marks interstitial myogenic progenitor cells in adult murine skeletal muscle. J Histochem Cytochem 60: 31‐44, 2012.
 178. Liu JX , Hoglund AS , Karlsson P , Lindblad J , Qaisar R , Aare S , Bengtsson E , Larsson L . Myonuclear domain size and myosin isoform expression in muscle fibres from mammals representing a 100,000‐fold difference in body size. Exp Physiol 94: 117‐129, 2009.
 179. Liu L , Cheung TH , Charville GW , Hurgo BM , Leavitt T , Shih J , Brunet A , Rando TA . Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Reports 4: 189‐204, 2013.
 180. Liu N , Nelson BR , Bezprozvannaya S , Shelton JM , Richardson JA , Bassel‐Duby R , Olson EN . Requirement of MEF2A, C, and D for skeletal muscle regeneration. Proc Natl Acad Sci U S A 111: 4109‐4114, 2014.
 181. Liu Y , Schneider MF . FGF2 activates TRPC and Ca(2+) signaling leading to satellite cell activation. Front Physiol 5: 38, 2014.
 182. Lluis F , Ballestar E , Suelves M , Esteller M , Munoz‐Canoves P . E47 phosphorylation by p38 MAPK promotes MyoD/E47 association and muscle‐specific gene transcription. EMBO J 24: 974‐984, 2005.
 183. Lu H , Huang D , Saederup N , Charo IF , Ransohoff RM , Zhou L . Macrophages recruited via CCR2 produce insulin‐like growth factor‐1 to repair acute skeletal muscle injury. FASEB J 25: 358‐369, 2011.
 184. Lu J , McKinsey TA , Nicol RL , Olson EN . Signal‐dependent activation of the MEF2 transcription factor by dissociation from histone deacetylases. Proc Natl Acad Sci U S A 97: 4070‐4075, 2000.
 185. Luo G , Hershko DD , Robb BW , Wray CJ , Hasselgren PO . IL‐1beta stimulates IL‐6 production in cultured skeletal muscle cells through activation of MAP kinase signaling pathway and NF‐kappa B. Am J Physiol Regul Integr Comp Physiol 284: R1249‐R1254, 2003.
 186. Ma K , Chan JK , Zhu G , Wu Z . Myocyte enhancer factor 2 acetylation by p300 enhances its DNA binding activity, transcriptional activity, and myogenic differentiation. Mol Cell Biol 25: 3575‐3582, 2005.
 187. Machida S , Spangenburg EE , Booth FW . Forkhead transcription factor FoxO1 transduces insulin‐like growth factor's signal to p27Kip1 in primary skeletal muscle satellite cells. J Cell Physiol 196: 523‐531, 2003.
 188. MacIntosh BR , Gardiner PF , McComas AJ . Skeletal Mmuscle: Form and Function, second edition. Champaign, Ill.; Leeds: Human Kinetics, 2006.
 189. Maher P . p38 mitogen‐activated protein kinase activation is required for fibroblast growth factor‐2‐stimulated cell proliferation but not differentiation. J Biol Chem 274: 17491‐17498, 1999.
 190. Mal AK . Histone methyltransferase Suv39h1 represses MyoD‐stimulated myogenic differentiation. EMBO J 25: 3323‐3334, 2006.
 191. Manzur AY , Kuntzer T , Pike M , Swan A . Glucocorticoid corticosteroids for Duchenne muscular dystrophy. Cochrane Database Syst Rev 1: CD003725, 2008.
 192. Mauro A . Satellite cell of skeletal muscle fibers. J Biophy Biochem Cytol 9: 493‐495, 1961.
 193. McCall GE , Allen DL , Linderman JK , Grindeland RE , Roy RR , Mukku VR , Edgerton VR . Maintenance of myonuclear domain size in rat soleus after overload and growth hormone/IGF‐I treatment. J Appl Physiol (1985) 84: 1407‐1412, 1998.
 194. McCarthy JJ , Mula J , Miyazaki M , Erfani R , Garrison K , Farooqui AB , Srikuea R , Lawson BA , Grimes B , Keller C , Van Zant G , Campbell KS , Esser KA , Dupont‐Versteegden EE , Peterson CA . Effective fiber hypertrophy in satellite cell‐depleted skeletal muscle. Development 138: 3657‐3666, 2011.
 195. McKinnell IW , Ishibashi J , Le Grand F , Punch VG , Addicks GC , Greenblatt JF , Dilworth FJ , Rudnicki MA . Pax7 activates myogenic genes by recruitment of a histone methyltransferase complex. Nat Cell Biol 10: 77‐84, 2008.
 196. McKinsey TA , Zhang CL , Lu J , Olson EN . Signal‐dependent nuclear export of a histone deacetylase regulates muscle differentiation. Nature 408: 106‐111, 2000.
 197. McKinsey TA , Zhang CL , Olson EN . Control of muscle development by dueling HATs and HDACs. Curr Opin Genet Dev 11: 497‐504, 2001.
 198. Megeney LA , Kablar B , Garrett K , Anderson JE , Rudnicki MA . MyoD is required for myogenic stem cell function in adult skeletal muscle. Genes Dev 10: 1173‐1183, 1996.
 199. Meng J , Chun S , Asfahani R , Lochmuller H , Muntoni F , Morgan J . Human skeletal muscle‐derived CD133(+) cells form functional satellite cells after intramuscular transplantation in immunodeficient host mice. Mol Ther 22: 1008‐1017, 2014.
 200. Millay DP , O'Rourke JR , Sutherland LB , Bezprozvannaya S , Shelton JM , Bassel‐Duby R , Olson EN . Myomaker is a membrane activator of myoblast fusion and muscle formation. Nature 499: 301‐305, 2013.
 201. Millay DP , Sutherland LB , Bassel‐Duby R , Olson EN . Myomaker is essential for muscle regeneration. Genes Dev 28: 1641‐1646, 2014.
 202. Mintz B , Baker WW . Normal mammalian muscle differentiation and gene control of isocitrate dehydrogenase synthesis. Proc Natl Acad Sci U S A 58: 592, 1967.
 203. Mitchell KJ , Pannerec A , Cadot B , Parlakian A , Besson V , Gomes ER , Marazzi G , Sassoon DA . Identification and characterization of a non‐satellite cell muscle resident progenitor during postnatal development. Nat Cell Biol 12: 257‐266, 2010.
 204. 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.
 205. Montarras D , L'Honore A , Buckingham M . Lying low but ready for action: The quiescent muscle satellite cell. FEBS J 280: 4036‐4050, 2013.
 206. Montarras D , Morgan J , Collins C , Relaix F , Zaffran S , Cumano A , Partridge T , Buckingham M . Direct isolation of satellite cells for skeletal muscle regeneration. Science 309: 2064‐2067, 2005.
 207. Morgan J , Rouche A , Bausero P , Houssaini A , Gross J , Fiszman MY , Alameddine HS . MMP‐9 overexpression improves myogenic cell migration and engraftment. Muscle Nerve 42: 584‐595, 2010.
 208. Mourikis P , Sambasivan R , Castel D , Rocheteau P , Bizzarro V , Tajbakhsh S . A critical requirement for notch signaling in maintenance of the quiescent skeletal muscle stem cell state. Stem Cells 30: 243‐252, 2012.
 209. Murphy MM , Lawson JA , Mathew SJ , Hutcheson DA , Kardon G . Satellite cells, connective tissue fibroblasts and their interactions are crucial for muscle regeneration. Development 138: 3625‐3637, 2011.
 210. Musaro A , McCullagh K , Paul A , Houghton L , Dobrowolny G , Molinaro M , Barton ER , Sweeney HL , Rosenthal N . Localized Igf‐1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat Genet 27: 195‐200, 2001.
 211. Muskiewicz KR , Frank NY , Flint AF , Gussoni E . Myogenic potential of muscle side and main population cells after intravenous injection into sub‐lethally irradiated mdx mice. J Histochem Cytochem 53: 861‐873, 2005.
 212. Mylona E , Jones KA , Mills ST , Pavlath GK . CD44 regulates myoblast migration and differentiation. J Cell Physiol 209: 314‐321, 2006.
 213. Nagata Y , Kobayashi H , Umeda M , Ohta N , Kawashima S , Zammit PS , Matsuda R . Sphingomyelin levels in the plasma membrane correlate with the activation state of muscle satellite cells. J Histochem Cytochem 54: 375‐384, 2006.
 214. Nagata Y , Partridge TA , Matsuda R , Zammit PS . Entry of muscle satellite cells into the cell cycle requires sphingolipid signaling. J Cell Biol 174: 245‐253, 2006.
 215. Neuhaus P , Oustanina S , Loch T , Kruger M , Bober E , Dono R , Zeller R , Braun T . Reduced mobility of fibroblast growth factor (FGF)‐deficient myoblasts might contribute to dystrophic changes in the musculature of FGF2/FGF6/mdx triple‐mutant mice. Mol Cell Biol 23: 6037‐6048, 2003.
 216. Ng RK , Gurdon JB . Epigenetic memory of an active gene state depends on histone H3.3 incorporation into chromatin in the absence of transcription. Nat Cell Biol 10: 102‐109, 2008.
 217. Nishimura T , Nakamura K , Kishioka Y , Kato‐Mori Y , Wakamatsu J , Hattori A . Inhibition of matrix metalloproteinases suppresses the migration of skeletal muscle cells. J Muscle Res Cell Motil 29: 37‐44, 2008.
 218. Olguin HC , Yang Z , Tapscott SJ , Olwin BB . Reciprocal inhibition between Pax7 and muscle regulatory factors modulates myogenic cell fate determination. J Cell Biol 177: 769‐779, 2007.
 219. Ono Y , Masuda S , Nam HS , Benezra R , Miyagoe‐Suzuki Y , Takeda S . Slow‐dividing satellite cells retain long‐term self‐renewal ability in adult muscle. J Cell Sci 125: 1309‐1317, 2012.
 220. Otis JS , Niccoli S , Hawdon N , Sarvas JL , Frye MA , Chicco AJ , Lees SJ . Pro‐inflammatory mediation of myoblast proliferation. PloS One 9: e92363, 2014.
 221. Oustanina S , Hause G , Braun T . Pax7 directs postnatal renewal and propagation of myogenic satellite cells but not their specification. EMBO J 23: 3430‐3439, 2004.
 222. Palacios D , Mozzetta C , Consalvi S , Caretti G , Saccone V , Proserpio V , Marquez VE , Valente S , Mai A , Forcales SV , Sartorelli V , Puri PL . TNF/p38alpha/polycomb signaling to Pax7 locus in satellite cells links inflammation to the epigenetic control of muscle regeneration. Cell Stem Cell 7: 455‐469, 2010.
 223. Pallafacchina G , Francois S , Regnault B , Czarny B , Dive V , Cumano A , Montarras D , Buckingham M . An adult tissue‐specific stem cell in its niche: A gene profiling analysis of in vivo quiescent and activated muscle satellite cells. Stem Cell Res 4: 77‐91, 2010.
 224. Pannerec A , Marazzi G , Sassoon D . Stem cells in the hood: The skeletal muscle niche. Trends Mol Med 18: 599‐606, 2012.
 225. Park IH , Chen J . Mammalian target of rapamycin (mTOR) signaling is required for a late‐stage fusion process during skeletal myotube maturation. J Biol Chem 280: 32009‐32017, 2005.
 226. Park IH , Erbay E , Nuzzi P , Chen J . Skeletal myocyte hypertrophy requires mTOR kinase activity and S6K1. Exp Cell Res 309: 211‐219, 2005.
 227. Parsons SA , Millay DP , Wilkins BJ , Bueno OF , Tsika GL , Neilson JR , Liberatore CM , Yutzey KE , Crabtree GR , Tsika RW , Molkentin JD . Genetic loss of calcineurin blocks mechanical overload‐induced skeletal muscle fiber type switching but not hypertrophy. J Biol Chem 279: 26192‐26200, 2004.
 228. 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 337: 176‐179, 1989.
 229. Pasut A , Oleynik P , Rudnicki MA . Isolation of muscle stem cells by fluorescence activated cell sorting cytometry. Methods Mol Biol 798: 53‐64, 2012.
 230. Perdiguero E , Ruiz‐Bonilla V , Serrano AL , Munoz‐Canoves P . Genetic deficiency of p38alpha reveals its critical role in myoblast cell cycle exit: The p38alpha‐JNK connection. Cell Cycle 6: 1298‐1303, 2007.
 231. Perrone CE , Fenwick‐Smith D , Vandenburgh HH . Collagen and stretch modulate autocrine secretion of insulin‐like growth factor‐1 and insulin‐like growth factor binding proteins from differentiated skeletal muscle cells. J Biol Chem 270: 2099‐2106, 1995.
 232. Pietsch P . Differentiation in regeneration. I. The development of muscle and cartilage following deplantation of of regenerating limb blastemata of Amblystoma larvae. Dev Biol 3: 255‐264, 1961.
 233. Pisconti A , Cornelison DD , Olguin HC , Antwine TL , Olwin BB . Syndecan‐3 and Notch cooperate in regulating adult myogenesis. J Cell Biol 190: 427‐441, 2010.
 234. Pizza FX , McLoughlin TJ , McGregor SJ , Calomeni EP , Gunning WT . Neutrophils injure cultured skeletal myotubes. Am J Physiol Cell Physiol 281: C335‐C341, 2001.
 235. 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.
 236. Prelovsek O , Mars T , Jevsek M , Podbregar M , Grubic Z . High dexamethasone concentration prevents stimulatory effects of TNF‐alpha and LPS on IL‐6 secretion from the precursors of human muscle regeneration. Am J Physiol Regul Integr Comp Physiol 291: R1651‐R1656, 2006.
 237. Price FD , von Maltzahn J , Bentzinger CF , Dumont NA , Yin H , Chang NC , Wilson DH , Frenette J , Rudnicki MA . Inhibition of JAK‐STAT signaling stimulates adult satellite cell function. Nat Med 20: 1174‐1181, 2014.
 238. Proctor DN , O'Brien PC , Atkinson EJ , Nair KS . Comparison of techniques to estimate total body skeletal muscle mass in people of different age groups. Am J Physiol 277: E489‐E495, 1999.
 239. Punch VG , Jones AE , Rudnicki MA . Transcriptional networks that regulate muscle stem cell function. Wiley Interdiscip Rev Syst Biol Med 1: 128‐140, 2009.
 240. Puri PL , Iezzi S , Stiegler P , Chen TT , Schiltz RL , Muscat GE , Giordano A , Kedes L , Wang JY , Sartorelli V . Class I histone deacetylases sequentially interact with MyoD and pRb during skeletal myogenesis. Mol Cell 8: 885‐897, 2001.
 241. Rampalli S , Li L , Mak E , Ge K , Brand M , Tapscott SJ , Dilworth FJ . p38 MAPK signaling regulates recruitment of Ash2L‐containing methyltransferase complexes to specific genes during differentiation. Nat Struct Mol Biol 14: 1150‐1156, 2007.
 242. Rawls A , Valdez MR , Zhang W , Richardson J , Klein WH , Olson EN . Overlapping functions of the myogenic bHLH genes MRF4 and MyoD revealed in double mutant mice. Development 125: 2349‐2358, 1998.
 243. Reimann J , Irintchev A , Wernig A . Regenerative capacity and the number of satellite cells in soleus muscles of normal and mdx mice. Neuromuscul Disord 10: 276‐282, 2000.
 244. Relaix F , Montarras D , Zaffran S , Gayraud‐Morel B , Rocancourt D , Tajbakhsh S , Mansouri A , Cumano A , Buckingham M . Pax3 and Pax7 have distinct and overlapping functions in adult muscle progenitor cells. J Cell Biol 172: 91‐102, 2006.
 245. Relaix F , Rocancourt D , Mansouri A , Buckingham M . Divergent functions of murine Pax3 and Pax7 in limb muscle development. Genes Dev 18: 1088‐1105, 2004.
 246. Relaix F , Rocancourt D , Mansouri A , Buckingham M . A Pax3/Pax7‐dependent population of skeletal muscle progenitor cells. Nature 435: 948‐953, 2005.
 247. Rocheteau P , Gayraud‐Morel B , Siegl‐Cachedenier I , Blasco MA , Tajbakhsh S . A subpopulation of adult skeletal muscle stem cells retains all template DNA strands after cell division. Cell 148: 112‐125, 2012.
 248. Rochlin K , Yu S , Roy S , Baylies MK . Myoblast fusion: When it takes more to make one. Dev Biol 341: 66‐83, 2010.
 249. Rommel C , Bodine SC , Clarke BA , Rossman R , Nunez L , Stitt TN , Yancopoulos GD , Glass DJ . Mediation of IGF‐1‐induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat Cell Biol 3: 1009‐1013, 2001.
 250. Rosenblatt JD , Parry DJ , Partridge TA . Phenotype of adult mouse muscle myoblasts reflects their fiber type of origin. Differentiation 60: 39‐45, 1996.
 251. Rudnicki MA , Braun T , Hinuma S , Jaenisch R . Inactivation of MyoD in mice leads to up‐regulation of the myogenic HLH gene Myf‐5 and results in apparently normal muscle development. Cell 71: 383‐390, 1992.
 252. Rudnicki MA , Schnegelsberg PN , Stead RH , Braun T , Arnold HH , Jaenisch R . MyoD or Myf‐5 is required for the formation of skeletal muscle. Cell 75: 1351‐1359, 1993.
 253. Ruffell D , Mourkioti F , Gambardella A , Kirstetter P , Lopez RG , Rosenthal N , Nerlov C . A CREB‐C/EBPbeta cascade induces M2 macrophage‐specific gene expression and promotes muscle injury repair. Proc Natl Acad Sci U S A 106: 17475‐17480, 2009.
 254. Sabourin LA , Girgis‐Gabardo A , Seale P , Asakura A , Rudnicki MA . Reduced differentiation potential of primary MyoD‐/‐ myogenic cells derived from adult skeletal muscle. J Cell Biol 144: 631‐643, 1999.
 255. Sacco A , Doyonnas R , Kraft P , Vitorovic S , Blau HM . Self‐renewal and expansion of single transplanted muscle stem cells. Nature 456: 502‐506, 2008.
 256. Sacco A , Mourkioti F , Tran R , Choi J , Llewellyn M , Kraft P , Shkreli M , Delp S , Pomerantz JH , Artandi SE , Blau HM . Short telomeres and stem cell exhaustion model Duchenne muscular dystrophy in mdx/mTR mice. Cell 143: 1059‐1071, 2010.
 257. Saclier M , Yacoub‐Youssef H , Mackey AL , Arnold L , Ardjoune H , Magnan M , Sailhan F , Chelly J , Pavlath GK , Mounier R , Kjaer M , Chazaud B . Differentially activated macrophages orchestrate myogenic precursor cell fate during human skeletal muscle regeneration. Stem Cells 31: 384‐396, 2013.
 258. Sadeh M . Effects of aging on skeletal muscle regeneration. J Neurol Sci 87: 67‐74, 1988.
 259. Sambasivan R , Yao R , Kissenpfennig A , Van Wittenberghe L , Paldi A , Gayraud‐Morel B , Guenou H , Malissen B , Tajbakhsh S , Galy A . Pax7‐expressing satellite cells are indispensable for adult skeletal muscle regeneration. Development 138: 3647‐3656, 2011.
 260. Sampaolesi M , Torrente Y , Innocenzi A , Tonlorenzi R , D'Antona G , Pellegrino MA , Barresi R , Bresolin N , De Angelis MG , Campbell KP , Bottinelli R , Cossu G . Cell therapy of alpha‐sarcoglycan null dystrophic mice through intra‐arterial delivery of mesoangioblasts. Science 301: 487‐492, 2003.
 261. Sandri M , Sandri C , Gilbert A , Skurk C , Calabria E , Picard A , Walsh K , Schiaffino S , Lecker SH , Goldberg AL . Foxo transcription factors induce the atrophy‐related ubiquitin ligase atrogin‐1 and cause skeletal muscle atrophy. Cell 117: 399‐412, 2004.
 262. Sanes JR . The basement membrane/basal lamina of skeletal muscle. J Biol Chem 278: 12601‐12604, 2003.
 263. Sartorelli V , Puri PL , Hamamori Y , Ogryzko V , Chung G , Nakatani Y , Wang JY , Kedes L . Acetylation of MyoD directed by PCAF is necessary for the execution of the muscle program. Mol Cell 4: 725‐734, 1999.
 264. Scharner J , Zammit PS . The muscle satellite cell at 50: The formative years. Skelet Muscle 1: 28, 2011.
 265. Schnoor M , Cullen P , Lorkowski J , Stolle K , Robenek H , Troyer D , Rauterberg J , Lorkowski S . Production of type VI collagen by human macrophages: A new dimension in macrophage functional heterogeneity. J Immunol 180: 5707‐5719, 2008.
 266. Schultz E . Satellite cell proliferative compartments in growing skeletal muscles. Dev Biol 175: 84‐94, 1996.
 267. Schultz E , Jaryszak DL , Valliere CR . Response of satellite cells to focal skeletal muscle injury. Muscle Nerve 8: 217‐222, 1985.
 268. Schwander M , Leu M , Stumm M , Dorchies OM , Ruegg UT , Schittny J , Muller U . Beta1 integrins regulate myoblast fusion and sarcomere assembly. Dev Cell 4: 673‐685, 2003.
 269. Seale P , Sabourin LA , Girgis‐Gabardo A , Mansouri A , Gruss P , Rudnicki MA . Pax7 is required for the specification of myogenic satellite cells. Cell 102: 777‐786, 2000.
 270. Seenundun S , Rampalli S , Liu QC , Aziz A , Palii C , Hong S , Blais A , Brand M , Ge K , Dilworth FJ . UTX mediates demethylation of H3K27me3 at muscle‐specific genes during myogenesis. EMBO J 29: 1401‐1411, 2010.
 271. Serra C , Palacios D , Mozzetta C , Forcales SV , Morantte I , Ripani M , Jones DR , Du K , Jhala US , Simone C , Puri PL . Functional interdependence at the chromatin level between the MKK6/p38 and IGF1/PI3K/AKT pathways during muscle differentiation. Mol Cell 28: 200‐213, 2007.
 272. Serrano AL , Baeza‐Raja B , Perdiguero E , Jardi M , Munoz‐Canoves P . Interleukin‐6 is an essential regulator of satellite cell‐mediated skeletal muscle hypertrophy. Cell Metab 7: 33‐44, 2008.
 273. Serrano AL , Mann CJ , Vidal B , Ardite E , Perdiguero E , Munoz‐Canoves P . Cellular and molecular mechanisms regulating fibrosis in skeletal muscle repair and disease. Curr Top Dev Biol 96: 167‐201, 2011.
 274. Shea KL , Xiang W , LaPorta VS , Licht JD , Keller C , Basson MA , Brack AS . Sprouty1 regulates reversible quiescence of a self‐renewing adult muscle stem cell pool during regeneration. Cell Stem Cell 6: 117‐129, 2010.
 275. Shefer G , Van de Mark DP , Richardson JB , Yablonka‐Reuveni Z . Satellite‐cell pool size does matter: Defining the myogenic potency of aging skeletal muscle. Dev Biol 294: 50‐66, 2006.
 276. Shi X , Garry DJ . Muscle stem cells in development, regeneration, and disease. Genes Dev 20: 1692‐1708, 2006.
 277. Shinin V , Gayraud‐Morel B , Gomes D , Tajbakhsh S . Asymmetric division and cosegregation of template DNA strands in adult muscle satellite cells. Nat Cell Biol 8: 677‐687, 2006.
 278. Siegel AL , Atchison K , Fisher KE , Davis GE , Cornelison DD . 3D timelapse analysis of muscle satellite cell motility. Stem Cells 27: 2527‐2538, 2009.
 279. Simone C , Forcales SV , Hill DA , Imbalzano AN , Latella L , Puri PL . p38 pathway targets SWI‐SNF chromatin‐remodeling complex to muscle‐specific loci. Nat Genet 36: 738‐743, 2004.
 280. Skuk D , Goulet M , Roy B , Tremblay JP . Efficacy of myoblast transplantation in nonhuman primates following simple intramuscular cell injections: Toward defining strategies applicable to humans. Exp Neurol 175: 112‐126, 2002.
 281. Soehnlein O , Lindbom L . Phagocyte partnership during the onset and resolution of inflammation. Nat Rev Immunol 10: 427‐439, 2010.
 282. Sohn RL , Huang P , Kawahara G , Mitchell M , Guyon J , Kalluri R , Kunkel LM , Gussoni E . A role for nephrin, a renal protein, in vertebrate skeletal muscle cell fusion. Proc Natl Acad Sci U S A 106: 9274‐9279, 2009.
 283. Soleimani VD , Punch VG , Kawabe Y , Jones AE , Palidwor GA , Porter CJ , Cross JW , Carvajal JJ , Kockx CE , van IWF , Perkins TJ , Rigby PW , Grosveld F , Rudnicki MA . Transcriptional dominance of Pax7 in adult myogenesis is due to high‐affinity recognition of homeodomain motifs. Dev Cell 22: 1208‐1220, 2012.
 284. Sonnet C , Lafuste P , Arnold L , Brigitte M , Poron F , Authier FJ , Chretien F , Gherardi RK , Chazaud B . Human macrophages rescue myoblasts and myotubes from apoptosis through a set of adhesion molecular systems. J Cell Sci 119: 2497‐2507, 2006.
 285. Sousa‐Victor P , Gutarra S , Garcia‐Prat L , Rodriguez‐Ubreva J , Ortet L , Ruiz‐Bonilla V , Jardi M , Ballestar E , Gonzalez S , Serrano AL , Perdiguero E , Munoz‐Canoves P . Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 506: 316‐321, 2014.
 286. Stark DA , Karvas RM , Siegel AL , Cornelison DD . Eph/ephrin interactions modulate muscle satellite cell motility and patterning. Development 138: 5279‐5289, 2011.
 287. Stradal TE , Rottner K , Disanza A , Confalonieri S , Innocenti M , Scita G . Regulation of actin dynamics by WASP and WAVE family proteins. Trends Cell Biol 14: 303‐311, 2004.
 288. Strunkelnberg M , Bonengel B , Moda LM , Hertenstein A , de Couet HG , Ramos RG , Fischbach KF . rst and its paralogue kirre act redundantly during embryonic muscle development in Drosophila. Development 128: 4229‐4239, 2001.
 289. Sun L , Ma K , Wang H , Xiao F , Gao Y , Zhang W , Wang K , Gao X , Ip N , Wu Z . JAK1‐STAT1‐STAT3, a key pathway promoting proliferation and preventing premature differentiation of myoblasts. J Cell Biol 179: 129‐138, 2007.
 290. Tajbakhsh S . Skeletal muscle stem cells in developmental versus regenerative myogenesis. J Intern Med 266: 372‐389, 2009.
 291. Tanaka KK , Hall JK , Troy AA , Cornelison DD , Majka SM , Olwin BB . Syndecan‐4‐expressing muscle progenitor cells in the SP engraft as satellite cells during muscle regeneration. Cell Stem Cell 4: 217‐225, 2009.
 292. Tang F , Barbacioru C , Nordman E , Li B , Xu N , Bashkirov VI , Lao K , Surani MA . RNA‐Seq analysis to capture the transcriptome landscape of a single cell. Nat Protoc 5: 516‐535, 2010.
 293. Taniguti AP , Pertille A , Matsumura CY , Santo Neto H , Marques MJ . Prevention of muscle fibrosis and myonecrosis in mdx mice by suramin, a TGF‐beta1 blocker. Muscle Nerve 43: 82‐87, 2011.
 294. Tatsumi R . Mechano‐biology of skeletal muscle hypertrophy and regeneration: Possible mechanism of stretch‐induced activation of resident myogenic stem cells. Anim Sci J 81: 11‐20, 2010.
 295. 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. Dev Biol 194: 114‐128, 1998.
 296. Taylor SM , Jones PA . Multiple new phenotypes induced in 10T1/2 and 3T3 cells treated with 5‐azacytidine. Cell 17: 771‐779, 1979.
 297. Tedesco FS , Dellavalle A , Diaz‐Manera J , Messina G , Cossu G . Repairing skeletal muscle: Regenerative potential of skeletal muscle stem cells. J Clin Invest 120: 11‐19, 2010.
 298. Tidball JG , Villalta SA . Regulatory interactions between muscle and the immune system during muscle regeneration. Am J Physiol Regul Integr Comp Physiol 298: R1173‐R1187, 2010.
 299. Tierney MT , Aydogdu T , Sala D , Malecova B , Gatto S , Puri PL , Latella L , Sacco A . STAT3 signaling controls satellite cell expansion and skeletal muscle repair. Nat Med 20: 1182‐1186, 2014.
 300. Torrente Y , Belicchi M , Sampaolesi M , Pisati F , Meregalli M , D'Antona G , Tonlorenzi R , Porretti L , Gavina M , Mamchaoui K , Pellegrino MA , Furling D , Mouly V , Butler‐Browne GS , Bottinelli R , Cossu G , Bresolin N . Human circulating AC133(+) stem cells restore dystrophin expression and ameliorate function in dystrophic skeletal muscle. J Clin Invest 114: 182‐195, 2004.
 301. Tremblay P , Dietrich S , Mericskay M , Schubert FR , Li Z , Paulin D . A crucial role for Pax3 in the development of the hypaxial musculature and the long‐range migration of muscle precursors. Dev Biol 203: 49‐61, 1998.
 302. Troy A , Cadwallader AB , Fedorov Y , Tyner K , Tanaka KK , Olwin BB . Coordination of satellite cell activation and self‐renewal by Par‐complex‐dependent asymmetric activation of p38alpha/beta MAPK. Cell Stem Cell 11: 541‐553, 2012.
 303. Uezumi A , Fukada S , Yamamoto N , Takeda S , Tsuchida K . Mesenchymal progenitors distinct from satellite cells contribute to ectopic fat cell formation in skeletal muscle. Nat Cell Biol 12: 143‐152, 2010.
 304. Urciuolo A , Quarta M , Morbidoni V , Gattazzo F , Molon S , Grumati P , Montemurro F , Tedesco FS , Blaauw B , Cossu G , Vozzi G , Rando TA , Bonaldo P . Collagen VI regulates satellite cell self‐renewal and muscle regeneration. Nat Commun 4: 1964, 2013.
 305. van der Vlag J , Otte AP . Transcriptional repression mediated by the human polycomb‐group protein EED involves histone deacetylation. Nat Genet 23: 474‐478, 1999.
 306. Verrier L , Escaffit F , Chailleux C , Trouche D , Vandromme M . A new isoform of the histone demethylase JMJD2A/KDM4A is required for skeletal muscle differentiation. PLoS Genet 7: e1001390, 2011.
 307. 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. Hum Mol Genet 18: 482‐496, 2009.
 308. von Maltzahn J , Bentzinger CF , Rudnicki MA . Wnt7a‐Fzd7 signalling directly activates the Akt/mTOR anabolic growth pathway in skeletal muscle. Nat Cell Biol 14: 186‐191, 2012.
 309. von Maltzahn J , Jones AE , Parks RJ , Rudnicki MA . Pax7 is critical for the normal function of satellite cells in adult skeletal muscle. Proc Natl Acad Sci U S A 110: 16474‐16479, 2013.
 310. von Maltzahn J , Renaud JM , Parise G , Rudnicki MA . Wnt7a treatment ameliorates muscular dystrophy. Proc Natl Acad Sci U S A 109: 20614‐20619, 2012.
 311. von Maltzahn J , Zinoviev R , Chang NC , Bentzinger CF , Rudnicki MA . A truncated Wnt7a retains full biological activity in skeletal muscle. Nat Commun 4: 2869, 2013.
 312. Wang X , Wu H , Zhang Z , Liu S , Yang J , Chen X , Fan M , Wang X . Effects of interleukin‐6, leukemia inhibitory factor, and ciliary neurotrophic factor on the proliferation and differentiation of adult human myoblasts. Cell Mol Neurobiol 28: 113‐124, 2008.
 313. Watt DJ , Morgan JE , Clifford MA , Partridge TA . The movement of muscle precursor cells between adjacent regenerating muscles in the mouse. Anat Embryol (Berl) 175: 527‐536, 1987.
 314. Webster C , Blau HM . Accelerated age‐related decline in replicative life‐span of Duchenne muscular dystrophy myoblasts: Implications for cell and gene therapy. Somat Cell Mol Genet 16: 557‐565, 1990.
 315. Wei L , Zhou W , Croissant JD , Johansen FE , Prywes R , Balasubramanyam A , Schwartz RJ . RhoA signaling via serum response factor plays an obligatory role in myogenic differentiation. J Biol Chem 273: 30287‐30294, 1998.
 316. Weintraub H , Davis R , Tapscott S , Thayer M , Krause M , Benezra R , Blackwell TK , Turner D , Rupp R , Hollenberg S , et al. The myoD gene family: Nodal point during specification of the muscle cell lineage. Science 251: 761‐766, 1991.
 317. Weintraub H , Tapscott SJ , Davis RL , Thayer MJ , Adam MA , Lassar AB , Miller AD . Activation of muscle‐specific genes in pigment, nerve, fat, liver, and fibroblast cell lines by forced expression of MyoD. Proc Natl Acad Sci U S A 86: 5434‐5438, 1989.
 318. Weir AP , Morgan JE , Davies KE . A‐utrophin up‐regulation in mdx skeletal muscle is independent of regeneration. Neuromuscul Disord 14: 19‐23, 2004.
 319. Wen Y , Bi P , Liu W , Asakura A , Keller C , Kuang S . Constitutive Notch activation upregulates Pax7 and promotes the self‐renewal of skeletal muscle satellite cells. Mol Cell Biol 32: 2300‐2311, 2012.
 320. Wong CF , Tellam RL . MicroRNA‐26a targets the histone methyltransferase Enhancer of Zeste homolog 2 during myogenesis. J Biol Chem 283: 9836‐9843, 2008.
 321. Wu Z , Woodring PJ , Bhakta KS , Tamura K , Wen F , Feramisco JR , Karin M , Wang JY , Puri PL . p38 and extracellular signal‐regulated kinases regulate the myogenic program at multiple steps. Mol Cell Biol 20: 3951‐3964, 2000.
 322. Xu Q , Yu L , Liu L , Cheung CF , Li X , Yee SP , Yang XJ , Wu Z . p38 Mitogen‐activated protein kinase‐, calcium‐calmodulin‐dependent protein kinase‐, and calcineurin‐mediated signaling pathways transcriptionally regulate myogenin expression. Mol Biol Cell 13: 1940‐1952, 2002.
 323. Yablonka‐Reuveni Z , Rivera AJ . Influence of PDGF‐BB on proliferation and transition through the MyoD‐myogenin‐MEF2A expression program during myogenesis in mouse C2 myoblasts. Growth Factors 15: 1‐27, 1997.
 324. Yamada M , Tatsumi R , Yamanouchi K , Hosoyama T , Shiratsuchi S , Sato A , Mizunoya W , Ikeuchi Y , Furuse M , Allen RE . High concentrations of HGF inhibit skeletal muscle satellite cell proliferation in vitro by inducing expression of myostatin: A possible mechanism for reestablishing satellite cell quiescence in vivo. Am J Physiol Cell Physiol 298: C465‐C476, 2010.
 325. Yamaguchi M , Ogawa R , Watanabe Y , Uezumi A , Miyagoe‐Suzuki Y , Tsujikawa K , Yamamoto H , Takeda S , Fukada S . Calcitonin receptor and Odz4 are differently expressed in Pax7‐positive cells during skeletal muscle regeneration. J Mol Histol 43: 581‐587, 2012.
 326. Yennek S , Burute M , Thery M , Tajbakhsh S . Cell adhesion geometry regulates non‐random DNA segregation and asymmetric cell fates in mouse skeletal muscle stem cells. Cell Reports 7: 961‐970, 2014.
 327. Yin H , Price F , Rudnicki MA . Satellite cells and the muscle stem cell niche. Physiol Rev 93: 23‐67, 2013.
 328. Yoshida N , Yoshida S , Koishi K , Masuda K , Nabeshima Y . Cell heterogeneity upon myogenic differentiation: Down‐regulation of MyoD and Myf‐5 generates ‘reserve cells’. J Cell Sci 111(Pt 6): 769‐779, 1998.
 329. Zammit PS , Golding JP , Nagata Y , Hudon V , Partridge TA , Beauchamp JR . Muscle satellite cells adopt divergent fates: A mechanism for self‐renewal? J Cell Biol 166: 347‐357, 2004.
 330. Zhang CL , McKinsey TA , Olson EN . Association of class II histone deacetylases with heterochromatin protein 1: Potential role for histone methylation in control of muscle differentiation. Mol Cell Biol 22: 7302‐7312, 2002.
 331. Zhang W , Behringer RR , Olson EN . Inactivation of the myogenic bHLH gene MRF4 results in up‐regulation of myogenin and rib anomalies. Genes Dev 9: 1388‐1399, 1995.
 332. Zhao M , New L , Kravchenko VV , Kato Y , Gram H , di Padova F , Olson EN , Ulevitch RJ , Han J . Regulation of the MEF2 family of transcription factors by p38. Mol Cell Biol 19: 21‐30, 1999.
 333. Zhao X , Sternsdorf T , Bolger TA , Evans RM , Yao TP . Regulation of MEF2 by histone deacetylase 4‐ and SIRT1 deacetylase‐mediated lysine modifications. Mol Cell Biol 25: 8456‐8464, 2005.
 334. Zhao XD , Han X , Chew JL , Liu J , Chiu KP , Choo A , Orlov YL , Sung WK , Shahab A , Kuznetsov VA , Bourque G , Oh S , Ruan Y , Ng HH , Wei CL . Whole‐genome mapping of histone H3 Lys4 and 27 trimethylations reveals distinct genomic compartments in human embryonic stem cells. Cell Stem Cell 1: 286‐298, 2007.
 335. Zhou VW , Goren A , Bernstein BE . Charting histone modifications and the functional organization of mammalian genomes. Nat Rev Genet 12: 7‐18, 2011.

Contact Editor

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

* Required Field

How to Cite

Nicolas A. Dumont, C. Florian Bentzinger, Marie‐Claude Sincennes, Michael A. Rudnicki. Satellite Cells and Skeletal Muscle Regeneration. Compr Physiol 2015, 5: 1027-1059. doi: 10.1002/cphy.c140068