Hanyang Med Rev.  2015 Nov;35(4):222-228. 10.7599/hmr.2015.35.4.222.

Recent Advances in Skeletal Muscle Stem Cells for Duchenne Muscular Dystrophy Treatment

Affiliations
  • 1Department of Surgery, Hanyang University College of Medicine, Seoul, Korea. jmj1103@hanyang.ac.kr

Abstract

Muscle stem cells, which are known as satellite cells have heterogeneous components of committed myogenic progenitors, non-committed satellite cells, and mesenchymal stem cells. This distinguishing organization of self-renewal and differentiation capacities encourages the remarkable regenerative ability of skeletal muscles. Lately it has been proved that the satellite cell is the derivation of muscle regeneration and with the self-renew function, it roles as a true muscle stem cell. Therefore, stem cell therapy using satellite cells is considered to be ideal therapy for muscular dystrophies, which is deficient in specific muscle protein and causes muscle degeneration. Especially, Duchenne Muscular Dystrophy (DMD), which is caused by mutations at the dystrophin gene, has been targeted by much research. In this article the satellite cell characteristics, regulation of cell function, and stem cell therapy for DMD and the present progressive clinical trials will be reviewed.

Keyword

Satellite Cells, Skeletal Muscle; Muscular Dystrophy, Duchenne; Dystrophin; Clinical Trial

MeSH Terms

Dystrophin
Mesenchymal Stromal Cells
Muscle Proteins
Muscle, Skeletal*
Muscular Dystrophies
Muscular Dystrophy, Duchenne*
Regeneration
Satellite Cells, Skeletal Muscle
Stem Cells*
Dystrophin
Muscle Proteins

Figure

  • Fig. 1 The self-renewal and differentiation capacity of satellite cells. Activated satellite cells are differentiated into myoblasts and then fuse with each other to create de novo myofibers. In de novo myofibers, nucleuses are centralized.

  • Fig. 2 Dystrophin (red) is not expressed in mdx mouse. (A) C57BL/10ScSn control mouse. (B) C57BL/10ScSn-Dmd.


Cited by  1 articles

New Horizons in Stem Cell Research
Dongho Choi
Hanyang Med Rev. 2015;35(4):187-189.    doi: 10.7599/hmr.2015.35.4.187.


Reference

1. Snow MH. Myogenic cell formation in regenerating rat skeletal muscle injured by mincing. II. An autoradiographic study. Anat Rec. 1977; 188:201–217.
Article
2. Collins CA, Olsen I, Zammit PS, Heslop L, Petrie A, Partridge TA, et al. Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell. 2005; 122:289–301.
Article
3. Kuang S, Kuroda K, Le Grand F, Rudnicki MA. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell. 2007; 129:999–1010.
Article
4. Emery AE. The muscular dystrophies. Lancet. 2002; 359:687–695.
Article
5. Cossu G, Sampaolesi M. New therapies for duchenne muscular dystrophy: challenges, prospects and clinical trials. Trends Mol Med. 2007; 13:520–526.
Article
6. Mauro A. Satellite cell of skeletal muscle fibers. J Biophys Biochem Cytol. 1961; 9:493–495.
Article
7. Kuang S, Rudnicki MA. The emerging biology of satellite cells and their therapeutic potential. Trends Mol Med. 2008; 14:82–91.
Article
8. Bischoff R. Regeneration of single skeletal muscle fibers in vitro. Anat Rec. 1975; 182:215–235.
Article
9. Charge SB, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiol Rev. 2004; 84:209–238.
Article
10. Nagata Y, Partridge TA, Matsuda R, Zammit PS. Entry of muscle satellite cells into the cell cycle requires sphingolipid signaling. J Cell Biol. 2006; 174:245–253.
Article
11. Wozniak AC, Anderson JE. Nitric oxide-dependence of satellite stem cell activation and quiescence on normal skeletal muscle fibers. Dev Dyn. 2007; 236:240–250.
Article
12. Pisconti A, Brunelli S, Di Padova M, De Palma C, Deponti D, Baesso S, et al. Follistatin induction by nitric oxide through cyclic GMP: a tightly regulated signaling pathway that controls myoblast fusion. J Cell Biol. 2006; 172:233–244.
Article
13. Jones NC, Tyner KJ, Nibarger L, Stanley HM, Cornelison DD, Fedorov YV, et al. The p38alpha/beta MAPK functions as a molecular switch to activate the quiescent satellite cell. J Cell Biol. 2005; 169:105–116.
Article
14. Moss FP, Leblond CP. Satellite cells as the source of nuclei in muscles of growing rats. Anat Rec. 1971; 170:421–435.
Article
15. Fuchs E, Tumbar T, Guasch G. Socializing with the neighbors: stem cells and their niche. Cell. 2004; 116:769–778.
16. Kuang S, Gillespie MA, Rudnicki MA. Niche regulation of muscle satellite cell self-renewal and differentiation. Cell Stem Cell. 2008; 2:22–31.
Article
17. Christov C, Chretien F, Abou-Khalil R, Bassez G, Vallet G, Authier FJ, et al. Muscle satellite cells and endothelial cells: close neighbors and privileged partners. Mol Biol Cell. 2007; 18:1397–1409.
Article
18. Quarta M, Rando TA. Mimicking the niche: cytokines expand muscle stem cells. Cell Res. 2015; 25:761–762.
Article
19. Brack AS, Rando TA. Tissue-specific stem cells: lessons from the skeletal muscle satellite cell. Cell Stem Cell. 2012; 10:504–514.
Article
20. 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. 2012; 30:232–242.
Article
21. Conboy IM, Rando TA. The regulation of Notch signaling controls satellite cell activation and cell fate determination in postnatal myogenesis. Dev Cell. 2002; 3:397–409.
Article
22. Abou-Khalil R, Brack AS. Muscle stem cells and reversible quiescence: the role of sprouty. Cell Cycle. 2010; 9:2575–2580.
Article
23. McCroskery S, Thomas M, Maxwell L, Sharma M, Kambadur R. Myostatin negatively regulates satellite cell activation and self-renewal. J Cell Biol. 2003; 162:1135–1147.
Article
24. Kitamoto T, Hanaoka K. Notch3 null mutation in mice causes muscle hyperplasia by repetitive muscle regeneration. Stem Cells. 2010; 28:2205–2216.
Article
25. Shea KL, Xiang W, LaPorta VS, Licht JD, Keller C, Basson MA, et al. Sprouty1 regulates reversible quiescence of a self-renewing adult muscle stem cell pool during regeneration. Cell Stem Cell. 2010; 6:117–129.
Article
26. Petersen PH, Zou K, Krauss S, Zhong W. Continuing role for mouse numb and numbl in maintaining progenitor cells during cortical neurogenesis. Nat Neurosci. 2004; 7:803–811.
Article
27. Church JC. Satellite cells and myogenesis; a study in the fruit-bat web. J Anat. 1969; 105:419–438.
28. Brack AS, Rando TA. Intrinsic changes and extrinsic influences of myogenic stem cell function during aging. Stem Cell Rev. 2007; 3:226–237.
Article
29. McKinnell IW, Ishibashi J, Le Grand F, Punch VG, Addicks GC, Greenblatt JF, et al. Pax7 activates myogenic genes by recruitment of a histone methyltransferase complex. Nat Cell Biol. 2008; 10:77–84.
Article
30. Seale P, Sabourin LA, Girgis-Gabardo A, Mansouri A, Gruss P, Rudnicki MA. Pax7 is required for the specification of myogenic satellite cells. Cell. 2000; 102:777–786.
Article
31. Beauchamp JR, Heslop L, Yu DS, Tajbakhsh S, Kelly RG, Wernig A, et al. Expression of CD34 and Myf5 defines the majority of quiescent adult skeletal muscle satellite cells. J Cell Biol. 2000; 151:1221–1234.
Article
32. Collins CA, Gnocchi VF, White RB, Boldrin L, Perez-Ruiz A, Relaix F, et al. Integrated functions of Pax3 and Pax7 in the regulation of proliferation, cell size and myogenic differentiation. PLoS One. 2009; 4:e4475.
Article
33. Meech R, Gonzalez KN, Barro M, Gromova A, Zhuang L, Hulin JA, et al. Barx2 is expressed in satellite cells and is required for normal muscle growth and regeneration. Stem Cells. 2012; 30:253–265.
Article
34. Hollnagel A, Grund C, Franke WW, Arnold HH. The cell adhesion molecule M-cadherin is not essential for muscle development and regeneration. Mol Cell Biol. 2002; 22:4760–4770.
Article
35. Wozniak AC, Pilipowicz O, Yablonka-Reuveni Z, Greenway S, Craven S, Scott E, et al. C-Met expression and mechanical activation of satellite cells on cultured muscle fibers. J Histochem Cytochem. 2003; 51:1437–1445.
Article
36. Ozeki N, Lim M, Yao CC, Tolar M, Kramer RH. alpha7 integrin expressing human fetal myogenic progenitors have stem cell-like properties and are capable of osteogenic differentiation. Exp Cell Res. 2006; 312:4162–4180.
Article
37. Ieronimakis N, Balasundaram G, Rainey S, Srirangam K, Yablonka-Reuveni Z, Reyes M. Absence of CD34 on murine skeletal muscle satellite cells marks a reversible state of activation during acute injury. PLoS One. 2010; 5:e10920.
Article
38. Ratajczak MZ, Majka M, Kucia M, Drukala J, Pietrzkowski Z, Peiper S, et al. 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. 2003; 21:363–371.
Article
39. Cornelison DD, Filla MS, Stanley HM, Rapraeger AC, Olwin BB. Syndecan-3 and syndecan-4 specifically mark skeletal muscle satellite cells and are implicated in satellite cell maintenance and muscle regeneration. Dev Biol. 2001; 239:79–94.
Article
40. 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. 2009; 4:e5205.
Article
41. Yamaguchi M, Ogawa R, Watanabe Y, Uezumi A, Miyagoe-Suzuki Y, Tsujikawa K, et al. Calcitonin receptor and Odz4 are differently expressed in Pax7-positive cells during skeletal muscle regeneration. J Mol Histol. 2012; 43:581–587.
Article
42. Frock RL, Kudlow BA, Evans AM, Jameson SA, Hauschka SD, Kennedy BK. Lamin A/C and emerin are critical for skeletal muscle satellite cell differentiation. Genes Dev. 2006; 20:486–500.
Article
43. 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. 2000; 10:113–120.
Article
44. Manzur AY, Kinali M, Muntoni F. Update on the management of duchenne muscular dystrophy. Arch Dis Child. 2008; 93:986–990.
Article
45. Peault B, Rudnicki M, Torrente Y, Cossu G, Tremblay JP, Partridge T, et al. Stem and progenitor cells in skeletal muscle development, maintenance, and therapy. Mol Ther. 2007; 15:867–877.
Article
46. Guerette B, Asselin I, Skuk D, Entman M, Tremblay JP. Control of inflammatory damage by anti-LFA-1: increase success of myoblast transplantation. Cell Transplant. 1997; 6:101–107.
Article
47. Skuk D, Roy B, Goulet M, Tremblay JP. Successful myoblast transplantation in primates depends on appropriate cell delivery and induction of regeneration in the host muscle. Exp Neurol. 1999; 155:22–30.
Article
48. Partridge TA, Morgan JE, Coulton GR, Hoffman EP, Kunkel LM. Conversion of mdx myofibres from dystrophin-negative to -positive by injection of normal myoblasts. Nature. 1989; 337:176–179.
Article
49. Dezawa M, Ishikawa H, Itokazu Y, Yoshihara T, Hoshino M, Takeda S, et al. Bone marrow stromal cells generate muscle cells and repair muscle degeneration. Science. 2005; 309:314–317.
Article
50. Sampaolesi M, Blot S, D'Antona G, Granger N, Tonlorenzi R, Innocenzi A, et al. Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs. Nature. 2006; 444:574–579.
Article
Full Text Links
  • HMR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr