Stem Cell Aging in Skeletal Muscle Regeneration and Disease
Abstract
:1. Introduction
2. MuSCs
2.1. MuSC Quiescence and Differentiation
2.2. Cellular Interactions in the Maintenance of MuSCs
3. Aging in MuSCs
3.1. The Decline of Regenerative Capacity with Aging
3.2. Sarcopenia and MuSCs
3.3. Stem Cell Aging in Diseased Condition
4. Prospects for Clinical Application Utilizing MuSCs
4.1. Cell-Autonomous Rejuvenation Strategy
4.2. Non-Cell Autonomous Rejuvenation Strategy
4.3. Regenerative Strategy
5. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Janssen, I.; Heymsfield, S.B.; Wang, Z.; Ross, R. Skeletal muscle mass and distribution in 468 men and women aged 18–88 yr. J. Appl. Physiol. 2000, 89, 81–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, N.C.; Rudnicki, M.A. Satellite cells: The architects of skeletal muscle. Curr. Top. Dev. Biol. 2014, 107, 161–181. [Google Scholar] [CrossRef] [PubMed]
- Mauro, A. Satellitse cell of skeletal muscle fibers. J. Biophys. Biochem. Cytol. 1961, 9, 493–495. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.H.; Jin, P.; Duan, R.; Chen, E.H. Mechanisms of myoblast fusion during muscle development. Curr. Opin. Genet. Dev. 2015, 32, 162–170. [Google Scholar] [CrossRef] [Green Version]
- Schmidt, M.; Schüler, S.C.; Hüttner, S.S.; von Eyss, B.; von Maltzahn, J. Adult stem cells at work: Regenerating skeletal muscle. Cell. Mol. Life Sci. 2019, 76, 2559–2570. [Google Scholar] [CrossRef] [Green Version]
- Von Maltzahn, J.; Bentzinger, C.F.; Rudnicki, M.A. Characteristics of satellite cells and multipotent adult stem cells in the skeletal muscle. In Stem Cells and Cancer Stem Cells; Springer: Berlin, Germany, 2014; Volume 12, pp. 63–73. [Google Scholar]
- Fukada, S.-I.; Ma, Y.; Ohtani, T.; Watanabe, Y.; Murakami, S.; Yamaguchi, M. Isolation, characterization, and molecular regulation of muscle stem cells. Front. Physiol. 2013, 4. [Google Scholar] [CrossRef] [Green Version]
- Seale, P.; Sabourin, L.A.; Girgis-Gabardo, A.; Mansouri, A.; Gruss, P.; Rudnicki, M.A. Pax7 is required for the specification of myogenic satellite cells. Cell 2000, 102, 777–786. [Google Scholar] [CrossRef] [Green Version]
- Shavlakadze, T.; McGeachie, J.; Grounds, M.D. Delayed but excellent myogenic stem cell response of regenerating geriatric skeletal muscles in mice. Biogerontology 2010, 11, 363–376. [Google Scholar] [CrossRef]
- 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 2011, 138, 3647–3656. [Google Scholar] [CrossRef] [Green Version]
- Biressi, S.; Rando, T.A. Heterogeneity in the muscle satellite cell population. Semin. Cell Dev. Biol. 2010, 21, 845–854. [Google Scholar] [CrossRef] [Green Version]
- Gros, J.; Manceau, M.; Thomé, V.; Marcelle, C. A common somitic origin for embryonic muscle progenitors and satellite cells. Nature 2005, 435, 954–958. [Google Scholar] [CrossRef] [PubMed]
- Kuang, S.; Kuroda, K.; Le Grand, F.; Rudnicki, M.A. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell 2007, 129, 999–1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moss, F.P.; Leblond, C.P. Satellite cells as the source of nuclei in muscles of growing rats. Anat. Rec. 1971, 170, 421–435. [Google Scholar] [CrossRef] [PubMed]
- Shea, K.L.; Xiang, W.; LaPorta, V.S.; Licht, J.D.; Keller, C.; Basson, M.A.; Brack, A.S. Sprouty1 regulates reversible quiescence of a self-renewing adult muscle stem cell pool during regeneration. Cell Stem Cell 2010, 6, 117–129. [Google Scholar] [CrossRef] [Green Version]
- Bray, S.J. Notch signalling in context. Nat. Rev. Mol. Cell Biol. 2016, 17, 722–735. [Google Scholar] [CrossRef]
- Kitamoto, T.; Hanaoka, K. Notch3 null mutation in mice causes muscle hyperplasia by repetitive muscle regeneration. Stem Cells (Dayton, Ohio) 2010, 28, 2205–2216. [Google Scholar] [CrossRef]
- Zalc, A.; Hayashi, S.; Auradé, F.; Bröhl, D.; Chang, T.; Mademtzoglou, D.; Mourikis, P.; Yao, Z.; Cao, Y.; Birchmeier, C.; et al. Antagonistic regulation of p57kip2 by Hes/Hey downstream of Notch signaling and muscle regulatory factors regulates skeletal muscle growth arrest. Development (Cambridge, England) 2014, 141, 2780–2790. [Google Scholar] [CrossRef] [Green Version]
- Mourikis, P.; Tajbakhsh, S. Distinct contextual roles for Notch signalling in skeletal muscle stem cells. BMC Dev. Biol. 2014, 14, 2. [Google Scholar] [CrossRef] [Green Version]
- Bjornson, C.R.R.; Cheung, T.H.; Liu, L.; Tripathi, P.V.; Steeper, K.M.; Rando, T.A. Notch signaling is necessary to maintain quiescence in adult muscle stem cells. Stem Cells 2012, 30, 232–242. [Google Scholar] [CrossRef] [Green Version]
- 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 2012, 30, 243–252. [Google Scholar] [CrossRef]
- Shen, H.; McElhinny, A.S.; Cao, Y.; Gao, P.; Liu, J.; Bronson, R.; Griffin, J.D.; Wu, L. The notch coactivator, MAML1, functions as a novel coactivator for MEF2C-mediated transcription and is required for normal myogenesis. Genes Dev. 2006, 20, 675–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baghdadi, M.B.; Firmino, J.; Soni, K.; Evano, B.; Di Girolamo, D.; Mourikis, P.; Castel, D.; Tajbakhsh, S. Notch-induced MiR-708 antagonizes satellite cell migration and maintains quiescence. Cell Stem Cell 2018, 23, 859–868.e5. [Google Scholar] [CrossRef] [PubMed]
- Gopinath, S.D.; Webb, A.E.; Brunet, A.; Rando, T.A. FOXO3 promotes quiescence in adult muscle stem cells during the process of self-renewal. Stem Cell Rep. 2014, 2, 414–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, B.; Shan, T. The role of satellite and other functional cell types in muscle repair and regeneration. J. Muscle Res. Cell Motil. 2019, 40, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Cornelison, D.D.W. Context matters: In vivo and in vitro influences on muscle satellite cell activity. J. Cell. Biochem. 2008, 105, 663–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wosczyna, M.N.; Rando, T.A. A muscle stem cell support group: Coordinated cellular responses in muscle regeneration. Dev. Cell 2018, 46, 135–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abou-Khalil, R.; Le Grand, F.; Pallafacchina, G.; Valable, S.; Authier, F.-J.; Rudnicki, M.A.; Gherardi, R.K.; Germain, S.; Chretien, F.; Sotiropoulos, A.; et al. Autocrine and paracrine angiopoietin 1/Tie-2 signaling promotes muscle satellite cell self-renewal. Cell Stem Cell 2009, 5, 298–309. [Google Scholar] [CrossRef] [Green Version]
- Chen, S.-E.; Gerken, E.; Zhang, Y.; Zhan, M.; Mohan, R.K.; Li, A.S.; Reid, M.B.; Li, Y.-P. Role of TNF-[1] signaling in regeneration of cardiotoxin-injured muscle. Am. J. Physiol. Cell Physiol. 2005, 289, C1179–C1187. [Google Scholar] [CrossRef]
- Chen, S.-E.; Jin, B.; Li, Y.-P. TNF-alpha regulates myogenesis and muscle regeneration by activating p38 MAPK. Am. J. Physiol. Cell Physiol. 2007, 292, C1660–C1671. [Google Scholar] [CrossRef]
- Mourkioti, F.; Rosenthal, N. IGF-1, inflammation and stem cells: Interactions during muscle regeneration. Trends Immunol. 2005, 26, 535–542. [Google Scholar] [CrossRef]
- Liu, L.; Cheung, T.H.; Charville, G.W.; Hurgo, B.M.C.; Leavitt, T.; Shih, J.; Brunet, A.; Rando, T.A. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep. 2013, 4, 189–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blum, R.; Vethantham, V.; Bowman, C.; Rudnicki, M.; Dynlacht, B.D. Genome-wide identification of enhancers in skeletal muscle: The role of MyoD1. Genes Dev. 2012, 26, 2763–2779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, Y.; Yao, Z.; Sarkar, D.; Lawrence, M.; Sanchez, G.J.; Parker, M.H.; MacQuarrie, K.L.; Davison, J.; Morgan, M.T.; Ruzzo, W.L.; et al. Genome-wide MyoD binding in skeletal muscle cells: A potential for broad cellular reprogramming. Dev. Cell 2010, 18, 662–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chargé, S.B.P.; Rudnicki, M.A. Cellular and molecular regulation of muscle regeneration. Physiol. Rev. 2004, 84, 209–238. [Google Scholar] [CrossRef] [PubMed]
- Tidball, J.G.; Villalta, S.A. Regulatory interactions between muscle and the immune system during muscle regeneration. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2010, 298, R1173–R1187. [Google Scholar] [CrossRef] [Green Version]
- Saclier, M.; Cuvellier, S.; Magnan, M.; Mounier, R.; Chazaud, B. Monocyte/macrophage interactions with myogenic precursor cells during skeletal muscle regeneration. FEBS J. 2013, 280, 4118–4130. [Google Scholar] [CrossRef]
- Heredia, J.E.; Mukundan, L.; Chen, F.M.; Mueller, A.A.; Deo, R.C.; Locksley, R.M.; Rando, T.A.; Chawla, A. Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell 2013, 153, 376–388. [Google Scholar] [CrossRef] [Green Version]
- Burzyn, D.; Kuswanto, W.; Kolodin, D.; Shadrach, J.L.; Cerletti, M.; Jang, Y.; Sefik, E.; Tan, T.G.; Wagers, A.J.; Benoist, C.; et al. A special population of regulatory T cells potentiates muscle repair. Cell 2013, 155, 1282–1295. [Google Scholar] [CrossRef] [Green Version]
- Joe, A.W.B.; Yi, L.; Natarajan, A.; Le Grand, F.; So, L.; Wang, J.; Rudnicki, M.A.; Rossi, F.M.V. Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nat. Cell Biol. 2010, 12, 153–163. [Google Scholar] [CrossRef] [Green Version]
- Uezumi, A.; Fukada, S.; Yamamoto, N.; Ikemoto-Uezumi, M.; Nakatani, M.; Morita, M.; Yamaguchi, A.; Yamada, H.; Nishino, I.; Hamada, Y.; et al. Identification and characterization of PDGFRα+ mesenchymal progenitors in human skeletal muscle. Cell Death Dis. 2014, 5, e1186. [Google Scholar] [CrossRef] [Green Version]
- Uezumi, A.; Ito, T.; Morikawa, D.; Shimizu, N.; Yoneda, T.; Segawa, M.; Yamaguchi, M.; Ogawa, R.; Matev, M.M.; Miyagoe-Suzuki, Y.; et al. Fibrosis and adipogenesis originate from a common mesenchymal progenitor in skeletal muscle. J. Cell Sci. 2011, 124, 3654–3664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lemos, D.R.; Paylor, B.; Chang, C.; Sampaio, A.; Underhill, T.M.; Rossi, F.M.V. Functionally convergent white adipogenic progenitors of different lineages participate in a diffused system supporting tissue regeneration. Stem Cells 2012, 30, 1152–1162. [Google Scholar] [CrossRef]
- Liu, N.; Garry, G.A.; Li, S.; Bezprozvannaya, S.; Sanchez-Ortiz, E.; Chen, B.; Shelton, J.M.; Jaichander, P.; Bassel-Duby, R.; Olson, E.N. A Twist2-dependent progenitor cell contributes to adult skeletal muscle. Nat. Cell Biol. 2017, 19, 202–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pannérec, A.; Formicola, L.; Besson, V.; Marazzi, G.; Sassoon, D.A. Defining skeletal muscle resident progenitors and their cell fate potentials. Development 2013, 140, 2879–2891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Latroche, C.; Weiss-Gayet, M.; Muller, L.; Gitiaux, C.; Leblanc, P.; Liot, S.; Ben-Larbi, S.; Abou-Khalil, R.; Verger, N.; Bardot, P.; et al. Coupling between myogenesis and angiogenesis during skeletal muscle regeneration is stimulated by restorative macrophages. Stem Cell Rep. 2017, 9, 2018–2033. [Google Scholar] [CrossRef] [Green Version]
- Christov, C.; Chrétien, F.; Abou-Khalil, R.; Bassez, G.; Vallet, G.; Authier, F.-J.; Bassaglia, Y.; Shinin, V.; Tajbakhsh, S.; Chazaud, B.; et al. Muscle satellite cells and endothelial cells: Close neighbors and privileged partners. Mol. Biol. Cell 2007, 18, 1397–1409. [Google Scholar] [CrossRef] [Green Version]
- Tatsumi, R.; Anderson, J.E.; Nevoret, C.J.; Halevy, O.; Allen, R.E. HGF/SF is present in normal adult skeletal muscle and is capable of activating satellite cells. Dev. Biol. 1998, 194, 114–128. [Google Scholar] [CrossRef] [Green Version]
- Bryan, B.A.; Walshe, T.E.; Mitchell, D.C.; Havumaki, J.S.; Saint-Geniez, M.; Maharaj, A.S.; Maldonado, A.E.; D’Amore, P.A. Coordinated vascular endothelial growth factor expression and signaling during skeletal myogenic differentiation. Mol. Biol. Cell 2008, 19, 994–1006. [Google Scholar] [CrossRef] [Green Version]
- Verma, M.; Asakura, Y.; Murakonda, B.S.R.; Pengo, T.; Latroche, C.; Chazaud, B.; McLoon, L.K.; Asakura, A. Muscle satellite cell cross-talk with a vascular niche maintains quiescence via VEGF and notch signaling. Cell Stem Cell 2018, 23, 530–543.e9. [Google Scholar] [CrossRef] [Green Version]
- Mathew, S.J.; Hansen, J.M.; Merrell, A.J.; Murphy, M.M.; Lawson, J.A.; Hutcheson, D.A.; Hansen, M.S.; Angus-Hill, M.; Kardon, G. Connective tissue fibroblasts and Tcf4 regulate myogenesis. Development 2011, 138, 371–384. [Google Scholar] [CrossRef] [Green Version]
- Murphy, M.M.; Lawson, J.A.; Mathew, S.J.; Hutcheson, D.A.; Kardon, G. Satellite cells, connective tissue fibroblasts and their interactions are crucial for muscle regeneration. Development 2011, 138, 3625–3637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamazaki, T.; Mukouyama, Y.-S. Tissue specific origin, development, and pathological perspectives of pericytes. Front. Cardiovasc. Med. 2018, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kostallari, E.; Baba-Amer, Y.; Alonso-Martin, S.; Ngoh, P.; Relaix, F.; Lafuste, P.; Gherardi, R.K. Pericytes in the myovascular niche promote post-natal myofiber growth and satellite cell quiescence. Development 2015, 142, 1242–1253. [Google Scholar] [CrossRef] [Green Version]
- Díaz-Manera, J.; Gallardo, E.; de Luna, N.; Navas, M.; Soria, L.; Garibaldi, M.; Rojas-García, R.; Tonlorenzi, R.; Cossu, G.; Illa, I. The increase of pericyte population in human neuromuscular disorders supports their role in muscle regeneration in vivo. J. Pathol. 2012, 228, 544–553. [Google Scholar] [CrossRef] [PubMed]
- Welle, S. Cellular and molecular basis of age-related sarcopenia. Can. J. Appl. Physiol. 2002, 27, 19–41. [Google Scholar] [CrossRef] [PubMed]
- Grounds, M.D. Age-associated changes in the response of skeletal muscle cells to exercise and regeneration. Ann. N.Y. Acad. Sci. 1998, 854, 78–91. [Google Scholar] [CrossRef] [PubMed]
- Sadeh, M. Effects of aging on skeletal muscle regeneration. J. Neurol. Sci. 1988, 87, 67–74. [Google Scholar] [CrossRef]
- Schultz, E.; Lipton, B.H. Skeletal muscle satellite cells: Changes in proliferation potential as a function of age. Mech. Ageing Dev. 1982, 20, 377–383. [Google Scholar] [CrossRef]
- Carlson, M.E.; Suetta, C.; Conboy, M.J.; Aagaard, P.; Mackey, A.; Kjaer, M.; Conboy, I. Molecular aging and rejuvenation of human muscle stem cells. EMBO Mol. Med. 2009, 1, 381–391. [Google Scholar] [CrossRef] [Green Version]
- Jejurikar, S.S.; Henkelman, E.A.; Cederna, P.S.; Marcelo, C.L.; Urbanchek, M.G.; Kuzon, W.M. Aging increases the susceptibility of skeletal muscle derived satellite cells to apoptosis. Exp. Gerontol. 2006, 41, 828–836. [Google Scholar] [CrossRef]
- Taylor-Jones, J.M.; McGehee, R.E.; Rando, T.A.; Lecka-Czernik, B.; Lipschitz, D.A.; Peterson, C.A. Activation of an adipogenic program in adult myoblasts with age. Mech. Ageing Dev. 2002, 123, 649–661. [Google Scholar] [CrossRef]
- Carlson, B.M.; Faulkner, J.A. Muscle transplantation between young and old rats: Age of host determines recovery. Am. J. Physiol. Cell Physiol. 1989, 256, C1262–C1266. [Google Scholar] [CrossRef]
- Kawabe, Y.-I.; Wang, Y.X.; McKinnell, I.W.; Bedford, M.T.; Rudnicki, M.A. Carm1 regulates Pax7 transcriptional activity through MLL1/2 recruitment during asymmetric satellite stem cell divisions. Cell Stem Cell 2012, 11, 333–345. [Google Scholar] [CrossRef] [Green Version]
- Chang, N.C.; Sincennes, M.-C.; Chevalier, F.P.; Brun, C.E.; Lacaria, M.; Segalés, J.; Muñoz-Cánoves, P.; Ming, H.; Rudnicki, M.A. The dystrophin glycoprotein complex regulates the epigenetic activation of muscle stem cell commitment. Cell Stem Cell 2018, 22, 755–768.e6. [Google Scholar] [CrossRef] [Green Version]
- Bernet, J.D.; Doles, J.D.; Hall, J.K.; Tanaka, K.K.; Carter, T.A.; Olwin, B.B. P38 MAPK signaling underlies a cell-autonomous loss of stem cell self-renewal in skeletal muscle of aged mice. Nat. Med. 2014, 20, 265–271. [Google Scholar] [CrossRef] [PubMed]
- Conboy, I.M.; Conboy, M.J.; Smythe, G.M.; Rando, T.A. Notch-mediated restoration of regenerative potential to aged muscle. Science 2003, 302, 1575–1577. [Google Scholar] [CrossRef] [PubMed]
- Shefer, G.; Van de Mark, D.P.; Richardson, J.B.; Yablonka-Reuveni, Z. Satellite-cell pool size does matter: Defining the myogenic potency of aging skeletal muscle. Dev. Biol. 2006, 294, 50–66. [Google Scholar] [CrossRef] [Green Version]
- Collins, C.A.; Zammit, P.S.; Ruiz, A.P.; Morgan, J.E.; Partridge, T.A. A population of myogenic stem cells that survives skeletal muscle aging. Stem Cells 2007, 25, 885–894. [Google Scholar] [CrossRef] [PubMed]
- Motohashi, N.; Asakura, A. Muscle satellite cell heterogeneity and self-renewal. Front. Cell Dev. Biol. 2014, 2. [Google Scholar] [CrossRef]
- Shefer, G.; Rauner, G.; Yablonka-Reuveni, Z.; Benayahu, D. Reduced satellite cell numbers and myogenic capacity in aging can be alleviated by endurance exercise. PLoS ONE 2010, 5, e13307. [Google Scholar] [CrossRef] [Green Version]
- Roth, S.M.; Martel, G.F.; Ivey, F.M.; Lemmer, J.T.; Metter, E.J.; Hurley, B.F.; Rogers, M.A. Skeletal muscle satellite cell populations in healthy young and older men and women. Anat. Rec. 2000, 260, 351–358. [Google Scholar] [CrossRef]
- Wagers, A.J.; Conboy, I.M. Cellular and molecular signatures of muscle regeneration: Current concepts and controversies in adult myogenesis. Cell 2005, 122, 659–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van der Meer, S.F.T.; Jaspers, R.T.; Jones, D.A.; Degens, H. Time-course of changes in the myonuclear domain during denervation in young-adult and old rat gastrocnemius muscle. Muscle Nerve 2011, 43, 212–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carlson, M.E.; Hsu, M.; Conboy, I.M. Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature 2008, 454, 528–532. [Google Scholar] [CrossRef] [Green Version]
- Brack, A.S.; Conboy, M.J.; Roy, S.; Lee, M.; Kuo, C.J.; Keller, C.; Rando, T.A. Increased wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 2007, 317, 807–810. [Google Scholar] [CrossRef] [PubMed]
- Sousa-Victor, P.; Gutarra, S.; García-Prat, L.; Rodriguez-Ubreva, J.; Ortet, L.; Ruiz-Bonilla, V.; Jardí, M.; Ballestar, E.; González, S.; Serrano, A.L.; et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 2014, 506, 316–321. [Google Scholar] [CrossRef] [PubMed]
- Segalés, J.; Perdiguero, E.; Muñoz-Cánoves, P. Regulation of muscle stem cell functions: A focus on the p38 mapk signaling pathway. Front. Cell Dev. Biol. 2016, 4, 91. [Google Scholar] [CrossRef] [Green Version]
- Price, F.D.; von Maltzahn, J.; Bentzinger, C.F.; Dumont, N.A.; Yin, H.; Chang, N.C.; Wilson, D.H.; Frenette, J.; Rudnicki, M.A. Inhibition of JAK-STAT signaling stimulates adult satellite cell function. Nat. Med. 2014, 20, 1174–1181. [Google Scholar] [CrossRef] [Green Version]
- Tierney, M.T.; Aydogdu, T.; Sala, D.; Malecova, B.; Gatto, S.; Puri, P.L.; Latella, L.; Sacco, A. STAT3 signaling controls satellite cell expansion and skeletal muscle repair. Nat. Med. 2014, 20, 1182–1186. [Google Scholar] [CrossRef] [Green Version]
- Chakkalakal, J.V.; Jones, K.M.; Basson, M.A.; Brack, A.S. The aged niche disrupts muscle stem cell quiescence. Nature 2012, 490, 355–360. [Google Scholar] [CrossRef] [Green Version]
- Sousa-Victor, P.; Perdiguero, E.; Muñoz-Cánoves, P. Geroconversion of aged muscle stem cells under regenerative pressure. Cell Cycle 2014, 13, 3183–3190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cosgrove, B.D.; Gilbert, P.M.; Porpiglia, E.; Mourkioti, F.; Lee, S.P.; Corbel, S.Y.; Llewellyn, M.E.; Delp, S.L.; Blau, H.M. Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat. Med. 2014, 20, 255–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Doherty, T.J. Invited review: Aging and sarcopenia. J. Appl. Physiol. 2003, 95, 1717–1727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verdijk, L.B.; Snijders, T.; Drost, M.; Delhaas, T.; Kadi, F.; van Loon, L.J.C. Satellite cells in human skeletal muscle; from birth to old age. Age (Dordr) 2014, 36, 545–547. [Google Scholar] [CrossRef] [Green Version]
- Fry, C.S.; Lee, J.D.; Mula, J.; Kirby, T.J.; Jackson, J.R.; Liu, F.; Yang, L.; Mendias, C.L.; Dupont-Versteegden, E.E.; McCarthy, J.J.; et al. Inducible depletion of satellite cells in adult, sedentary mice impairs muscle regenerative capacity without affecting sarcopenia. Nat. Med. 2015, 21, 76–80. [Google Scholar] [CrossRef]
- Keefe, A.C.; Lawson, J.A.; Flygare, S.D.; Fox, Z.D.; Colasanto, M.P.; Mathew, S.J.; Yandell, M.; Kardon, G. Muscle stem cells contribute to myofibres in sedentary adult mice. Nat. Commun. 2015, 6, 7087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brack, A.S.; Bildsoe, H.; Hughes, S.M. Evidence that satellite cell decrement contributes to preferential decline in nuclear number from large fibres during murine age-related muscle atrophy. J. Cell Sci. 2005, 118, 4813–4821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hikida, R.S. Aging changes in satellite cells and their functions. Curr. Aging Sci. 2011, 4, 279–297. [Google Scholar] [CrossRef]
- Sousa-Victor, P.; Muñoz-Cánoves, P. Regenerative decline of stem cells in sarcopenia. Mol. Asp. Med. 2016, 50, 109–117. [Google Scholar] [CrossRef]
- Brack, A.S.; Muñoz-Cánoves, P. The ins and outs of muscle stem cell aging. Skelet. Muscle 2016, 6, 1. [Google Scholar] [CrossRef] [Green Version]
- Snijders, T.; Nederveen, J.P.; McKay, B.R.; Joanisse, S.; Verdijk, L.B.; van Loon, L.J.C.; Parise, G. Satellite cells in human skeletal muscle plasticity. Front. Physiol. 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grounds, M.D. Therapies for sarcopenia and regeneration of old skeletal muscles. Bioarchitecture 2014, 4, 81–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alsharidah, M.; Lazarus, N.R.; George, T.E.; Agley, C.C.; Velloso, C.P.; Harridge, S.D.R. Primary human muscle precursor cells obtained from young and old donors produce similar proliferative, differentiation and senescent profiles in culture. Aging Cell 2013, 12, 333–344. [Google Scholar] [CrossRef] [PubMed]
- Barberi, L.; Scicchitano, B.M.; De Rossi, M.; Bigot, A.; Duguez, S.; Wielgosik, A.; Stewart, C.; McPhee, J.; Conte, M.; Narici, M.; et al. Age-dependent alteration in muscle regeneration: The critical role of tissue niche. Biogerontology 2013, 14, 273–292. [Google Scholar] [CrossRef] [Green Version]
- Fulle, S.; Sancilio, S.; Mancinelli, R.; Gatta, V.; Di Pietro, R. Dual role of the caspase enzymes in satellite cells from aged and young subjects. Cell Death Dis. 2013, 4, e955. [Google Scholar] [CrossRef] [Green Version]
- Franco, I.; Johansson, A.; Olsson, K.; Vrtačnik, P.; Lundin, P.; Helgadottir, H.T.; Larsson, M.; Revêchon, G.; Bosia, C.; Pagnani, A.; et al. Somatic mutagenesis in satellite cells associates with human skeletal muscle aging. Nat. Commun. 2018, 9, 800. [Google Scholar] [CrossRef]
- Powers, S.K.; Lynch, G.S.; Murphy, K.T.; Reid, M.B.; Zijdewind, I. Disease-induced skeletal muscle atrophy and fatigue. Med. Sci. Sports Exerc. 2016, 48, 2307–2319. [Google Scholar] [CrossRef]
- Brooks, N.; Myburgh, K. Skeletal muscle wasting with disuse atrophy is multi-dimensional: The response and interaction of myonuclei, satellite cells and signaling pathways. Front. Physiol. 2014, 5. [Google Scholar] [CrossRef]
- Matsuba, Y.; Goto, K.; Morioka, S.; Naito, T.; Akema, T.; Hashimoto, N.; Sugiura, T.; Ohira, Y.; Beppu, M.; Yoshioka, T. Gravitational unloading inhibits the regenerative potential of atrophied soleus muscle in mice. Acta Physiol. 2009, 196, 329–339. [Google Scholar] [CrossRef]
- Suetta, C.; Frandsen, U.; Mackey, A.L.; Jensen, L.; Hvid, L.G.; Bayer, M.L.; Petersson, S.J.; Schrøder, H.D.; Andersen, J.L.; Aagaard, P.; et al. Ageing is associated with diminished muscle re-growth and myogenic precursor cell expansion early after immobility-induced atrophy in human skeletal muscle. J. Physiol. 2013, 591, 3789–3804. [Google Scholar] [CrossRef]
- Verdijk, L.B.; Dirks, M.L.; Snijders, T.; Prompers, J.J.; Beelen, M.; Jonkers, R.A.M.; Thijssen, D.H.J.; Hopman, M.T.E.; Van Loon, L.J.C. Reduced satellite cell numbers with spinal cord injury and aging in humans. Med. Sci. Sports Exerc. 2012, 44, 2322–2330. [Google Scholar] [CrossRef] [PubMed]
- Darr, K.C.; Schultz, E. Hindlimb suspension suppresses muscle growth and satellite cell proliferation. J. Appl. Physiol. 1989, 67, 1827–1834. [Google Scholar] [CrossRef] [PubMed]
- Mitchell, P.O.; Pavlath, G.K. Skeletal muscle atrophy leads to loss and dysfunction of muscle precursor cells. Am. J. Physiol. Cell Physiol. 2004, 287, C1753–C1762. [Google Scholar] [CrossRef] [Green Version]
- Mozdziak, P.E.; Pulvermacher, P.M.; Schultz, E. Muscle regeneration during hindlimb unloading results in a reduction in muscle size after reloading. J. Appl. Physiol. 2001, 91, 183–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hara, M.; Yuasa, S.; Shimoji, K.; Onizuka, T.; Hayashiji, N.; Ohno, Y.; Arai, T.; Hattori, F.; Kaneda, R.; Kimura, K.; et al. G-CSF influences mouse skeletal muscle development and regeneration by stimulating myoblast proliferation. J. Exp. Med. 2011, 208, 715–727. [Google Scholar] [CrossRef] [Green Version]
- Emery, A.E.H. The muscular dystrophies. Lancet 2002, 359, 687–695. [Google Scholar] [CrossRef]
- Bushby, K.; Finkel, R.; Birnkrant, D.J.; Case, L.E.; Clemens, P.R.; Cripe, L.; Kaul, A.; Kinnett, K.; McDonald, C.; Pandya, S.; et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: Diagnosis, and pharmacological and psychosocial management. Lancet Neurol. 2010, 9, 77–93. [Google Scholar] [CrossRef]
- Hoffman, E.; Monaco, A.; Feener, C.; Kunkel, L. Conservation of the Duchenne muscular dystrophy gene in mice and humans. Science 1987, 238, 347–350. [Google Scholar] [CrossRef]
- Luz, M.A.M.; Marques, M.J.; Santo Neto, H. Impaired regeneration of dystrophin-deficient muscle fibers is caused by exhaustion of myogenic cells. Braz. J. Med Biol. Res. 2002, 35, 691–695. [Google Scholar] [CrossRef] [Green Version]
- Sacco, A.; Mourkioti, F.; Tran, R.; Choi, J.; Llewellyn, M.; Kraft, P.; Shkreli, M.; Delp, S.; Pomerantz, J.H.; Artandi, S.E.; et al. Short telomeres and stem cell exhaustion model duchenne muscular dystrophy in mdx/mTR mice. Cell 2010, 143, 1059–1071. [Google Scholar] [CrossRef] [Green Version]
- Tichy, E.D.; Sidibe, D.K.; Tierney, M.T.; Stec, M.J.; Sharifi-Sanjani, M.; Hosalkar, H.; Mubarak, S.; Johnson, F.B.; Sacco, A.; Mourkioti, F. Single stem cell imaging and analysis reveals telomere length differences in diseased human and mouse skeletal muscles. Stem Cell Rep. 2017, 9, 1328–1341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kottlors, M.; Kirschner, J. Elevated satellite cell number in Duchenne muscular dystrophy. Cell Tissue Res. 2010, 340, 541–548. [Google Scholar] [CrossRef] [PubMed]
- Chakkalakal, J.V.; Christensen, J.; Xiang, W.; Tierney, M.T.; Boscolo, F.S.; Sacco, A.; Brack, A.S. Early forming label-retaining muscle stem cells require p27kip1 for maintenance of the primitive state. Development 2014, 141, 1649–1659. [Google Scholar] [CrossRef] [Green Version]
- Dumont, N.A.; Wang, Y.X.; von Maltzahn, J.; Pasut, A.; Bentzinger, C.F.; Brun, C.E.; Rudnicki, M.A. Dystrophin expression in muscle stem cells regulates their polarity and asymmetric division. Nat. Med. 2015, 21, 1455–1463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Troy, A.; Cadwallader, A.B.; Fedorov, Y.; Tyner, K.; Tanaka, K.K.; Olwin, B.B. Coordination of satellite cell activation and self-renewal by par-complex-dependent asymmetric activation of p38α/β MAPK. Cell Stem Cell 2012, 11, 541–553. [Google Scholar] [CrossRef] [Green Version]
- Hayashiji, N.; Yuasa, S.; Miyagoe-Suzuki, Y.; Hara, M.; Ito, N.; Hashimoto, H.; Kusumoto, D.; Seki, T.; Tohyama, S.; Kodaira, M.; et al. G-CSF supports long-term muscle regeneration in mouse models of muscular dystrophy. Nat. Commun. 2015, 6, 6745. [Google Scholar] [CrossRef] [Green Version]
- Chang, N.C.; Chevalier, F.P.; Rudnicki, M.A. Satellite cells in muscular dystrophy – Lost in polarity. Trends Mol. Med. 2016, 22, 479–496. [Google Scholar] [CrossRef] [Green Version]
- Peay, H.L.; Fischer, R.; Tzeng, J.P.; Hesterlee, S.E.; Morris, C.; Martin, A.S.; Rensch, C.; Smith, E.; Ricotti, V.; Beaverson, K.; et al. Gene therapy as a potential therapeutic option for Duchenne muscular dystrophy: A qualitative preference study of patients and parents. Plos ONE 2019, 14, e0213649. [Google Scholar] [CrossRef]
- Mendell, J.R.; Rodino-Klapac, L.R. Duchenne muscular dystrophy: CRISPR/Cas9 treatment. Cell Res. 2016, 26, 513–514. [Google Scholar] [CrossRef] [Green Version]
- Reinig, A.M.; Mirzaei, S.; Berlau, D.J. Advances in the treatment of duchenne muscular dystrophy: New and emerging pharmacotherapies. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2017, 37, 492–499. [Google Scholar] [CrossRef]
- Ousterout, D.G.; Kabadi, A.M.; Thakore, P.I.; Majoros, W.H.; Reddy, T.E.; Gersbach, C.A. Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy. Nat. Commun. 2015, 6, 6244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, M.-J.; Chen, J.; Chen, F.; Liu, Q.; Sun, Y.; Yan, C.; Yang, T.; Bao, Y.; Hu, Y.-P. Rejuvenating strategies of tissue-specific stem cells for healthy aging. Aging Dis. 2019, 10, 871–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kitajima, Y.; Suzuki, N.; Nunomiya, A.; Osana, S.; Yoshioka, K.; Tashiro, Y.; Takahashi, R.; Ono, Y.; Aoki, M.; Nagatomi, R. The ubiquitin-proteasome system is indispensable for the maintenance of muscle stem cells. Stem Cell Rep. 2018, 11, 1523–1538. [Google Scholar] [CrossRef] [Green Version]
- Rufini, A.; Tucci, P.; Celardo, I.; Melino, G. Senescence and aging: The critical roles of p53. Oncogene 2013, 32, 5129–5143. [Google Scholar] [CrossRef]
- Stantzou, A.; Schirwis, E.; Swist, S.; Alonso-Martin, S.; Polydorou, I.; Zarrouki, F.; Mouisel, E.; Beley, C.; Julien, A.; Le Grand, F.; et al. BMP signaling regulates satellite cell-dependent postnatal muscle growth. Development (Cambridge, England) 2017, 144, 2737–2747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Flamini, V.; Ghadiali, R.S.; Antczak, P.; Rothwell, A.; Turnbull, J.E.; Pisconti, A. The satellite cell niche regulates the balance between myoblast differentiation and self-renewal via p53. Stem Cell Rep. 2018, 10, 970–983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hansen, M.; Rubinsztein, D.C.; Walker, D.W. Autophagy as a promoter of longevity: Insights from model organisms. Nat. Rev. Mol. Cell Biol. 2018, 19, 579–593. [Google Scholar] [CrossRef] [PubMed]
- Fiacco, E.; Castagnetti, F.; Bianconi, V.; Madaro, L.; De Bardi, M.; Nazio, F.; D’Amico, A.; Bertini, E.; Cecconi, F.; Puri, P.L.; et al. Autophagy regulates satellite cell ability to regenerate normal and dystrophic muscles. Cell Death Differ. 2016, 23, 1839–1849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, A.H.; Rando, T.A. Induction of autophagy supports the bioenergetic demands of quiescent muscle stem cell activation. EMBO J. 2014, 33, 2782–2797. [Google Scholar] [CrossRef] [Green Version]
- García-Prat, L.; Martínez-Vicente, M.; Perdiguero, E.; Ortet, L.; Rodríguez-Ubreva, J.; Rebollo, E.; Ruiz-Bonilla, V.; Gutarra, S.; Ballestar, E.; Serrano, A.L.; et al. Autophagy maintains stemness by preventing senescence. Nature 2016, 529, 37–42. [Google Scholar] [CrossRef]
- White, J.P.; Billin, A.N.; Campbell, M.E.; Russell, A.J.; Huffman, K.M.; Kraus, W.E. The AMPK/p27Kip1 Axis regulates autophagy/apoptosis decisions in aged skeletal muscle stem cells. Stem Cell Rep. 2018, 11, 425–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oh, J.; Lee, Y.D.; Wagers, A.J. Stem cell aging: Mechanisms, regulators and therapeutic opportunities. Nat. Med. 2014, 20, 870–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beerman, I.; Rossi, D.J. Epigenetic control of stem cell potential during homeostasis, aging, and disease. Cell Stem Cell 2015, 16, 613–625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hernando-Herraez, I.; Evano, B.; Stubbs, T.; Commere, P.-H.; Jan Bonder, M.; Clark, S.; Andrews, S.; Tajbakhsh, S.; Reik, W. Ageing affects DNA methylation drift and transcriptional cell-to-cell variability in mouse muscle stem cells. Nat. Commun. 2019, 10, 4361. [Google Scholar] [CrossRef] [Green Version]
- Schwörer, S.; Becker, F.; Feller, C.; Baig, A.H.; Köber, U.; Henze, H.; Kraus, J.M.; Xin, B.; Lechel, A.; Lipka, D.B.; et al. Epigenetic stress responses induce muscle stem-cell ageing by Hoxa9 developmental signals. Nature 2016, 540, 428–432. [Google Scholar] [CrossRef] [Green Version]
- Minetti, G.C.; Colussi, C.; Adami, R.; Serra, C.; Mozzetta, C.; Parente, V.; Fortuni, S.; Straino, S.; Sampaolesi, M.; Di Padova, M.; et al. Functional and morphological recovery of dystrophic muscles in mice treated with deacetylase inhibitors. Nat. Med. 2006, 12, 1147–1150. [Google Scholar] [CrossRef] [Green Version]
- Consalvi, S.; Mozzetta, C.; Bettica, P.; Germani, M.; Fiorentini, F.; Del Bene, F.; Rocchetti, M.; Leoni, F.; Monzani, V.; Mascagni, P.; et al. Preclinical studies in the mdx mouse model of duchenne muscular dystrophy with the histone deacetylase inhibitor givinostat. Mol. Med. 2013, 19, 79–87. [Google Scholar] [CrossRef]
- Colussi, C.; Gurtner, A.; Rosati, J.; Illi, B.; Ragone, G.; Piaggio, G.; Moggio, M.; Lamperti, C.; D’Angelo, G.; Clementi, E.; et al. Nitric oxide deficiency determines global chromatin changes in Duchenne muscular dystrophy. FASEB J. 2009, 23, 2131–2141. [Google Scholar] [CrossRef] [Green Version]
- Conboy, I.M.; Conboy, M.J.; Wagers, A.J.; Girma, E.R.; Weissman, I.L.; Rando, T.A. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 2005, 433, 760–764. [Google Scholar] [CrossRef]
- Morrissey, J.B.; Cheng, R.Y.; Davoudi, S.; Gilbert, P.M. Biomechanical origins of muscle stem cell signal transduction. J. Mol. Biol. 2016, 428, 1441–1454. [Google Scholar] [CrossRef]
- Sorensen, J.R.; Fuqua, J.D.; Deyhle, M.R.; Parmley, J.; Skousen, C.; Hancock, C.; Parcell, A.C.; Hyldahl, R.D. Preclinical characterization of the JAK/STAT inhibitor SGI-1252 on skeletal muscle function, morphology, and satellite cell content. Plos ONE 2018, 13, e0198611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, L.; Charville, G.W.; Cheung, T.H.; Yoo, B.; Santos, P.J.; Schroeder, M.; Rando, T.A. Impaired notch signaling leads to a decrease in p53 activity and mitotic catastrophe in aged muscle stem cells. Cell Stem cell 2018, 23, 544–556.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sinha, M.; Jang, Y.C.; Oh, J.; Khong, D.; Wu, E.Y.; Manohar, R.; Miller, C.; Regalado, S.G.; Loffredo, F.S.; Pancoast, J.R.; et al. Restoring systemic GDF11 levels reverses age-related dysfunction in mouse skeletal muscle. Science (New York, N.Y.) 2014, 344, 649–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jia, Z.; Nie, Y.; Yue, F.; Kong, Y.; Gu, L.; Gavin, T.P.; Liu, X.; Kuang, S. A requirement of Polo-like kinase 1 in murine embryonic myogenesis and adult muscle regeneration. eLife 2019, 8, e47097. [Google Scholar] [CrossRef] [PubMed]
- Blau, H.M.; Cosgrove, B.D.; Ho, A.T.V. The central role of muscle stem cells in regenerative failure with aging. Nat. Med. 2015, 21, 854–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scadden, D.T. Nice neighborhood: Emerging concepts of the stem cell niche. Cell 2014, 157, 41–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Evano, B.; Tajbakhsh, S. Skeletal muscle stem cells in comfort and stress. NPJ Regen. Med. 2018, 3, 24. [Google Scholar] [CrossRef]
- Lukjanenko, L.; Jung, M.J.; Hegde, N.; Perruisseau-Carrier, C.; Migliavacca, E.; Rozo, M.; Karaz, S.; Jacot, G.; Schmidt, M.; Li, L.; et al. Loss of fibronectin from the aged stem cell niche affects the regenerative capacity of skeletal muscle in mice. Nat. Med. 2016, 22, 897–905. [Google Scholar] [CrossRef] [Green Version]
- Oh, J.; Sinha, I.; Tan, K.Y.; Rosner, B.; Dreyfuss, J.M.; Gjata, O.; Tran, P.; Shoelson, S.E.; Wagers, A.J. Age-associated NF-κB signaling in myofibers alters the satellite cell niche and re-strains muscle stem cell function. Aging (Albany NY) 2016, 8, 2871–2896. [Google Scholar] [CrossRef] [Green Version]
- Carlson, M.E.; Conboy, M.J.; Hsu, M.; Barchas, L.; Jeong, J.; Agrawal, A.; Mikels, A.J.; Agrawal, S.; Schaffer, D.V.; Conboy, I.M. Relative roles of TGF-beta1 and Wnt in the systemic regulation and aging of satellite cell responses. Aging Cell 2009, 8, 676–689. [Google Scholar] [CrossRef]
- Elabd, C.; Cousin, W.; Upadhyayula, P.; Chen, R.Y.; Chooljian, M.S.; Li, J.; Kung, S.; Jiang, K.P.; Conboy, I.M. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat. Commun. 2014, 5, 4082. [Google Scholar] [CrossRef] [Green Version]
- Rozo, M.; Li, L.; Fan, C.-M. Targeting β1-integrin signaling enhances regeneration in aged and dystrophic muscle in mice. Nat. Med. 2016, 22, 889–896. [Google Scholar] [CrossRef] [Green Version]
- Feige, P.; Brun, C.E.; Ritso, M.; Rudnicki, M.A. Orienting muscle stem cells for regeneration in homeostasis, aging, and disease. Cell Stem Cell 2018, 23, 653–664. [Google Scholar] [CrossRef] [PubMed]
- Sun, C.; Serra, C.; Lee, G.; Wagner, K.R. Stem cell-based therapies for Duchenne muscular dystrophy. Exp. Neurol. 2020, 323, 113086. [Google Scholar] [CrossRef]
- Price, F.D.; Kuroda, K.; Rudnicki, M.A. Stem cell based therapies to treat muscular dystrophy. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2007, 1772, 272–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kodaka, Y.; Rabu, G.; Asakura, A. Skeletal muscle cell induction from pluripotent stem cells. Stem Cells Int. 2017, 2017, 1376151. [Google Scholar] [CrossRef] [PubMed]
- Miyagoe-Suzuki, Y.; Takeda, S.I. Skeletal muscle generated from induced pluripotent stem cells—Induction and application. World J. Stem Cells 2017, 9, 89–97. [Google Scholar] [CrossRef]
- Van der Wal, E.; Herrero-Hernandez, P.; Wan, R.; Broeders, M.; In ‘t Groen, S.L.M.; van Gestel, T.J.M.; van Ijcken, W.F.J.; Cheung, T.H.; van der Ploeg, A.T.; Schaaf, G.J.; et al. Large-scale expansion of human iPSC-derived skeletal muscle cells for disease modeling and cell-based therapeutic strategies. Stem Cell Rep. 2018, 10, 1975–1990. [Google Scholar] [CrossRef]
Intrinsic Rejuvenating Strategies of Aged MuSC | |||
---|---|---|---|
Target | Mechanism | Function | Reference |
Fgfr1 inhibitor SU5402 Spry1 overexpression | reducing FGF signaling | loss of quiescence, regenerative capacity | [81] |
Fibronection injection | rescue FAK signaling | proliferative and myogenic potential | [149] |
Sodium salicylate | inhibition of NF-κB signaling | regenerative capacity | [150] |
TS2/16 | activation of b1-integrin/FGFR | regenerative capacity | [153] |
Tyr AG 490 | inhibition of JAK/STAT | MuSC number; self-renewal; regenerative capacity | [79] |
5,15 diphenylporphrine | inhibition of JAK/STAT | MuSC number; self-renewal; regenerative capacity | [79] |
Systemic environment to reverse aging of MuSC | |||
Target | Mechanism | Rejuvenation on Function | Reference |
Frizzled-related protein 3 (sFRP3) incubation | suppression of Wnt signaling | proliferative potential; muscle regeneration | [76] |
Dickkopf-1 (Dkk1) injection | suppression of Wnt signaling | muscle regeneration | [76] |
TGF-beta receptor kinase inhibitor | attenuating TGFb signaling | regenerative potential | [151] |
Growth differentiation factor 11 (GDF11) injection | unknown | regenerative potential | [144] |
Oxytocin | activation of MAPK/ERK signaling | MuSC activation and proliferation; regenerative potential | [152] |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Yamakawa, H.; Kusumoto, D.; Hashimoto, H.; Yuasa, S. Stem Cell Aging in Skeletal Muscle Regeneration and Disease. Int. J. Mol. Sci. 2020, 21, 1830. https://doi.org/10.3390/ijms21051830
Yamakawa H, Kusumoto D, Hashimoto H, Yuasa S. Stem Cell Aging in Skeletal Muscle Regeneration and Disease. International Journal of Molecular Sciences. 2020; 21(5):1830. https://doi.org/10.3390/ijms21051830
Chicago/Turabian StyleYamakawa, Hiroyuki, Dai Kusumoto, Hisayuki Hashimoto, and Shinsuke Yuasa. 2020. "Stem Cell Aging in Skeletal Muscle Regeneration and Disease" International Journal of Molecular Sciences 21, no. 5: 1830. https://doi.org/10.3390/ijms21051830
APA StyleYamakawa, H., Kusumoto, D., Hashimoto, H., & Yuasa, S. (2020). Stem Cell Aging in Skeletal Muscle Regeneration and Disease. International Journal of Molecular Sciences, 21(5), 1830. https://doi.org/10.3390/ijms21051830