1. Trang chủ
  2. » Y Tế - Sức Khỏe

Sarcopenia Age-Related Muscle Wasting and Weakness: Mechanisms and Treatments P24 pdf

10 240 0

Đang tải... (xem toàn văn)

THÔNG TIN TÀI LIỆU

Nội dung

216 A.P. Russell and B. Lèger Artavanis-Tsakonas, S., Rand, M. D., Lake, R. J. (1999). Notch signaling: Cell fate control and signal integration in development. Science, 284, 770–776. Baumann, A. P., Ibebunjo, C., Grasser, W. A., Paralkar, V. M. (2003). Myostatin expression in age and denervation-induced skeletal muscle atrophy. Journal of Musculoskeletal & Neuronal Interactions, 3, 8–16. Bodine, S. C., Latres, E., Baumhueter, S., Lai, V. K., Nunez, L., Clarke, B. A., Poueymirou, W. T., Panaro, F. J., Na, E., Dharmarajan, K., Pan, Z. Q., Valenzuela, D. M., Dechiara, T. M., Stitt, T. N., Yancopoulos, G. D., Glass, D. J. (2001). Identification of ubiquitin ligases required for skeletal muscle atrophy. Science, 294, 1704–1708. Bodine, S. C., Stitt, T. N., Gonzalez, M., Kline, W. O., Stover, G. L., Bauerlein, R., Zlotchenko, E., Scrimgeour, A., Lawrence, J. C., Glass, D. J., Yancopoulos, G. D. (2001). Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nature Cell Biology, 3, 1014–1019. Brack, A. S. & Rando, T. A. (2007). Intrinsic changes and extrinsic influences of myogenic stem cell function during aging. Stem Cell Reviews, 3, 226–237. Brack, A. S., Bildsoe, H., Hughes, S. M. (2005). Evidence that satellite cell decrement contributes to preferential decline in nuclear number from large fibres during murine age-related muscle atrophy. Journal of Cell Science, 118, 4813–4821. Brunet, A., Bonni, A., Zigmond, M. J., Lin, M. Z., Juo, P., Hu, L. S., Anderson, M. J., Arden, K. C., Blenis, J., Greenberg, M. E. (1999). Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell, 96, 857–868. Carson, J. A., Schwartz, R. J., Booth, F. W. (1996). SRF and TEF–1 control of chicken skeletal alpha-actin gene during slow-muscle hypertrophy. The American Journal of Physiology, 270, C1624–C1633. Charvet, C., Houbron, C., Parlakian, A., Giordani, J., Lahoute, C., Bertrand, A., Sotiropoulos, A., Renou, l., Schmitt, A., Melki, J., Li, Z., Daegelen, D., Tuil, D. (2006). New role for serum response factor in postnatal skeletal muscle growth and regeneration via the interleukin 4 and insulin-like growth factor 1 pathways. Molecular and Cellular Biology, 26, 6664–6674. Christov, C., Chretien, F., Abou-Khalil, R., Bassez, G., Vallet, G., Authier, F. J., Bassaglia, Y., Shinin, V., Tajbakhsh, S., Chazaud, B., Gherardi, R. K. (2007). Muscle satellite cells and endothelial cells: Close neighbors and privileged partners. Molecular Biology of the Cell, 18, 1397–1409. Clavel, S., Coldefy, A. S., Kurkdjian, E., Salles, J., Margaritis, I., Derijard, B. (2006). Atrophy- related ubiquitin ligases, atrogin-1 and MuRF1 are up-regulated in aged rat Tibialis Anterior muscle. Mechanisms of Ageing and Development, 127, 794–801. Conboy, I. M. & Rando, T. A. (2002). The regulation of Notch signaling controls satellite cell activa- tion and cell fate determination in postnatal myogenesis. Developmental Cell, 3, 397–409. Conboy, I. M., Conboy, M. J., Smythe, G. M., Rando, T. A. (2003). Notch-mediated restoration of regenerative potential to aged muscle. Science, 302, 1575–1577. Conboy, I. M., Conboy, M. J., Wagers, A. J., Girma, E. R., Weissman, I. L., Rando, T. A. (2005). Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature, 433, 760–764. Corpas, E., Harman, S. M., Blackman, M. R. (1993). Human growth hormone and human aging. Endocrine Reviews, 14, 20–39. Cuthbertson, D., Smith, K., Babraj, J., Leese, G., Waddell, T., Atherton, P., Wackerhage, H., Taylor, P. M., Rennie, M. J. (2005). Anabolic signaling deficits underlie amino acid resistance of wasting, aging muscle. The Faseb Journal, 19, 422–424. Datta, S. R., Brunet, A., Greenberg, M. E. (1999). Cellular survival: A play in three Akts. Genes & Development, 13, 2905–2927. Dedkov, E. I., Borisov, A. B., Wernig, A., Carlson, B. M. (2003). Aging of skeletal muscle does not affect the response of satellite cells to denervation. The Journal of Histochemistry and Cytochemistry, 51, 853–863. Dreyer, H. C., Blanco, C. E., Sattler, F. R., Schroeder, E. T., Wiswell, R. A. (2006). Satellite cell numbers in young and older men 24 hours after eccentric exercise. Muscle & Nerve, 33, 242–253. 217Age-Related Changes in the Molecular Regulation of Skeletal Muscle Mass Edstrom, E., Altun, M., Hagglund, M., Ulfhake, B. (2006). Atrogin-1/MAFbx and MuRF1 are downregulated in aging-related loss of skeletal muscle. The Journals of Gerontology. Series A: Biological Sciences and Medical Sciences, 61, 663–674. Emanuelli, B., Peraldi, P., Filloux, C., Chavey, C., Freidinger, K., Hilton, D. J., Hotamisligil, G. S., Van Obberghen, E. (2001). SOCS-3 inhibits insulin signaling and is up-regulated in response to tumor necrosis factor-alpha in the adipose tissue of obese mice. The Journal of Biological Chemistry, 276, 47944–47949. Frost, R. A., Nystrom, G. J., Lang, C. H. (2003). Tumor necrosis factor-alpha decreases insulin- like growth factor-I messenger ribonucleic acid expression in C2C12 myoblasts via a Jun N-terminal kinase pathway. Endocrinology, 144, 1770–1779. Gibson, M. C. & Schultz, E. (1983). Age-related differences in absolute numbers of skeletal muscle satellite cells. Muscle & Nerve, 6, 574–580. Glass, D. J. (2003). Signalling pathways that mediate skeletal muscle hypertrophy and atrophy. Nature Cell Biology, 5, 87–90. Glass, D. J. (2005). Skeletal muscle hypertrophy and atrophy signaling pathways. The International Journal of Biochemistry & Cell Biology, 37, 1974–1984. Gomes, M. D., Lecker, S. H., Jagoe, R. T., Navon, A., Goldberg, A. L. (2001). Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy. Proceedings of the National Academy of Sciences of the United States of America, 98, 14440–14445. Gonzalez-Cadavid, N. F., Taylor, W. E., Yarasheski, K., Sinha-Hikim, I., MA, K., Ezzat, S., Shen, R., Lalani, R., Asa, S., Mamita, M., Nair, G., Arver, S., Bhasin, S. (1998). Organization of the human myostatin gene and expression in healthy men and HIV-infected men with muscle wasting. Proceedings of the National Academy of Sciences of the United States of America, 95, 14938–14943. Greiwe, J. S., Cheng, B., Rubin, D. C., Yarasheski, K. E., Semenkovich, C. F. (2001). Resistance exercise decreases skeletal muscle tumor necrosis factor alpha in frail elderly humans. The FASEB Journal, 15, 475–482. Grobet, L., Martin, L. J., Poncelet, D., Pirottin, D., Brouwers, B., Riquet, J., Schoeberlein, A., Dunner, S., Menissier, F., Massabanda, J., Fries, R., Hanset, R., Georges, M. (1997). A dele- tion in the bovine myostatin gene causes the double-muscled phenotype in cattle. Nature Genetics, 17, 71–74. Haddad, F. & Adams, G. R. (2006). Aging-sensitive cellular and molecular mechanisms associ- ated with skeletal muscle hypertrophy. Journal of Applied Physiology, 100, 1188–1203. Hansen, J. A., Lindberg, K., Hilton, D. J., Nielsen, J. H., Billestrup, N. (1999). Mechanism of inhibition of growth hormone receptor signaling by suppressor of cytokine signaling proteins. Molecular Endocrinology, 13, 1832–1843. Hawke, T. J. & Garry, D. J. (2001). Myogenic satellite cells: Physiology to molecular biology. Journal of Applied Physiology, 91, 534–551. Herrington, J. & Carter-Su, C. (2001). Signaling pathways activated by the growth hormone recep- tor. Trends in Endocrinology and Metabolism, 12, 252–257. Hornberger, T. A., Stuppard, R., Conley, K. E., Fedele, M. J., Fiorotto, M. L., Chin, E. R., Esser, K. A. (2004). Mechanical stimuli regulate rapamycin-sensitive signalling by a phosphoinositide 3-kinase-, protein kinase B- and growth factor-independent mechanism. The Biochemical Journal, 380, 795–804. Jefferson, L. S., Fabian, J. R., Kimball, S. R. (1999). Glycogen synthase kinase-3 is the predomi- nant insulin-regulated eukaryotic initiation factor 2B kinase in skeletal muscle. The International Journal of Biochemistry & Cell Biology, 31, 191–200. Kadi, F., Charifi, N., Denis, C., Lexell, J. (2004). Satellite cells and myonuclei in young and elderly women and men. Muscle & Nerve, 29, 120–127. Kadi, F., Charifi, N., Denis, C., Lexell, J., Andersen, J. L., Schjerling, P., Olsen, S., Kjaer, M. (2005). The behaviour of satellite cells in response to exercise: What have we learned from human studies? Pflugers Archives, 451, 319–327. Katsanos, C. S., Kobayashi, H., Sheffield-Moore, M., Aarsland, A., Wolfe, R. R. (2005). Aging is associated with diminished accretion of muscle proteins after the ingestion of a small 218 A.P. Russell and B. Lèger bolus of essential amino acids. The American Journal of Clinical Nutrition, 82, 1065–1073. Kawada, S., Tachi, C., Ishii, N. (2001). Content and localization of myostatin in mouse skeletal muscles during aging, mechanical unloading and reloading. Journal of Muscle Research and Cell Motility, 22, 627–633. Kimball, S. R., O’malley, J. P., Anthony, J. C., Crozier, S. J., Jefferson, L. S. (2004). Assessment of biomarkers of protein anabolism in skeletal muscle during the life span of the rat: Sarcopenia despite elevated protein synthesis. American Journal of Physiology. Endocrinology and Metabolism, 287, E772–E780. Kuwahara, K., Barrientos, T., Pipes, G. C., LI, S., Olson, E. N. (2005). Muscle-specific signaling mechanism that links actin dynamics to serum response factor. Molecular and Cellular Biology, 25, 3173–3181. Lahoute, C., Sotiropoulos, A., Favier, M., Guillet-Deniau, I., Charvet, C., Ferry, A., Butler- Browne, G., Metzger, D., Tuil, D., Daegelen, D. (2008). Premature aging in skeletal muscle lacking serum response factor. PLoS ONE, 3, e3910. Lamon, S., Wallace, M. A., Leger, B., Russell, A. P. (2009). Regulation of STARS and its down- stream targets suggest a novel pathway involved in human skeletal muscle hypertrophy and atrophy. Journal de Physiologie, 587, 1795–1803. Langley, B., Thomas, M., Bishop, A., Sharma, M., Gilmour, S., Kambadur, R. (2002). Myostatin inhibits myoblast differentiation by down-regulating MyoD expression. The Journal of Biological Chemistry, 277, 49831–49840. Latres, E., Amini, A. R., Amini, A. A., Griffiths, J., Martin, F. J., Wei, Y., Lin, H. C., Yancopoulos, G. D., Glass, D. J. (2005). Insulin-like growth factor-1 (IGF-1) inversely regulates atrophy- induced genes via the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway. The Journal of Biological Chemistry, 280, 2737–2744. Lecker, S. H., Solomon, V., Mitch, W. E., Goldberg, A. L. (1999). Muscle protein breakdown and the critical role of the ubiquitin-proteasome pathway in normal and disease states. The Journal of Nutrition, 129, 227S–237S. Leger, B., Derave, W., De Bock, K., Hespel, P., Russell, A. P. (2008). Human sarcopenia reveals an increase in SOCS-3 and myostatin and a reduced efficiency of Akt phosphorylation. Rejuvenation Research, 11, 163–175B. Lexell, J. (1995). Human aging, muscle mass, and fiber type composition. The Journals of Gerontology. Series A: Biological Sciences and Medical Sciences, 50 Spec No, 11–16. Liu, W., Thomas, S. G., Asa, S. L., Gonzalez-Cadavid, N., Bhasin, S., Ezzat, S. (2003). Myostatin is a skeletal muscle target of growth hormone anabolic action. The Journal of Clinical Endocrinology and Metabolism, 88, 5490–5496. Lupu, F., Terwilliger, J. D., Lee, K., Segre, G. V., Efstratiadis, A. (2001). Roles of growth hormone and insulin-like growth factor 1 in mouse postnatal growth. Developmental Biology, 229, 141–162. Mahadeva, H., Brooks, G., Lodwick, D., Chong, N. W., Samani, N. J. (2002). ms1, a novel stress- responsive, muscle-specific gene that is up-regulated in the early stages of pressure overload- induced left ventricular hypertrophy. FEBS Letters, 521, 100–104. Mahoney, J., Sager, M., Dunham, N. C., Johnson, J. (1994). Risk of falls after hospital discharge. Journal of the American Geriatrics Society, 42, 269–274. McFarlane, C., Plummer, E., Thomas, M., Hennebry, A., Ashby, M., Ling, N., Smith, H., Sharma, M., Kambadur, R. (2006). Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF-kappaB-independent, FoxO1-dependent mechanism. The Journal of Cell Physiology, 209(2), 501–514. McPherron, A. C., Lawler, A. M., Lee, S. J. (1997). Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature, 387, 83–90. Miralles, F., Posern, G., Zaromytidou, A. I., Treisman, R. (2003). Actin dynamics control SRF activity by regulation of its coactivator Mal. Cell, 113, 329–342. Morissette, M., Cook, S., Buranasombati, C., Rosenberg, M., Rosenzweig, A. (2009). Myostatin inhibits IGF-I induced myotube hypertrophy through Akt. American Journal of Physiology. Cell Physiology, 297(5), 1124–1132. 219Age-Related Changes in the Molecular Regulation of Skeletal Muscle Mass Morissette, M. R., Stricker, J. C., Rosenberg, M. A., Buranasombati, C., Levitan, E. B., Mittleman, M. A., Rosenzweig, A. (2009). Effects of myostatin deletion in aging mice. Aging Cell, 8, 573–583. Nishimura, T., Oyama, K., Kishioka, Y., Wakamatsu, J., Hattori, A. (2007). Spatiotemporal expression of decorin and myostatin during rat skeletal muscle development. Biochemical and Biophysical Research Communications, 361, 896–902. Nnodim, J. O. (2000). Satellite cell numbers in senile rat levator ani muscle. Mechanisms of Ageing and Development, 112, 99–111. Pallafacchina, G., Calabria, E., Serrano, A. L., Kalhovde, J. M., Schiaffino, S. (2002). A protein kinase B-dependent and rapamycin-sensitive pathway controls skeletal muscle growth but not fiber type specification. Proceedings of the National Academy of Sciences of the United States of America, 99, 9213–9218. Petrella, J. K., Kim, J. S., Cross, J. M., Kosek, D. J., Bamman, M. M. (2006). Efficacy of myonu- clear addition may explain differential myofiber growth among resistance-trained young and older men and women. American Journal of Physiology. Endocrinology and Metabolism, 291, E937–E946. Ram, P. A. & Waxman, D. J. (1999). SOCS/CIS protein inhibition of growth hormone-stimulated STAT5 signaling by multiple mechanisms. The Journal of Biological Chemistry, 274, 35553–35561. Raue, U., Slivka, D., Jemiolo, B., Hollon, C., Trappe, S. (2006). Myogenic gene expression at rest and after a bout of resistance exercise in young (18–30 yr) and old (80–89 yr) women. Journal of Applied Physiology, 101, 53–59. Raue, U., Slivka, D., Jemiolo, B., Hollon, C., Trappe, S. (2007). Proteolytic gene expression dif- fers at rest and after resistance exercise between young and old women. The Journals of Gerontology. Series A: Biological Sciences and Medical Sciences, 62, 1407–1412. Renault, V., Thornell, L. E., Eriksson, P. O., Butler-Browne, G., Mouly, V. (2002). Regenerative potential of human skeletal muscle during aging. Aging Cell, 1, 132–139. Rennie, M. J., Selby, A., Atherton, P., Smith, K., Kumar, V., Glover, E. L., Philips, S. M. (2009). Facts, noise and wishful thinking: Muscle protein turnover in aging and human disuse atrophy. Scandinavian Journal of Medicine & Science in Sports, 20(1), 5–9. Rhoads, R. E. (1999). Signal transduction pathways that regulate eukaryotic protein synthesis. The Journal of Biological Chemistry, 274, 30337–30340. Rommel, C., Bodine, S. C., Clarke, B. A., Rossman, R., Nunez, L., Stitt, T. N., Yancopoulos, G. D., Glass, D. J. (2001). Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/ mTOR and PI(3)K/Akt/GSK3 pathways. Nature Cell Biology, 3, 1009–1013. Roth, S. M., Martel, G. F., Ivey, F. M., Lemmer, J. T., Metter, E. J., Hurley, B. F., Rogers, M. A. (2000). Skeletal muscle satellite cell populations in healthy young and older men and women. The Anatomical Record, 260, 351–358. Russell, A. P. (2009). The molecular regulation of skeletal muscle mass. Clinical and Experimental Pharmacology & Physiology, 37(3), 378–384. Ryan, N. A., Zwetsloot, K. A., Westerkamp, L. M., Hickner, R. C., Pofahl, W. E., Gavin, T. P. (2006). Lower skeletal muscle capillarization and VEGF expression in aged vs. young men. Journal of Applied Physiology, 100, 178–185. Sajko, S., Kubinova, L., Cvetko, E., Kreft, M., Wernig, A., Erzen, I. (2004). Frequency of M-cadherin-stained satellite cells declines in human muscles during aging. The Journal of Histochemistry and Cytochemistry, 52, 179–185. Sakuma, K., Akiho, M., Nakashima, H., Akima, H., Yasuhara, M. (2008). Age-related reductions in expression of serum response factor and myocardin-related transcription factor A in mouse skeletal muscles. Biochimica et Biophysica Acta, 1782, 453–461. Sandri, M., Sandri, C., Gilbert, A., Skurk, C., Calabria, E., Picard, A., Walsh, K., Schiaffino, S., Lecker, S. H., Goldberg, A. L. (2004). Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell, 117, 399–412. Sandri, M., Lin, J., Handschin, C., Yang, W., Arany, Z. P., Lecker, S. H., Goldberg, A. L., Spiegelman, B. M. (2006). PGC-1alpha protects skeletal muscle from atrophy by suppressing 220 A.P. Russell and B. Lèger FoxO3 action and atrophy-specific gene transcription. Proceedings of the National Academy of Sciences of the United States of America, 103, 16260–16265. Schafer, R., Zweyer, M., Knauf, U., Mundegar, R. R., Wernig, A. (2005). The ontogeny of soleus muscles in mdx and wild type mice. Neuromuscular Disorders, 15, 57–64. Schuelke, M., Wagner, K. R., Stolz, L. E., Hubner, C., Riebel, T., Komen, W., Braun, T., Tobin, J. F., Lee, S. J. (2004). Myostatin mutation associated with gross muscle hypertrophy in a child. The New England Journal of Medicine, 350, 2682–2688. Shefer, G., Vande Mark, D. P., Richardson, J. B., Yablonka-Reuveni, Z. (2006). Satellite-cell pool size does matter: Defining the myogenic potency of aging skeletal muscle. Developmental Biology, 294, 50–66. Snijders, T., Verdijk, L. B., Van Loon, L. J. (2009). The impact of sarcopenia and exercise training on skeletal muscle satellite cells. Ageing Research Reviews, 8(4), 328–338. Sotiropoulos, A., Gineitis, D., Copeland, J., Treisman, R. (1999). Signal-regulated activation of serum response factor is mediated by changes in actin dynamics. Cell, 98, 159–169. Stitt, T. N., Drujan, D., Clarke, B. A., Panaro, F., Timofeyva, Y., Kline, W. O., Gonzalez, M., Yancopoulos, G. D., Glass, D. J. (2004). The Igf-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting Foxo transcription factors. Molecular Cell, 14, 395–403. Sun, K., Battle, M. A., Misra, R. P., Duncan, S. A. (2009). Hepatocyte expression of serum response factor is essential for liver function, hepatocyte proliferation and survival, and post- natal body growth in mice. Hepatology, 49, 1645–1654. Supakar, P. C. & Roy, A. K. (1996). Role of transcription factors in the age-dependent regulation of the androgen receptor gene in rat liver. Biological Signals, 5, 170–179. Thomas, M., Langley, B., Berry, C., Sharma, M., Kirk, S., Bass, J., Kambadur, R. (2000). Myostatin, a negative regulator of muscle growth, functions by inhibiting myoblast prolifera- tion. The Journal of Biological Chemistry, 275, 40235–40243. Tollet-Egnell, P., Flores-Morales, A., Stavreus-Evers, A., Sahlin, L., Norstedt, G. (1999). Growth hormone regulation of SOCS-2, SOCS-3, and CIS messenger ribonucleic acid expression in the rat. Endocrinology, 140, 3693–3704. Verdijk, L. B., Koopman, R., Schaart, G., Meijer, K., Savelberg, H. H., Van Loon, L. J. (2007). Satellite cell content is specifically reduced in type II skeletal muscle fibers in the elderly. American Journal of Physiology. Endocrinology and Metabolism, 292, E151–E157. Verney, J., Kadi, F., Charifi, N., Feasson, L., Saafi, M. A., Castells, J., Piehl-Aulin, K., Denis, C. (2008). Effects of combined lower body endurance and upper body resistance training on the satellite cell pool in elderly subjects. Muscle & Nerve, 38, 1147–1154. Vivanco, I. & Sawyers, C. L. (2002). The phosphatidylinositol 3-Kinase Akt pathway in human cancer. Nature Reviews. Cancer, 2, 489–501. Volpi, E., Sheffield-Moore, M., Rasmussen, B. B., Wolfe, R. R. (2001). Basal muscle amino acid kinetics and protein synthesis in healthy young and older men. JAMA, 286, 1206–1212. Welle, S. (2002). Cellular and molecular basis of age-related sarcopenia. Canadian Journal of Applied Physiology, 27, 19–41. Welle, S., Bhatt, K., Shah, B., Thornton, C. (2002). Insulin-like growth factor-1 and myostatin mRNA expression in muscle: Comparison between 62–77 and 21–31 yr old men. Experimental Gerontology, 37, 833–839. Welle, S., Brooks, A. I., Delehanty, J. M., Needler, N., Thornton, C. A. (2003). Gene expression profile of aging in human muscle. Physiological Genomics, 14, 149–159. Welsh, G. I., Stokes, C. M., Wang, X., Sakaue, H., Ogawa, W., Kasuga, M., Proud, C. G. (1997). Activation of translation initiation factor eIF2B by insulin requires phosphatidyl inositol 3-kinase. Febs Letters, 410, 418–422. Whitman, S. A., Wacker, M. J., Richmond, S. R., Godard, M. P. (2005). Contributions of the ubiquitin-proteasome pathway and apoptosis to human skeletal muscle wasting with age. Pflugers Archives, 450, 437–446. Wilkes, E. A., Selby, A. L., Atherton, P. J., Patel, R., Rankin, D., Smith, K., Rennie, M. J. (2009). Blunting of insulin inhibition of proteolysis in legs of older subjects may contribute to age- related sarcopenia. The American Journal of Clinical Nutrition, 90(5), 1343–1350. 221Age-Related Changes in the Molecular Regulation of Skeletal Muscle Mass Woelfle, J. & Rotwein, P. (2004). In vivo regulation of growth hormone-stimulated gene transcription by STAT5b. American Journal of Physiology. Endocrinology and Metabolism, 286, E393–E401. Zadik, Z., Chalew, S. A., Mccarter, R. J., JR Meistas, M., Kowarski, A. A. (1985). The influence of age on the 24-hour integrated concentration of growth hormone in normal individuals. The Journal of Clinical Endocrinology and Metabolism, 60, 513–516. 223 G.S. Lynch (ed.), Sarcopenia – Age-Related Muscle Wasting and Weakness, DOI 10.1007/978-90-481-9713-2_11, © Springer Science+Business Media B.V. 2011 Abstract Skeletal muscle is one of the most heritable quantitative traits studied to date, with heritability estimates ranging from 30% to 85% for muscle strength measures and 50–80% for lean mass measures. The strong genetic contribution to skeletal muscle traits indicates the possibility of using genetic approaches to indi- vidualize treatment approaches for sarcopenia or even aid in prevention strategies through the use of genetic screening prior to functional limitations. While these possibilities provide the rationale and motivation for genetic studies of skeletal muscle traits, few genes have been identified to date that appear to contribute to variation in either skeletal muscle strength or mass phenotypes, let alone sarcope- nia itself. The ACE, ACTN3, CNTF, and VDR genes have been associated with skeletal muscle strength in two or more papers each, while the AR, TRHR, and VDR genes have been similarly associated with muscle mass. Only the VDR gene has been significantly associated with sarcopenia itself as an endpoint phenotype but replication of this initial finding has not yet been performed. Large-scale clini- cal studies relying on advanced genome-wide association techniques are needed to provide further insights into potentially clinically relevant genes that contrib- ute to skeletal muscle traits, with identified genes then explored functionally to determine the likelihood that genetic screening can assist in the prevention and treatment of sarcopenia. Keywords Genotype • Heritability • Muscle mass • Muscle strength • Polymorphism Genetic Variation and Skeletal Muscle Traits: Implications for Sarcopenia Stephen M. Roth S.M. Roth (*) Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD 20742, USA e-mail: sroth1@umd.edu 224 S.M. Roth 1 Introduction Aging is associated with a decline in skeletal muscle mass, strength, power and physical functioning, generally termed sarcopenia (Dutta and Hadley 1995). These well-documented losses of muscle strength, mass, and muscle quality (limb strength/limb muscle mass) with age (Lindle et al. 1997; Baumgartner et al. 1998; Janssen et al. 2002; Lauretani et al. 2003; Sowers et al. 2005; Ploutz-Snyder et al. 2002) have important health consequences, because this deterioration in muscle structure and function is associated with an increased risk of falls, hip fractures, and functional decline (Schultz et al. 1997; Aniansson et al. 1984; Janssen et al. 2004; Newman et al. 2003a; Lauretani et al. 2003; Sowers et al. 2005). Muscle strength is independently associated with functional ability in the elderly (Hyatt et al. 1990; Visser et al. 2000a; Kwon et al. 2001; Purser et al. 2003; Rantanen et al. 1998; Foldvari et al. 2000; Lauretani et al. 2003; Pendergast et al. 1993) and may explain up to 25% of the variance in overall functional ability (Buchner and deLateur 1991). Furthermore, sarcopenia is related to a reduction in the performance of activities of daily living (Nybo et al. 2001), which may lead to further declines in muscle mass and strength and greater reductions in the performance of those activi- ties. The net effect of this cycle can result in marked disablement, predisposing older individuals to falls, injuries and disability (Rantanen et al. 2000). Although the loss of muscle mass is associated with the decline in strength in older adults, the strength decline is much more rapid than the concomitant loss of muscle mass, suggesting a decline in muscle quality (Goodpaster et al. 2006). The loss of muscle strength is an independent predictor of mortality in the elderly, more so than loss of muscle mass (Metter et al. 2002; Rantanen et al. 2000, 2003; Fujita et al. 1995; Laukkanen et al. 1995; Newman et al. 2003b). Thus, the relationship of muscle mass and strength to mortality may rest in the higher functional capacity associated with having more muscle strength and mass, and an inverse association with functional limitations and disability. Sex differences have been shown, with women showing an earlier age of onset of sarcopenia (Lauretani et al. 2003; Janssen et al. 2002), and a greater prevalence of functional impairment at any age in com- parison to men (Lauretani et al. 2003; Rantanen and Avela 1997; Ostchega et al. 2000; Dunlap et al. 2002; Visser et al. 2000b), most likely owing to their lower muscle mass and strength levels compared to men throughout the adult age span (Frontera et al. 1991; Lindle et al. 1997; Rantanen and Avela 1997; Lauretani et al. 2003). The consequences of sarcopenia-related disability are significant both in terms of personal quality of life and to the overall economy, with healthcare costs related to sarcopenia in the United States estimated to be $18.5 billion dollars for adults 60 years and older for the year 2000 (Janssen et al. 2004). Though the losses of muscle mass and strength begin on average between 40 and 50 years of age, losses for any particular individual are quite variable. For example, investigators from our laboratories at the University of Maryland have reported substantial age-related declines in strength and muscle quality in men and women from the Baltimore Longitudinal Study of Aging (BLSA) (Lindle et al. 1997; 225Genetic Variation and Skeletal Muscle Traits: Implications for Sarcopenia Lynch et al. 1999). However, we’ve observed enormous inter-individual variability in muscle strength within each age group that could not be explained by previous muscular activity levels. For example, the highest strength values for 80–96 year old men and women were two to four times higher than the lowest strength values in 20–39 year old men and women (Table 1). Furthermore, at least 15% of the men and women >60 year had strength values that were above the average values for 20 year old subjects. Similar inter-individual variations existed for leg muscle mass (Lindle et al. 1997) and for muscle quality in both older men and women (Lynch et al. 1999). Sarcopenia has been reported in community-dwelling men and women below the age of 50 year (Melton et al. 2000; Tanko et al. 2002; Janssen et al. 2002; Lauretani et al. 2003), and recently, sarcopenia associated with compromised physical functioning was shown to occur in nearly one in ten women aged 34–58 year (mid-life) (Sowers et al. 2005), providing further support for the variable onset of muscle strength losses and an indication of susceptibility to sarcopenia in some individuals. Various research groups are currently exploring the possibility that a portion of this inter-individual variability and susceptibility to early muscle losses is due to genetic factors, which could someday be used to identify susceptible men and women and individualize their prevention and treatment interventions. This review discusses the genetic aspects of skeletal muscle traits with an emphasis on sarcopenia, including examination of heritability, linkage analysis, and specific genes associated with relevant traits. While skeletal muscle remains one of the most heritable health-related quantitative phenotypes studied to date, the iden- tification of specific contributing genes remains at the early stages and much work remains to determine the future clinical importance of genetic contributions to sar- copenia risk. This review will not address the potential role of mitochondrial DNA mutations in the development of sarcopenia (Hiona and Leeuwenburgh 2008), as these genetic variations represent age-related, sporadic modifications of DNA sequence rather than stable, genome-wide genetic variants present since birth in all somatic cells. 2 Heritability of Skeletal Muscle Traits Variation in skeletal muscle traits among individuals can be attributed to environ- mental factors, genetic factors, or the interaction of both. While the influence of environmental factors such as physical activity and diet have been broadly investi- gated, only recently have studies begun to address the specific genetic influences on skeletal muscle traits that may explain the inter-individual variability noted above. The earliest of these studies examined familial aggregation of body compo- sition traits in twins, especially exploiting the slight but important differences between monozygotic and dizygotic twin pairs. Monozygotic twins share not only 100% of genetic variation in their DNA sequence, but also share the intrauterine environment and very likely a similar environment through adolescence. Dizygotic . (2001). Basal muscle amino acid kinetics and protein synthesis in healthy young and older men. JAMA, 286, 1206–1212. Welle, S. (2002). Cellular and molecular basis of age-related sarcopenia. Canadian. individuals. The Journal of Clinical Endocrinology and Metabolism, 60, 513–516. 223 G.S. Lynch (ed.), Sarcopenia – Age-Related Muscle Wasting and Weakness, DOI 10.1007/978-90-481-9713-2_11, ©. functioning, generally termed sarcopenia (Dutta and Hadley 1995). These well-documented losses of muscle strength, mass, and muscle quality (limb strength/limb muscle mass) with age (Lindle

Ngày đăng: 07/07/2014, 10:20

TỪ KHÓA LIÊN QUAN

TÀI LIỆU CÙNG NGƯỜI DÙNG

TÀI LIỆU LIÊN QUAN