Molecular aging and rejuvenation of human muscle stem cells

Very little remains known about the regulation of human organ stem cells (in general, and during the aging process), and most previous data were collected in short‐lived rodents. We examined whether stem cell aging in rodents could be extrapolated to genetically and environmentally variable humans. Our findings establish key evolutionarily conserved mechanisms of human stem cell aging. We find that satellite cells are maintained in aged human skeletal muscle, but fail to activate in response to muscle attrition, due to diminished activation of Notch compounded by elevated transforming growth factor beta (TGF‐β)/phospho Smad3 (pSmad3). Furthermore, this work reveals that mitogen‐activated protein kinase (MAPK)/phosphate extracellular signal‐regulated kinase (pERK) signalling declines in human muscle with age, and is important for activating Notch in human muscle stem cells. This molecular understanding, combined with data that human satellite cells remain intrinsically young, introduced novel therapeutic targets. Indeed, activation of MAPK/Notch restored ‘youthful’ myogenic responses to satellite cells from 70‐year‐old humans, rendering them similar to cells from 20‐year‐old humans. These findings strongly suggest that aging of human muscle maintenance and repair can be reversed by ‘youthful’ calibration of specific molecular pathways.

[1]  M. Brooke,et al.  Muscle fiber types: how many and what kind? , 1970, Archives of neurology.

[2]  K. Sahlin,et al.  Influence of severe potassium depletion and subsequent repletion with potassium on muscle electrolytes, metabolites and amino acids in man. , 1976, Clinical science and molecular medicine.

[3]  E. Schultz,et al.  Skeletal muscle satellite cells: Changes in proliferation potential as a function of age , 1982, Mechanisms of Ageing and Development.

[4]  J. Kuiper,et al.  Age‐related changes in wound healing , 1994, Clinical and experimental dermatology.

[5]  M. Brown,et al.  Differential effects of reduced muscle use (hindlimb unweighting) on skeletal muscle with aging , 1996, Aging.

[6]  M. Grounds Age‐associated Changes in the Response of Skeletal Muscle Cells to Exercise and Regeneration a , 1998, Annals of the New York Academy of Sciences.

[7]  G. Butler-Browne,et al.  Regenerative potential of human skeletal muscle during aging , 2002, Aging cell.

[8]  Iva Greenwald,et al.  Endocytosis-mediated downregulation of LIN-12/Notch upon Ras activation in Caenorhabditis elegans , 2002, Nature.

[9]  Marc S Halfon,et al.  Reciprocal regulatory interactions between the Notch and Ras signaling pathways in the Drosophila embryonic mesoderm. , 2002, Developmental biology.

[10]  T. Rando,et al.  The regulation of Notch signaling controls satellite cell activation and cell fate determination in postnatal myogenesis. , 2002, Developmental cell.

[11]  Gayle M. Smythe,et al.  Notch-Mediated Restoration of Regenerative Potential to Aged Muscle , 2003, Science.

[12]  H. Degens,et al.  Skeletal muscle function and hypertrophy are diminished in old age , 2003, Muscle & nerve.

[13]  Per Aagaard,et al.  Training-induced changes in muscle CSA, muscle strength, EMG, and rate of force development in elderly subjects after long-term unilateral disuse. , 2004, Journal of applied physiology.

[14]  A. Wagers,et al.  Cellular and Molecular Signatures of Muscle Regeneration: Current Concepts and Controversies in Adult Myogenesis , 2005, Cell.

[15]  A. Petrie,et al.  Stem Cell Function, Self-Renewal, and Behavioral Heterogeneity of Cells from the Adult Muscle Satellite Cell Niche , 2005, Cell.

[16]  F. Booth,et al.  Changes in signalling molecule levels in 10-day hindlimb immobilized rat muscles. , 2005, Acta physiologica Scandinavica.

[17]  I. Weissman,et al.  Rejuvenation of aged progenitor cells by exposure to a young systemic environment , 2005, Nature.

[18]  G. Shefer,et al.  Satellite-cell pool size does matter: defining the myogenic potency of aging skeletal muscle. , 2006, Developmental biology.

[19]  K. Bushby,et al.  The childhood limb-girdle muscular dystrophies. , 2006, Seminars in pediatric neurology.

[20]  T. Partridge,et al.  A Population of Myogenic Stem Cells That Survives Skeletal Muscle Aging , 2007, Stem Cells.

[21]  H. Langberg,et al.  Myofibre damage in human skeletal muscle: effects of electrical stimulation versus voluntary contraction , 2007, The Journal of physiology.

[22]  E. Pistilli,et al.  Interleukin-15 responses to aging and unloading-induced skeletal muscle atrophy. , 2007, American journal of physiology. Cell physiology.

[23]  I. Conboy,et al.  Loss of stem cell regenerative capacity within aged niches , 2007, Aging cell.

[24]  I. Conboy,et al.  Aging of signal transduction pathways, and pathology. , 2008, Experimental cell research.

[25]  G Castellani,et al.  Human models of aging and longevity. , 2008, Expert opinion on biological therapy.

[26]  Jan Vijg,et al.  Puzzles, promises and a cure for ageing , 2008, Nature.

[27]  Jeanne Shen,et al.  A temporal switch from notch to Wnt signaling in muscle stem cells is necessary for normal adult myogenesis. , 2008, Cell stem cell.

[28]  I. Conboy,et al.  Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells , 2008, Nature.