Hostname: page-component-8448b6f56d-xtgtn Total loading time: 0 Render date: 2024-04-24T14:49:31.764Z Has data issue: false hasContentIssue false

Stem cell ageing: does it happen and can we intervene?

Published online by Cambridge University Press:  19 November 2007

Ilaria Bellantuono*
Affiliation:
Academic Unit of Bone Biology, University of Sheffield Medical School, Sheffield, S10 2RX, UK.
W. Nicol Keith
Affiliation:
Centre for Oncology and Applied Pharmacology, University of Glasgow, Cancer Research UK Beatson Laboratories, Glasgow, G61 1BD, UK.
*
*Corresponding author: Ilaria Bellantuono, Room EU28, Academic Unit of Bone Biology, University of Sheffield Medical School, Beech Hill Rd, Sheffield, S10 2RX, UK. Tel: +44 (0)114 2711798; Fax: +44 (0)114 2711711; E-mail: i.bellantuono@shef.ac.uk

Abstract

Adult stem cells have become the focus of intense research in recent years as a result of their role in the maintenance and repair of tissues. They exert this function through their extensive expansion (self-renewal) and multipotent differentiation capacity. Understanding whether adult stem cells retain this capacity throughout the lifespan of the individual, or undergo a process of ageing resulting in a decreased stem cell pool, is an important area of investigation. Progress in this area has been hampered by lack of suitable models and of appropriate markers and assays to identify stem cells. However, recent data suggest that an understanding of the mechanisms governing stem cell ageing can give insight into the mechanism of tissue ageing and, most importantly, advance our ability to use stem cells in cell and gene therapy strategies.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2007

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Thomson, J.A. et al. (1998) Embryonic stem cell lines derived from human blastocysts. Science 282, 1145-1147CrossRefGoogle ScholarPubMed
2Reubinoff, B.E. et al. (2000) Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol 18, 399-404CrossRefGoogle ScholarPubMed
3Weissman, I.L. (2000) Stem cells: units of development, units of regeneration, and units in evolution. Cell 100, 157-168CrossRefGoogle ScholarPubMed
4Watt, F.M. (2000) Epidermal stem cells as targets for gene transfer. Hum Gene Ther 11, 2261-2266CrossRefGoogle ScholarPubMed
5Whitehead, R.H. et al. (1999) Clonogenic growth of epithelial cells from normal colonic mucosa from both mice and humans. Gastroenterology 117, 858-865CrossRefGoogle ScholarPubMed
6Potten, C.S. et al. (2003) Identification of a putative intestinal stem cell and early lineage marker; musashi-1. Differentiation 71, 28-41Google Scholar
7Wagers, A.J. and Conboy, I.M. (2005) Cellular and molecular signatures of muscle regeneration: current concepts and controversies in adult myogenesis. Cell 122, 659-667CrossRefGoogle ScholarPubMed
8Gage, F.H. (2000) Mammalian neural stem cells. Science 287, 1433-1438CrossRefGoogle ScholarPubMed
9Alison, M. (2002) Hepatic stem cells. Transplant Proc 34, 2702-2705Google Scholar
10Clarke, R.B. (2005) Isolation and characterization of human mammary stem cells. Cell Proliferation 38, 375-386CrossRefGoogle ScholarPubMed
11Lechner, A. and Habener, J.F. (2003) Stem/progenitor cells derived from adult tissues: potential for the treatment of diabetes mellitus. Am J Physiol Endocrinol Metab 284, E259-266Google Scholar
12Siminovitch, L., Till, J.E. and McCulloch, E.A. (1964) Decline In Colony-Forming Ability Of Marrow Cells Subjected To Serial Transplantation Into Irradiated Mice. J Cell Physiol 64, 23-31CrossRefGoogle ScholarPubMed
13Rando, T.A. (2007) The immortal strand hypothesis: segregation and reconstruction. Cell 129, 1239-1243Google Scholar
14Lansdorp, P.M. (2007) Immortal strands? Give me a break. Cell 129, 1244-1247CrossRefGoogle ScholarPubMed
15Wright, L.S. et al. (2003) Gene expression in human neural stem cells: effects of leukemia inhibitory factor. J Neurochem 86, 179-195CrossRefGoogle ScholarPubMed
16Rossi, D.J. et al. (2005) Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc Natl Acad Sci U S A 102, 9194-9199CrossRefGoogle ScholarPubMed
17Rossi, D.J. et al. (2007) Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725CrossRefGoogle ScholarPubMed
18Campisi, J. (2001) Cellular senescence as a tumor-suppressor mechanism. Trends Cell Biol 11, S27-31CrossRefGoogle ScholarPubMed
19Sharpless, N.E. and DePinho, R.A. (2004) Telomeres, stem cells, senescence, and cancer. J Clin Invest 113, 160-168CrossRefGoogle ScholarPubMed
20Pelicci, P.G. (2004) Do tumor-suppressive mechanisms contribute to organism aging by inducing stem cell senescence? J Clin Invest 113, 4-7Google Scholar
21Gil, J. and Peters, G. (2006) Regulation of the INK4b-ARF-INK4a tumour suppressor locus: all for one or one for all. Nat Rev Mol Cell Biol 7, 667-677Google Scholar
22Dimri, G.P. et al. (1995) A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A 92, 9363-9367CrossRefGoogle ScholarPubMed
23Paradis, V. et al. (2001) Replicative senescence in normal liver, chronic hepatitis C, and hepatocellular carcinomas. Hum Pathol 32, 327-332Google Scholar
24Lechel, A. et al. (2005) The cellular level of telomere dysfunction determines induction of senescence or apoptosis in vivo. EMBO Rep 6, 275-281CrossRefGoogle ScholarPubMed
25Wiemann, S.U. et al. (2002) Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis. Faseb J 16, 935-942Google Scholar
26Satyanarayana, A. et al. (2003) Telomere shortening impairs organ regeneration by inhibiting cell cycle re-entry of a subpopulation of cells. Embo J 22, 4003-4013CrossRefGoogle ScholarPubMed
27Sharpless, N.E. and DePinho, R.A. (2004) Telomeres, stem cells, senescence, and cancer. J Clin Invest 113, 160-168CrossRefGoogle ScholarPubMed
28Kim, W.Y. and Sharpless, N.E. (2006) The regulation of INK4/ARF in cancer and aging. Cell 127, 265-275Google Scholar
29Serrano, M. et al. (1997) Oncogenic ras provokes premature cell senescence associated with accumulation of 53 and p16INK4a. Cell 88, 593-602Google Scholar
30Maehama, T. and Dixon, J.E. (1998) The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J Biol Chem 273, 13375-13378CrossRefGoogle ScholarPubMed
31Di Cristofano, A. and Pandolfi, P.P. (2000) The multiple roles of PTEN in tumor suppression. Cell 100, 387-390CrossRefGoogle ScholarPubMed
32Hayflick, L. and Moorhead, P.S. (1961) The serial cultivation of human diploid cell strains. Exp Cell Res 25, 585-621Google Scholar
33Blackburn, E.H. (2005) Telomeres and telomerase: their mechanisms of action and the effects of altering their functions. FEBS Lett 579, 859-862Google Scholar
34Harley, C.B., Futcher, A.B. and Greider, C.W. (1990) Telomeres shorten during ageing of human fibroblasts. Nature 345, 458-460Google Scholar
35de Lange, T. (2002) Protection of mammalian telomeres. Oncogene 21, 532-540CrossRefGoogle ScholarPubMed
36Geserick, C. and Blasco, M.A. (2006) Novel roles for telomerase in aging. Mech Ageing Dev 127, 579-583Google Scholar
37Sherr, C.J. and DePinho, R.A. (2000) Cellular senescence: mitotic clock or culture shock? Cell 102, 407-410CrossRefGoogle ScholarPubMed
38Drayton, S. and Peters, G. (2002) Immortalisation and transformation revisited. Curr Opin Genet Dev 12, 98-104CrossRefGoogle ScholarPubMed
39Chin, L. et al. (1999) 53 deficiency rescues the adverse effects of telomere loss and cooperates with telomere dysfunction to accelerate carcinogenesis. Cell 97, 527-538Google Scholar
40Molofsky, A.V. et al. (2003) Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature 425, 962-967Google Scholar
41Iwama, A. et al. (2004) Enhanced self-renewal of hematopoietic stem cells mediated by the polycomb gene product Bmi-1. Immunity 21, 843-851Google Scholar
42Park, I.K. et al. (2003) Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature 423, 302-305Google Scholar
43Jacobs, J.J. et al. (1999) The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature 397, 164-168Google Scholar
44Itahana, K. et al. (2003) Control of the replicative life span of human fibroblasts by 16 and the polycomb protein Bmi-1. Mol Cell Biol 23, 389-401Google Scholar
45Guney, I., Wu, S. and Sedivy, J.M. (2006) Reduced c-Myc signaling triggers telomere-independent senescence by regulating Bmi-1 and 16(INK4a). Proc Natl Acad Sci U S A 103, 3645-3650Google Scholar
46Bruggeman, S.W. et al. (2005) Ink4a and Arf differentially affect cell proliferation and neural stem cell self-renewal in Bmi1-deficient mice. Genes Dev 19, 1438-1443Google Scholar
47Cantley, L.C. and Neel, B.G. (1999) New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proc Natl Acad Sci U S A 96, 4240-4245Google Scholar
48Stiles, B. et al. (2004) PTENless means more. Dev Biol 273, 175-184Google Scholar
49Kondo, M. et al. (2003) Biology of hematopoietic stem cells and progenitors: implications for clinical application. Annu Rev Immunol 21, 759-806Google Scholar
50Civin, C.I. et al. (1990) Positive stem cell selection–basic science. Prog Clin Biol Res 333, 387-401; discussion 402Google Scholar
51Brugger, W. et al. (1994) Positively selected autologous blood CD34+ cells and unseparated peripheral blood progenitor cells mediate identical hematopoietic engraftment after high-dose VP16, ifosfamide, carboplatin, and epirubicin. Blood 84, 1421-1426Google Scholar
52Bhatia, M. et al. (1997) Purification of primitive human hematopoietic cells capable of repopulating immune-deficient mice. Proc Natl Acad Sci U S A 94, 5320-5325CrossRefGoogle ScholarPubMed
53Glimm, H. et al. (2001) Previously undetected human hematopoietic cell populations with short-term repopulating activity selectively engraft NOD/SCID-beta2 microglobulin-null mice. J Clin Invest 107, 199-206Google Scholar
54Lajtha, L.G. (1979) Stem cell concepts. Differentiation 14, 23-34CrossRefGoogle ScholarPubMed
55Barrandon, Y. and Green, H. (1987) Three clonal types of keratinocyte with different capacities for multiplication. Proc Natl Acad Sci U S A 84, 2302-2306Google Scholar
56Morrison, S.J. et al. (1997) Identification of a lineage of multipotent hematopoietic progenitors. Development 124, 1929-1939Google Scholar
57Lavker, R.M. et al. (1993) Hair follicle stem cells: their location, role in hair cycle, and involvement in skin tumor formation. J Invest Dermatol 101, 16S-26SGoogle Scholar
58Tumbar, T. et al. (2004) Defining the epithelial stem cell niche in skin. Science 303, 359-363CrossRefGoogle ScholarPubMed
59Tamaki, S. et al. (2002) Engraftment of sorted/expanded human central nervous system stem cells from fetal brain. J Neurosci Res 69, 976-986CrossRefGoogle ScholarPubMed
60Quirici, N. et al. (2002) Isolation of bone marrow mesenchymal stem cells by anti-nerve growth factor receptor antibodies. Exp Hematol 30, 783-791Google Scholar
61Bensidhoum, M. et al. (2004) Homing of in vitro expanded Stro-1- or Stro-1+ human mesenchymal stem cells into the NOD/SCID mouse and their role in supporting human CD34 cell engraftment. Blood 103, 3313-3319Google Scholar
62Dennis, J.E. et al. (2002) The STRO-1+ marrow cell population is multipotential. Cells Tissues Organs 170, 73-82Google Scholar
63Szilvassy, S.J. et al. (1990) Quantitative assay for totipotent reconstituting hematopoietic stem cells by a competitive repopulation strategy. Proc Natl Acad Sci U S A 87, 8736-8740Google Scholar
64McNiece, I.K. et al. (1990) Colony-forming cells with high proliferative potential (HPP-CFC). Int J Cell Cloning 8, 146-160Google Scholar
65Bhatia, M. et al. (1998) A newly discovered class of human hematopoietic cells with SCID-repopulating activity. Nat Med 4, 1038-1045Google Scholar
66Larochelle, A. et al. (1996) Identification of primitive human hematopoietic cells capable of repopulating NOD/SCID mouse bone marrow: implications for gene therapy. Nat Med 2, 1329-1337CrossRefGoogle ScholarPubMed
67Bonnet, D. (2002) Haematopoietic stem cells. J Pathol 197, 430-440CrossRefGoogle ScholarPubMed
68Larochelle, A. et al. (1995) Engraftment of immune-deficient mice with primitive hematopoietic cells from beta-thalassemia and sickle cell anemia patients: implications for evaluating human gene therapy protocols. Hum Mol Genet 4, 163-172CrossRefGoogle ScholarPubMed
69Cashman, J. et al. (1997) Sustained proliferation, multi-lineage differentiation and maintenance of primitive human haemopoietic cells in NOD/SCID mice transplanted with human cord blood. Br J Haematol 98, 1026-1036Google Scholar
70Horn, P.A. et al. (2003) Distinct hematopoietic stem/progenitor cell populations are responsible for repopulating NOD/SCID mice compared with nonhuman primates. Blood 102, 4329-4335Google Scholar
71Schmidt, M. et al. (2002) Polyclonal long-term repopulating stem cell clones in a primate model. Blood 100, 2737-2743Google Scholar
72Blanpain, C. et al. (2004) Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell 118, 635-648Google Scholar
73Claudinot, S. et al. (2005) Long-term renewal of hair follicles from clonogenic multipotent stem cells. Proc Natl Acad Sci U S A 102, 14677-14682Google Scholar
74Collins, C.A. et al. (2005) Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell 122, 289-301Google Scholar
75Collins, C.A. et al. (2007) A population of myogenic stem cells that survives skeletal muscle aging. Stem Cells 25, 885-894Google Scholar
76Ostenfeld, T. et al. (2000) Human neural precursor cells express low levels of telomerase in vitro and show diminishing cell proliferation with extensive axonal outgrowth following transplantation. Exp Neurol 164, 215-226Google Scholar
77Wright, W.E. and Shay, J.W. (2001) Cellular senescence as a tumor-protection mechanism: the essential role of counting. Curr Opin Genet Dev 11, 98-103Google Scholar
78Wright, W.E. and Shay, J.W. (2002) Historical claims and current interpretations of replicative aging. Nat Biotechnol 20, 682-688Google Scholar
79Smogorzewska, A. and de Lange, T. (2002) Different telomere damage signaling pathways in human and mouse cells. Embo J 21, 4338-4348Google Scholar
80Prowse, K.R. and Greider, C.W. (1995) Developmental and tissue-specific regulation of mouse telomerase and telomere length. Proc Natl Acad Sci U S A 92, 4818-4822Google Scholar
81Blasco, M.A. et al. (1997) Telomere shortening and tumor formation by mouse cells lacking telomerase RNA. Cell 91, 25-34Google Scholar
82Abkowitz, J.L. et al. (2002) Evidence that the number of hematopoietic stem cells per animal is conserved in mammals. Blood 100, 2665-2667Google Scholar
83Parrinello, S. et al. (2003) Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts. Nat Cell Biol 5, 741-747Google Scholar
84Itahana, K., Campisi, J. and Dimri, G.P. (2004) Mechanisms of cellular senescence in human and mouse cells. Biogerontology 5, 1-10Google Scholar
85Samper, E. et al. (2002) Long-term repopulating ability of telomerase-deficient murine hematopoietic stem cells. Blood 99, 2767-2775Google Scholar
86Rudolph, K.L. et al. (1999) Longevity, stress response, and cancer in aging telomerase-deficient mice. Cell 96, 701-712Google Scholar
87Lee, H.W. et al. (1998) Essential role of mouse telomerase in highly proliferative organs. Nature 392, 569-574Google Scholar
88Barlow, C. et al. (1996) Atm-deficient mice: a paradigm of ataxia telangiectasia. Cell 86, 159-171Google Scholar
89Xu, Y. et al. (1996) Targeted disruption of ATM leads to growth retardation, chromosomal fragmentation during meiosis, immune defects, and thymic lymphoma. Genes Dev 10, 2411-2422Google Scholar
90Wong, K.K. et al. (2003) Telomere dysfunction and Atm deficiency compromises organ homeostasis and accelerates ageing. Nature 421, 643-648CrossRefGoogle ScholarPubMed
91Epstein, C.J. (1995) Down syndrome (Trisomy 21) (Scriver, C.A. et al. , series eds), pp. 749-794, McGraw-Hill, New YorkGoogle Scholar
92Nespoli, L. et al. (1993) Immunological features of Down's syndrome: a review. J Intellect Disabil Res 37, 543-551Google Scholar
93Rumble, B. et al. (1989) Amyloid A4 protein and its precursor in Down's syndrome and Alzheimer's disease. N Engl J Med 320, 1446-1452Google Scholar
94Angelopoulou, N. et al. (2000) Bone mineral density and muscle strength in young men with mental retardation (with and without Down syndrome). Calcif Tissue Int 66, 176-180Google Scholar
95Creutzig, U. et al. (1996) Myelodysplasia and acute myelogenous leukemia in Down's syndrome. A report of 40 children of the AML-BFM Study Group. Leukemia 10, 1677-1686Google Scholar
96Vaziri, H. et al. (1993) Loss of telomeric DNA during aging of normal and trisomy 21 human lymphocytes. Am J Hum Genet 52, 661-667Google Scholar
97Holmes, D.K. et al. (2006) Hematopoietic progenitor cell deficiency in fetuses and children affected by Down's syndrome. Exp Hematol 34, 1611-1615Google Scholar
98Flores, I., Cayuela, M.L. and Blasco, M.A. (2005) Effects of telomerase and telomere length on epidermal stem cell behavior. Science 309, 1253-1256Google Scholar
99Ferron, S. et al. (2004) Telomere shortening and chromosomal instability abrogates proliferation of adult but not embryonic neural stem cells. Development 131, 4059-4070Google Scholar
100Prasher, J.M. et al. (2005) Reduced hematopoietic reserves in DNA interstrand crosslink repair-deficient Ercc1-/- mice. Embo J 24, 861-871Google Scholar
101Ito, K. et al. (2006) Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med 12, 446-451Google Scholar
102Stepanova, L. and Sorrentino, B.P. (2005) A limited role for p16Ink4a and p19Arf in the loss of hematopoietic stem cells during proliferative stress. Blood 106, 827-832Google Scholar
103Janzen, V. et al. (2006) Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature 443, 421-426Google Scholar
104Molofsky, A.V. et al. (2006) Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature 443, 448-452Google Scholar
105Yuan, Y. et al. (2004) In vivo self-renewing divisions of haematopoietic stem cells are increased in the absence of the early G1-phase inhibitor, p18INK4C. Nat Cell Biol 6, 436-442Google Scholar
106Yu, H. et al. (2006) Hematopoietic stem cell exhaustion impacted by p18 INK4C and p21 Cip1/Waf1 in opposite manners. Blood 107, 1200-1206Google Scholar
107Choudhury, A.R. et al. (2007) Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation. Nat Genet 39, 99-105Google Scholar
108Cheng, T. et al. (2000) Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 287, 1804-1808Google Scholar
109Kippin, T.E. et al. (2004) Neural stem cells show bidirectional experience-dependent plasticity in the perinatal mammalian brain. J Neurosci 24, 2832-2836Google Scholar
110Dumble, M. et al. (2007) The impact of altered p53 dosage on hematopoietic stem cell dynamics during aging. Blood 109, 1736-1742Google Scholar
111Walkley, C.R. et al. (2005) Negative cell-cycle regulators cooperatively control self-renewal and differentiation of haematopoietic stem cells. Nat Cell Biol 7, 172-178Google Scholar
112Cheng, T. et al. (2000) Stem cell repopulation efficiency but not pool size is governed by p27(kip1). Nat Med 6, 1235-1240Google Scholar
113Zhang, J. et al. (2006) PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention. Nature 441, 518-522Google Scholar
114Lessard, J. and Sauvageau, G. (2003) Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature 423, 255-260Google Scholar
115Molofsky, A.V. et al. (2005) Bmi-1 promotes neural stem cell self-renewal and neural development but not mouse growth and survival by repressing the p16Ink4a and p19Arf senescence pathways. Genes Dev 19, 1432-1437Google Scholar
116Brun, A.C. et al. (2004) Hoxb4-deficient mice undergo normal hematopoietic development but exhibit a mild proliferation defect in hematopoietic stem cells. Blood 103, 4126-4133Google Scholar
117Bjornsson, J.M. et al. (2003) Reduced proliferative capacity of hematopoietic stem cells deficient in Hoxb3 and Hoxb4. Mol Cell Biol 23, 3872-3883Google Scholar
118Miyake, N. et al. (2006) HOXB4-induced self-renewal of hematopoietic stem cells is significantly enhanced by p21 deficiency. Stem Cells 24, 653-661Google Scholar
119Walkley, C.R. and Orkin, S.H. (2006) Rb is dispensable for self-renewal and multilineage differentiation of adult hematopoietic stem cells. Proc Natl Acad Sci U S A 103, 9057-9062Google Scholar
120Chien, W.M. et al. (2006) Genetic mosaics reveal both cell-autonomous and cell-nonautonomous function of murine p27Kip1. Proc Natl Acad Sci U S A 103, 4122-4127Google Scholar
121Sudo, K. et al. (2000) Age-associated characteristics of murine hematopoietic stem cells. J Exp Med 192, 1273-1280Google Scholar
122Pearce, D.J. et al. (2006) Age-dependent increase in side population distribution within hematopoiesis: implications for our understanding of the mechanism of aging. Stem Cells 5, 828-835Google Scholar
123Morrison, S.J. et al. (1996) The aging of hematopoietic stem cells. Nat Med 2, 1011-1016Google Scholar
124Gibson, M.C. and Schultz, E. (1983) Age-related differences in absolute numbers of skeletal muscle satellite cells. Muscle Nerve 6, 574-580Google Scholar
125Conboy, I.M. et al. (2003) Notch-mediated restoration of regenerative potential to aged muscle. Science 302, 1575-1577Google Scholar
126Brack, A.S., Bildsoe, H. and 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. J Cell Sci 118, 4813-4821Google Scholar
127Ogden, D.A. and Mickliem, H.S. (1976) The fate of serially transplanted bone marrow cell populations from young and old donors. Transplantation 22, 287-293Google Scholar
128Liang, Y., Van Zant, G. and Szilvassy, S.J. (2005) Effects of aging on the homing and engraftment of murine hematopoietic stem and progenitor cells. Blood 106, 1479-1487Google Scholar
129Baxter, M.A. et al. (2004) Study of telomere length reveals rapid aging of human marrow stromal cells following in vitro expansion. Stem Cells 22, 675-682Google Scholar
130Lipschitz, D.A., Mitchell, C.O. and Thompson, C. (1981) The anemia of senescence. Am J Hematol 11, 47-54Google Scholar
131Harstock, R.J., Smith, E.B. and Ketter, C.S. (1965) Normal variation with aging of the amount of hematopoietic tissue in bone marrow from anterior iliac crest. Am J Clin Pathol 43, 325-333Google Scholar
132Bagnara, G.P. et al. (2000) Hemopoiesis in healthy old people and centenarians: well-maintained responsiveness of CD34+ cells to hemopoietic growth factors and remodeling of cytokine network. J Gerontol A Biol Sci Med Sci 55, B61-66; discussion B67-70Google Scholar
133Ho, A.D., Wagner, W. and Mahlknecht, U. (2005) Stem cells and ageing. The potential of stem cells to overcome age-related deteriorations of the body in regenerative medicine. EMBO Rep 6 Spec No, S35-38Google Scholar
134Gale, R.E. et al. (1997) Acquired skewing of X-chromosome inactivation patterns in myeloid cells of the elderly suggests stochastic clonal loss with age. Br J Haematol 98, 512-519Google Scholar
135Morrison, S.J. et al. (1996) Telomerase activity in hematopoietic cells is associated with self-renewal potential. Immunity 5, 207-216Google Scholar
136Lansdorp, P.M. (1995) Telomere length and proliferation potential of hematopoietic stem cells. J Cell Sci 108, 1-6Google Scholar
137Rufer, N. et al. (1998) Telomere length dynamics in human lymphocyte subpopulations measured by flow cytometry. Nat Biotechnol 16, 743-747Google Scholar
138Vaziri, H. et al. (1994) Evidence for a mitotic clock in human hematopoietic stem cells: loss of telomeric DNA with age. Proc Natl Acad Sci U S A 91, 9857-9860Google Scholar
139Wynn, R. et al. (1999) Telomere shortening in leucocyte subsets of long-term survivors of allogeneic bone marrow transplantation. Br J Haematol 105, 997-1001Google Scholar
140Notaro, R. et al. (1997) In vivo telomere dynamics of human hematopoietic stem cells. Proc Natl Acad Sci U S A 94, 13782-13785Google Scholar
141Allsopp, R.C. et al. (2003) Telomerase is required to slow telomere shortening and extend replicative lifespan of HSCs during serial transplantation. Blood 102, 517-520Google Scholar
142Mason, P.J., Wilson, D.B. and Bessler, M. (2005) Dyskeratosis congenita – a disease of dysfunctional telomere maintenance. Curr Mol Med 5, 159-170Google Scholar
143Cawthon, R.M. et al. (2003) Association between telomere length in blood and mortality in people aged 60 years or older. Lancet 361, 393-395Google Scholar
144Kruger, G.M. and Morrison, S.J. (2002) Brain repair by endogenous progenitors. Cell 110, 399-402Google Scholar
145De Felice, L. et al. (2005) Histone deacetylase inhibitor valproic acid enhances the cytokine-induced expansion of human hematopoietic stem cells. Cancer Res 65, 1505-1513Google Scholar
146Bug, G. et al. (2005) Valproic acid stimulates proliferation and self-renewal of hematopoietic stem cells. Cancer Res 65, 2537-2541Google Scholar
147Chen, S. et al. (2006) Self-renewal of embryonic stem cells by a small molecule. Proc Natl Acad Sci U S A 103, 17266-17271Google Scholar
148Trowbridge, J.J. et al. (2006) Glycogen synthase kinase-3 is an in vivo regulator of hematopoietic stem cell repopulation. Nat Med 12, 89-98Google Scholar
149Sato, N. et al. (2004) Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat Med 10, 55-63Google Scholar
150Halaschek-Wiener, J. and Brooks-Wilson, A. (2007) Progeria of stem cells: stem cell exhaustion in Hutchinson-Gilford progeria syndrome. J Gerontol A Biol Sci Med Sci 62, 3-8Google Scholar
151Ramirez, C.L et al. (2007) Human progeroid syndromes, aging and cancer: new genetic and epigenetic insights into old questions. Cell Mol Life Sci 64, 155-170Google Scholar
152Kudlow, B.A., Kennedy, B.K. and Monnat, R.J. Jr. (2007) Werner and Hutchinson-Gilford progeria syndromes: mechanistic basis of human progeroid diseases. Nat Rev Mol Cell Biol 8, 394-404Google Scholar
153Conboy, I.M. et al. (2005) Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433, 760-764Google Scholar

Further reading, resources and contacts

The International Society for Stem Cell Research (ISSCR) website has excellent links on stem cells:

Abcam, Miltenyi Biotech, Stem Cell Technologies and BD Biosciences have antibody and specialised reagents for the isolation and culture of murine and human stem cells:

http://www.nia.nih.gov/ (National Institute on Aging)Google Scholar
http://www.buckinstitute.org/site/ (Buck Institute for Age Research)Google Scholar
http://www.barshop.uthscsa.edu/ (The Sam and Ann Barshop Institute for Longevity and Aging Studies)Google Scholar
http://www.nia.nih.gov/ (National Institute on Aging)Google Scholar
http://www.buckinstitute.org/site/ (Buck Institute for Age Research)Google Scholar
http://www.barshop.uthscsa.edu/ (The Sam and Ann Barshop Institute for Longevity and Aging Studies)Google Scholar