Can ends justify the means?: telomeres and the mechanisms of replicative senescence and immortalization in mammalian cells.

Finite replicative lifespan, or senescence, of mammalian cells in culture is a phenomenon that has generated much curiosity since its description. The obvious significance of senescence to organismal aging and the development of cancer has engendered a long-lasting and lively debate about its mechanisms. Recent discoveries concerning the phenotypes of telomerase knockout mice, the consequences of telomerase reexpression in somatic cells, and genes that regulate senescence have provided striking molecular insights but also have uncovered important new questions. The objective of this review is to reconcile old observations with new molecular details and to focus attention on the key remaining puzzles.

[1]  M. Bibby Telomeres and Telomerase. Ciba Foundation Symposium 211 , 1999, British Journal of Cancer.

[2]  Robert A. Weinberg,et al.  Expression of TERT in early premalignant lesions and a subset of cells in normal tissues , 1998, Nature Genetics.

[3]  H. Niida,et al.  Severe growth defect in mouse cells lacking the telomerase RNA component , 1998, Nature Genetics.

[4]  A. Conn,et al.  Two regions of simian virus 40 large T-antigen independently extend the life span of primary C57BL/6 mouse embryo fibroblasts and cooperate in immortalization. , 1998, Virology.

[5]  L. Chin,et al.  Expression of mouse telomerase reverse transcriptase during development, differentiation and proliferation , 1998, Oncogene.

[6]  D. Sidransky,et al.  Role of the p16 tumor suppressor gene in cancer. , 1998, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[7]  S. Benchimol,et al.  Reconstitution of telomerase activity in normal human cells leads to elongation of telomeres and extended replicative life span , 1998, Current Biology.

[8]  J. Herman,et al.  Hypermethylation can selectively silence individual p16ink4A alleles in neoplasia. , 1998, Cancer research.

[9]  P. Söderkvist,et al.  Inactivations of p16INK4a-α, p16INK4a-β and p15INK4b genes in 2′, 3′-dideoxycytidine- and 1,3-butadiene-induced murine lymphomas , 1998, Oncogene.

[10]  Bas van Steensel,et al.  TRF2 Protects Human Telomeres from End-to-End Fusions , 1998, Cell.

[11]  V. Gouyer,et al.  Mechanisms of p16INK4A inactivation in non small-cell lung cancers , 1998, Oncogene.

[12]  C. Harley,et al.  Extension of life-span by introduction of telomerase into normal human cells. , 1998, Science.

[13]  Richard A. Ashmun,et al.  Tumor Suppression at the Mouse INK4a Locus Mediated by the Alternative Reading Frame Product p19 ARF , 1997, Cell.

[14]  T. Ide,et al.  Abnormal telomere dynamics of B-lymphoblastoid cell strains from Werner's syndrome patients transformed by Epstein – Barr virus , 1997, Oncogene.

[15]  María A Blasco,et al.  Telomere Shortening and Tumor Formation by Mouse Cells Lacking Telomerase RNA , 1997, Cell.

[16]  T. Davison,et al.  ATM‐dependent telomere loss in aging human diploid fibroblasts and DNA damage lead to the post‐translational activation of p53 protein involving poly(ADP‐ribose) polymerase , 1997, The EMBO journal.

[17]  Wenyi Wei,et al.  Bypass of senescence after disruption of p21CIP1/WAF1 gene in normal diploid human fibroblasts. , 1997, Science.

[18]  K. Prowse,et al.  Telomere length regulation during postnatal development and ageing in Mus spretus. , 1997, Nucleic acids research.

[19]  H. Tanke,et al.  Telomeres in the mouse have large inter-chromosomal variations in the number of T2AG3 repeats. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[20]  J. Shay,et al.  A survey of telomerase activity in human cancer. , 1997, European journal of cancer.

[21]  H. Varmus,et al.  Telomerase activation in mouse mammary tumors: lack of detectable telomere shortening and evidence for regulation of telomerase RNA with cell proliferation , 1996, Molecular and cellular biology.

[22]  S. Benchimol,et al.  From telomere loss to p53 induction and activation of a DNA-damage pathway at senescence: The telomere loss/DNA damage model of cell aging , 1996, Experimental Gerontology.

[23]  L. Chin,et al.  Role of the INK4a Locus in Tumor Suppression and Cell Mortality , 1996, Cell.

[24]  J. McDougall,et al.  Telomerase activation by the E6 gene product of human papillomavirus type 16 , 1996, Nature.

[25]  L. Naeger,et al.  Activation of the endogenous p53 growth inhibitory pathway in HeLa cervical carcinoma cells by expression of the bovine papillomavirus E2 gene. , 1996, Oncogene.

[26]  C. Harley,et al.  Telomerase activity in normal and malignant murine tissues. , 1995, Oncogene.

[27]  A. Levine,et al.  The Spectrum of Mutations at the p53 Locus , 1995, Annals of the New York Academy of Sciences.

[28]  Stephen J. Elledge,et al.  Mice Lacking p21 CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control , 1995, Cell.

[29]  C. Greider,et al.  Developmental and tissue-specific regulation of mouse telomerase and telomere length. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[30]  C B Harley,et al.  Specific association of human telomerase activity with immortal cells and cancer. , 1994, Science.

[31]  P. Jat,et al.  The biological clock that measures the mitotic life-span of mouse embryo fibroblasts continues to function in the presence of simian virus 40 large tumor antigen. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[32]  C. Harley,et al.  Stabilization of short telomeres and telomerase activity accompany immortalization of Epstein-Barr virus-transformed human B lymphocytes , 1994, Journal of virology.

[33]  T. Lange,et al.  Activation of telomerase in a human tumor. , 1994 .

[34]  M. Kastan,et al.  DNA strand breaks: the DNA template alterations that trigger p53-dependent DNA damage response pathways , 1994, Molecular and cellular biology.

[35]  S. Barber,et al.  Restoration of telomeres in human papillomavirus-immortalized human anogenital epithelial cells , 1994, Molecular and cellular biology.

[36]  T. Kondo,et al.  Immortalization of normal human cells is a multistep process and a rate limiting step of neoplastic transformation of the cells. , 1993, Human cell.

[37]  J. Shay,et al.  The frequency of immortalization of human fibroblasts and mammary epithelial cells transfected with SV40 large T-antigen. , 1993, Experimental cell research.

[38]  Sanjay K. Chhablani,et al.  Silent domains are assembled continuously from the telomere and are defined by promoter distance and strength, and by SIR3 dosage. , 1993, Genes & development.

[39]  C B Harley,et al.  Telomere length predicts replicative capacity of human fibroblasts. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[40]  J. Shay,et al.  Telomere positional effects and the regulation of cellular senescence. , 1992, Trends in genetics : TIG.

[41]  J. Steitz,et al.  Telomere shortening associated with chromosome instability is arrested in immortal cells which express telomerase activity. , 1992, The EMBO journal.

[42]  D. Murasko,et al.  Extension of lifespan of human T lymphocytes by transfection with SV40 large T antigen. , 1992, Experimental cell research.

[43]  J. Shay,et al.  Defining the molecular mechanisms of human cell immortalization. , 1991, Biochimica et biophysica acta.

[44]  C B Harley,et al.  Telomere loss: mitotic clock or genetic time bomb? , 1991, Mutation research.

[45]  D. Kaufman,et al.  Extended life span of human endometrial stromal cells transfected with cloned origin-defective, temperature-sensitive simian virus 40 , 1991, Journal of virology.

[46]  S. Goldstein Replicative senescence: the human fibroblast comes of age. , 1990, Science.

[47]  C. Harley,et al.  Telomeres shorten during ageing of human fibroblasts , 1990, Nature.

[48]  S. Jazwinski Aging and senescence of the budding yeast Saccharomyces cerevisiae , 1990, Molecular microbiology.

[49]  Edward L. Schneider,et al.  Handbook of the Biology of Aging , 1990 .

[50]  J. Shay,et al.  Quantitation of the frequency of immortalization of normal human diploid fibroblasts by SV40 large T-antigen. , 1989, Experimental cell research.

[51]  J. Shay,et al.  Reversible cellular senescence: implications for immortalization of normal human diploid fibroblasts , 1989, Molecular and cellular biology.

[52]  F. Traganos,et al.  Growth of immortal simian virus 40 tsA-transformed human fibroblasts is temperature dependent , 1989, Molecular and cellular biology.

[53]  P. Sharp,et al.  Cell lines established by a temperature-sensitive simian virus 40 large-T-antigen gene are growth restricted at the nonpermissive temperature , 1989, Molecular and cellular biology.

[54]  J. R. Smith,et al.  Genetic analysis of indefinite division in human cells: identification of four complementation groups. , 1988, Proceedings of the National Academy of Sciences of the United States of America.

[55]  K. Dittmann,et al.  Human skin fibroblasts in vitro differentiate along a terminal cell lineage. , 1988, Proceedings of the National Academy of Sciences of the United States of America.

[56]  J. Mccormick,et al.  Towards an understanding of the malignant transformation of diploid human fibroblasts. , 1988, Mutation research.

[57]  M. Stampfer,et al.  Human mammary epithelial cells in culture: differentiation and transformation. , 1988, Cancer treatment and research.

[58]  A. Henderson,et al.  Immortalization of human fibroblasts transformed by origin-defective simian virus 40 , 1987, Molecular and cellular biology.

[59]  D. Loo,et al.  Extended culture of mouse embryo cells without senescence: inhibition by serum. , 1987, Science.

[60]  B. Stanulis-Praeger Cellular Senescence revisited: a review , 1987, Mechanisms of Ageing and Development.

[61]  R. Weinberg,et al.  Behavior of myc and ras oncogenes in transformation of rat embryo fibroblasts , 1986, Molecular and cellular biology.

[62]  J. R. Smith,et al.  Existence of high abundance antiproliferative mRNA's in senescent human diploid fibroblasts. , 1986, Science.

[63]  S. A. Bruce,et al.  In vitro senescence of Syrian hamster mesenchymal cells of fetal to aged adult origin. Inverse relationship between in vivo donor age and in vitro proliferative capacity , 1986, Mechanisms of Ageing and Development.

[64]  G. Stein,et al.  Relationship of finite proliferative lifespan, senescence, and quiescence in human cells , 1985, Journal of cellular physiology.

[65]  T. Ide,et al.  Progress of aging in human diploid cells transformed with a tsA mutant of simian virus 40. , 1984, Experimental cell research.

[66]  R. Kamen,et al.  Expression of the large T protein of polyoma virus promotes the establishment in culture of "normal" rodent fibroblast cell lines. , 1983, Proceedings of the National Academy of Sciences of the United States of America.

[67]  R. Newbold,et al.  Induction of immortality is an early event in malignant transformation of mammalian cells by carcinogens , 1982, Nature.

[68]  D. Röhme Evidence for a relationship between longevity of mammalian species and life spans of normal fibroblasts in vitro and erythrocytes in vivo. , 1981, Proceedings of the National Academy of Sciences of the United States of America.

[69]  R. Meek,et al.  Establishment of rat embryonic cells in vitro. Relationship of DNA synthesis, senescence, and acquisition of unlimited growth potential. , 1980, Experimental cell research.

[70]  T. Matsumura,et al.  Senescent human diploid cells in culture: survival, DNA synthesis and morphology. , 1979, Journal of gerontology.

[71]  S. Goldstein Human Genetic Disorders That Feature Premature Onset and Accelerated Progression of Biological Aging , 1978 .

[72]  M. Dinowitz A continuous line of Rous sarcoma virus-transformed chick embryo cells. , 1977, Journal of the National Cancer Institute.

[73]  H. Green,et al.  Epidermal growth factor and the multiplication of cultured human epidermal keratinocytes , 1977, Nature.

[74]  E. Schneider,et al.  The relationship between in vitro cellular aging and in vivo human age. , 1976, Proceedings of the National Academy of Sciences of the United States of America.

[75]  T. Norwood,et al.  Dominance of the senescent phenotype in heterokaryons between replicative and post-replicative human fibroblast-like cells. , 1974, Proceedings of the National Academy of Sciences of the United States of America.

[76]  S. Goldstein Aging in vitro: Growth of cultured cells from the Galapagos tortoise , 1974 .

[77]  A M Olovnikov,et al.  A theory of marginotomy. The incomplete copying of template margin in enzymic synthesis of polynucleotides and biological significance of the phenomenon. , 1973, Journal of theoretical biology.

[78]  L. Orgel,et al.  Ageing of Clones of Mammalian Cells , 1973, Nature.

[79]  J. Pontén The growth capacity of normal and Rous‐virus‐transformed chicken fibroblasts in vitro , 1970, International journal of cancer.

[80]  H. Green,et al.  QUANTITATIVE STUDIES OF THE GROWTH OF MOUSE EMBRYO CELLS IN CULTURE AND THEIR DEVELOPMENT INTO ESTABLISHED LINES , 1963, The Journal of cell biology.

[81]  L. Hayflick,et al.  The serial cultivation of human diploid cell strains. , 1961, Experimental cell research.

[82]  B. Mcclintock,et al.  The Fusion of Broken Ends of Chromosomes Following Nuclear Fusion. , 1942, Proceedings of the National Academy of Sciences of the United States of America.