메뉴 건너뛰기




Volumn 28, Issue 2, 2015, Pages 51-64

Cellular senescence: a hitchhiker’s guide

Author keywords

Cellular senescence; Oncogene induced senescence; Replicative senescence; Senescence in embryogenesis; Stress induced senescence

Indexed keywords

PROTEIN P53; RETINOBLASTOMA PROTEIN; TELOMERASE;

EID: 84939975178     PISSN: 09147470     EISSN: 17490774     Source Type: Journal    
DOI: 10.1007/s13577-015-0110-x     Document Type: Article
Times cited : (56)

References (209)
  • 1
    • 34548186667 scopus 로고    scopus 로고
    • Cellular senescence: when bad things happen to good cells
    • COI: 1:CAS:528:DC%2BD2sXpsVartLg%3D, PID: 1766795
    • Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007;8(9):729–40.
    • (2007) Nat Rev Mol Cell Biol , vol.8 , Issue.9 , pp. 729-740
    • Campisi, J.1    d’Adda di Fagagna, F.2
  • 2
    • 0035545802 scopus 로고    scopus 로고
    • Putting the stress on senescence
    • COI: 1:CAS:528:DC%2BD3MXotFWht7s%3D, PID: 1169819
    • Serrano M, Blasco MA. Putting the stress on senescence. Curr Opin Cell Biol. 2001;13(6):748–53.
    • (2001) Curr Opin Cell Biol , vol.13 , Issue.6 , pp. 748-753
    • Serrano, M.1    Blasco, M.A.2
  • 3
    • 14644410453 scopus 로고    scopus 로고
    • The signals and pathways activating cellular senescence
    • COI: 1:CAS:528:DC%2BD2MXhvVGjs7c%3D, PID: 1574367
    • Ben-Porath I, Weinberg RA. The signals and pathways activating cellular senescence. Int J Biochem Cell Biol. 2005;37(5):961–76.
    • (2005) Int J Biochem Cell Biol , vol.37 , Issue.5 , pp. 961-976
    • Ben-Porath, I.1    Weinberg, R.A.2
  • 4
    • 34447543913 scopus 로고    scopus 로고
    • Cellular senescence in cancer and aging
    • COI: 1:CAS:528:DC%2BD2sXovFaltrc%3D, PID: 1766293
    • Collado M, Blasco MA, Serrano M. Cellular senescence in cancer and aging. Cell. 2007;130(2):223–33.
    • (2007) Cell , vol.130 , Issue.2 , pp. 223-233
    • Collado, M.1    Blasco, M.A.2    Serrano, M.3
  • 5
    • 8344230643 scopus 로고    scopus 로고
    • Senescent fibroblasts resist apoptosis by downregulating caspase-3
    • COI: 1:CAS:528:DC%2BD2cXpslGnu7c%3D, PID: 1554177
    • Marcotte R, Lacelle C, Wang E. Senescent fibroblasts resist apoptosis by downregulating caspase-3. Mech Ageing Dev. 2004;125(10–11):777–83.
    • (2004) Mech Ageing Dev , vol.125 , Issue.10-11 , pp. 777-783
    • Marcotte, R.1    Lacelle, C.2    Wang, E.3
  • 6
    • 33644825358 scopus 로고    scopus 로고
    • Apoptosis resistance of senescent human fibroblasts is correlated with the absence of nuclear IGFBP-3
    • COI: 1:CAS:528:DC%2BD28Xpt1Wg, PID: 1630048
    • Hampel B, et al. Apoptosis resistance of senescent human fibroblasts is correlated with the absence of nuclear IGFBP-3. Aging Cell. 2005;4(6):325–30.
    • (2005) Aging Cell , vol.4 , Issue.6 , pp. 325-330
    • Hampel, B.1
  • 7
    • 13944270339 scopus 로고    scopus 로고
    • Senescent cells, tumor suppression, and organismal aging: good citizens, bad neighbors
    • COI: 1:CAS:528:DC%2BD2MXitVWnu74%3D, PID: 1573468
    • Campisi J. Senescent cells, tumor suppression, and organismal aging: good citizens, bad neighbors. Cell. 2005;120(4):513–22.
    • (2005) Cell , vol.120 , Issue.4 , pp. 513-522
    • Campisi, J.1
  • 8
    • 33748327046 scopus 로고    scopus 로고
    • p16INK4A is a robust in vivo biomarker of cellular aging in human skin
    • COI: 1:CAS:528:DC%2BD28XhtFSisb3O, PID: 1691156
    • Ressler S, et al. p16INK4A is a robust in vivo biomarker of cellular aging in human skin. Aging Cell. 2006;5(5):379–89.
    • (2006) Aging Cell , vol.5 , Issue.5 , pp. 379-389
    • Ressler, S.1
  • 9
    • 33644692330 scopus 로고    scopus 로고
    • Cellular senescence in aging primates
    • COI: 1:CAS:528:DC%2BD28XitV2hsbk%3D, PID: 1645603
    • Herbig U, et al. Cellular senescence in aging primates. Science. 2006;311(5765):1257.
    • (2006) Science , vol.311 , Issue.5765 , pp. 1257
    • Herbig, U.1
  • 10
    • 33846090789 scopus 로고    scopus 로고
    • Accumulation of senescent cells in mitotic tissue of aging primates
    • COI: 1:CAS:528:DC%2BD2sXmtVWruw%3D%3D, PID: 1711631
    • Jeyapalan JC, et al. Accumulation of senescent cells in mitotic tissue of aging primates. Mech Ageing Dev. 2007;128(1):36–44.
    • (2007) Mech Ageing Dev , vol.128 , Issue.1 , pp. 36-44
    • Jeyapalan, J.C.1
  • 11
    • 23244447037 scopus 로고    scopus 로고
    • BRAFE600-associated senescence-like cell cycle arrest of human naevi
    • COI: 1:CAS:528:DC%2BD2MXmvFenuro%3D, PID: 1607985
    • Michaloglou C, et al. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature. 2005;436(7051):720–4.
    • (2005) Nature , vol.436 , Issue.7051 , pp. 720-724
    • Michaloglou, C.1
  • 12
    • 78650243948 scopus 로고    scopus 로고
    • Impact of cellular senescence signature on ageing research
    • COI: 1:CAS:528:DC%2BC3cXhsFyktrbI, PID: 2094697
    • Sikora E, et al. Impact of cellular senescence signature on ageing research. Ageing Res Rev. 2011;10(1):146–52.
    • (2011) Ageing Res Rev , vol.10 , Issue.1 , pp. 146-152
    • Sikora, E.1
  • 13
    • 0025731583 scopus 로고
    • Structure and function of telomeres
    • COI: 1:CAS:528:DyaK3MXisVWnsLg%3D, PID: 170811
    • Blackburn EH. Structure and function of telomeres. Nature. 1991;350(6319):569–73.
    • (1991) Nature , vol.350 , Issue.6319 , pp. 569-573
    • Blackburn, E.H.1
  • 14
    • 0030731928 scopus 로고    scopus 로고
    • Normal human chromosomes have long G-rich telomeric overhangs at one end
    • COI: 1:CAS:528:DyaK2sXntlOjsb0%3D, PID: 935325
    • Wright WE, et al. Normal human chromosomes have long G-rich telomeric overhangs at one end. Genes Dev. 1997;11(21):2801–9.
    • (1997) Genes Dev , vol.11 , Issue.21 , pp. 2801-2809
    • Wright, W.E.1
  • 15
    • 0030982721 scopus 로고    scopus 로고
    • The terminal DNA structure of mammalian chromosomes
    • COI: 1:CAS:528:DyaK2sXktlCnsbg%3D, PID: 921881
    • McElligott R, Wellinger RJ. The terminal DNA structure of mammalian chromosomes. EMBO J. 1997;16(12):3705–14.
    • (1997) EMBO J , vol.16 , Issue.12 , pp. 3705-3714
    • McElligott, R.1    Wellinger, R.J.2
  • 16
    • 0035929353 scopus 로고    scopus 로고
    • Switching and signaling at the telomere
    • COI: 1:CAS:528:DC%2BD3MXnslGkt7c%3D, PID: 1157277
    • Blackburn EH. Switching and signaling at the telomere. Cell. 2001;106(6):661–73.
    • (2001) Cell , vol.106 , Issue.6 , pp. 661-673
    • Blackburn, E.H.1
  • 17
    • 44349136891 scopus 로고    scopus 로고
    • Telomere dysfunction and tumour suppression: the senescence connection
    • COI: 1:CAS:528:DC%2BD1cXmt1Oksb8%3D, PID: 1850024
    • Deng Y, Chan SS, Chang S. Telomere dysfunction and tumour suppression: the senescence connection. Nat Rev Cancer. 2008;8(6):450–8.
    • (2008) Nat Rev Cancer , vol.8 , Issue.6 , pp. 450-458
    • Deng, Y.1    Chan, S.S.2    Chang, S.3
  • 18
    • 0027993344 scopus 로고
    • Genetic determination of telomere size in humans: a twin study of three age groups
    • COI: 1:STN:280:DyaK2M%2Fmsl2msA%3D%3D, PID: 797734
    • Slagboom PE, Droog S, Boomsma DI. Genetic determination of telomere size in humans: a twin study of three age groups. Am J Hum Genet. 1994;55(5):876–82.
    • (1994) Am J Hum Genet , vol.55 , Issue.5 , pp. 876-882
    • Slagboom, P.E.1    Droog, S.2    Boomsma, D.I.3
  • 19
    • 0037726517 scopus 로고    scopus 로고
    • The pattern of chromosome-specific variations in telomere length in humans is determined by inherited, telomere-near factors and is maintained throughout life
    • COI: 1:CAS:528:DC%2BD3sXjsVWmtr8%3D, PID: 1273590
    • Graakjaer J, et al. The pattern of chromosome-specific variations in telomere length in humans is determined by inherited, telomere-near factors and is maintained throughout life. Mech Ageing Dev. 2003;124(5):629–40.
    • (2003) Mech Ageing Dev , vol.124 , Issue.5 , pp. 629-640
    • Graakjaer, J.1
  • 21
    • 0026460896 scopus 로고
    • Telomere length predicts replicative capacity of human fibroblasts
    • COI: 1:CAS:528:DyaK3sXhvF2luw%3D%3D, PID: 143819
    • Allsopp RC, et al. Telomere length predicts replicative capacity of human fibroblasts. Proc Natl Acad Sci USA. 1992;89(21):10114–8.
    • (1992) Proc Natl Acad Sci USA , vol.89 , Issue.21 , pp. 10114-10118
    • Allsopp, R.C.1
  • 22
    • 0022402513 scopus 로고
    • Identification of a specific telomere terminal transferase activity in Tetrahymena extracts
    • COI: 1:CAS:528:DyaL28Xot12itQ%3D%3D, PID: 390785
    • Greider CW, Blackburn EH. Identification of a specific telomere terminal transferase activity in Tetrahymena extracts. Cell. 1985;43(2 Pt 1):405–13.
    • (1985) Cell , vol.43 , pp. 405-413
    • Greider, C.W.1    Blackburn, E.H.2
  • 23
    • 0028564951 scopus 로고
    • Specific association of human telomerase activity with immortal cells and cancer
    • COI: 1:CAS:528:DyaK2MXisl2lu7g%3D, PID: 760542
    • Kim NW, et al. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266(5193):2011–5.
    • (1994) Science , vol.266 , Issue.5193 , pp. 2011-2015
    • Kim, N.W.1
  • 24
    • 0029872174 scopus 로고    scopus 로고
    • Telomerase activity in human germline and embryonic tissues and cells
    • COI: 1:CAS:528:DyaK28Xis1Ort7o%3D, PID: 893487
    • Wright WE, et al. Telomerase activity in human germline and embryonic tissues and cells. Dev Genet. 1996;18(2):173–9.
    • (1996) Dev Genet , vol.18 , Issue.2 , pp. 173-179
    • Wright, W.E.1
  • 25
    • 0344622606 scopus 로고
    • The serial cultivation of human diploid cell strains
    • COI: 1:STN:280:DyaF38%2FkslSgtA%3D%3D, PID: 1390565
    • Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.
    • (1961) Exp Cell Res , vol.25 , pp. 585-621
    • Hayflick, L.1    Moorhead, P.S.2
  • 26
    • 0025279931 scopus 로고
    • Telomeres shorten during ageing of human fibroblasts
    • COI: 1:CAS:528:DyaK3cXksFKntb4%3D, PID: 234257
    • Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of human fibroblasts. Nature. 1990;345(6274):458–60.
    • (1990) Nature , vol.345 , Issue.6274 , pp. 458-460
    • Harley, C.B.1    Futcher, A.B.2    Greider, C.W.3
  • 27
    • 0033942779 scopus 로고    scopus 로고
    • Role of oxidative stress in telomere length regulation and replicative senescence
    • von Zglinicki T. Role of oxidative stress in telomere length regulation and replicative senescence. Ann N Y Acad Sci. 2000;908:99–110.
    • (2000) Ann N Y Acad Sci , vol.908 , pp. 99-110
    • von Zglinicki, T.1
  • 28
    • 0037382414 scopus 로고    scopus 로고
    • Erosion of the telomeric single-strand overhang at replicative senescence
    • COI: 1:CAS:528:DC%2BD3sXisVGitrY%3D, PID: 1265229
    • Stewart SA, et al. Erosion of the telomeric single-strand overhang at replicative senescence. Nat Genet. 2003;33(4):492–6.
    • (2003) Nat Genet , vol.33 , Issue.4 , pp. 492-496
    • Stewart, S.A.1
  • 29
    • 0344441890 scopus 로고    scopus 로고
    • A DNA damage checkpoint response in telomere-initiated senescence
    • PID: 1460836
    • d’Adda di Fagagna F. A DNA damage checkpoint response in telomere-initiated senescence. Nature. 2003;426(6963):194–8.
    • (2003) Nature , vol.426 , Issue.6963 , pp. 194-198
    • d’Adda di Fagagna, F.1
  • 30
    • 0029939848 scopus 로고    scopus 로고
    • Heterogeneity in telomere length of human chromosomes
    • COI: 1:CAS:528:DyaK28XivVOjt7s%3D, PID: 873313
    • Lansdorp PM, et al. Heterogeneity in telomere length of human chromosomes. Hum Mol Genet. 1996;5(5):685–91.
    • (1996) Hum Mol Genet , vol.5 , Issue.5 , pp. 685-691
    • Lansdorp, P.M.1
  • 31
    • 0029936681 scopus 로고    scopus 로고
    • Models of initiation of replicative senescence by loss of telomeric DNA
    • COI: 1:CAS:528:DyaK28XitlansLg%3D, PID: 870679
    • Allsopp RC. Models of initiation of replicative senescence by loss of telomeric DNA. Exp Gerontol. 1996;31(1–2):235–43.
    • (1996) Exp Gerontol , vol.31 , Issue.1-2 , pp. 235-243
    • Allsopp, R.C.1
  • 32
    • 0035812845 scopus 로고    scopus 로고
    • The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability
    • COI: 1:CAS:528:DC%2BD3MXnsFOntLs%3D, PID: 1159518
    • Hemann MT, et al. The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability. Cell. 2001;107(1):67–77.
    • (2001) Cell , vol.107 , Issue.1 , pp. 67-77
    • Hemann, M.T.1
  • 33
    • 84881071211 scopus 로고    scopus 로고
    • The length of the shortest telomere as the major determinant of the onset of replicative senescence
    • COI: 1:CAS:528:DC%2BC3sXhsV2lu77P, PID: 2373378
    • Xu Z, et al. The length of the shortest telomere as the major determinant of the onset of replicative senescence. Genetics. 2013;194:847.
    • (2013) Genetics , vol.194 , pp. 847
    • Xu, Z.1
  • 34
    • 0029127226 scopus 로고
    • Mild hyperoxia shortens telomeres and inhibits proliferation of fibroblasts: a model for senescence?
    • von Zglinicki T, et al. Mild hyperoxia shortens telomeres and inhibits proliferation of fibroblasts: a model for senescence? Exp Cell Res. 1995;220(1):186–93.
    • (1995) Exp Cell Res , vol.220 , Issue.1 , pp. 186-193
    • von Zglinicki, T.1
  • 35
    • 0042195820 scopus 로고    scopus 로고
    • Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts
    • COI: 1:CAS:528:DC%2BD3sXlvV2gt78%3D, PID: 1285595
    • Parrinello S, et al. Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts. Nat Cell Biol. 2003;5(8):741–7.
    • (2003) Nat Cell Biol , vol.5 , Issue.8 , pp. 741-747
    • Parrinello, S.1
  • 36
    • 0033583242 scopus 로고    scopus 로고
    • Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species
    • COI: 1:CAS:528:DyaK1MXitFSkuro%3D, PID: 1007568
    • Lee AC, et al. Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. J Biol Chem. 1999;274(12):7936–40.
    • (1999) J Biol Chem , vol.274 , Issue.12 , pp. 7936-7940
    • Lee, A.C.1
  • 37
    • 0032524563 scopus 로고    scopus 로고
    • Molecular analysis of H2O2-induced senescent-like growth arrest in normal human fibroblasts: p53 and Rb control G1 arrest but not cell replication
    • COI: 1:CAS:528:DyaK1cXjslSgt74%3D, PID: 957684
    • Chen QM, et al. Molecular analysis of H2O2-induced senescent-like growth arrest in normal human fibroblasts: p53 and Rb control G1 arrest but not cell replication. Biochem J. 1998;332(Pt 1):43–50.
    • (1998) Biochem J , vol.332 , pp. 43-50
    • Chen, Q.M.1
  • 38
    • 0033673408 scopus 로고    scopus 로고
    • Protein oxidation and degradation during cellular senescence of human BJ fibroblasts: part I–effects of proliferative senescence
    • COI: 1:CAS:528:DC%2BD3MXht1SntL4%3D, PID: 1109946
    • Sitte N, et al. Protein oxidation and degradation during cellular senescence of human BJ fibroblasts: part I–effects of proliferative senescence. FASEB J. 2000;14(15):2495–502.
    • (2000) FASEB J , vol.14 , Issue.15 , pp. 2495-2502
    • Sitte, N.1
  • 39
    • 0033674181 scopus 로고    scopus 로고
    • Protein oxidation and degradation during cellular senescence of human BJ fibroblasts: part II–aging of nondividing cells
    • COI: 1:CAS:528:DC%2BD3MXht1SntL8%3D, PID: 1109946
    • Sitte N, et al. Protein oxidation and degradation during cellular senescence of human BJ fibroblasts: part II–aging of nondividing cells. FASEB J. 2000;14(15):2503–10.
    • (2000) FASEB J , vol.14 , Issue.15 , pp. 2503-2510
    • Sitte, N.1
  • 40
    • 0029051426 scopus 로고
    • Oxidative DNA damage and senescence of human diploid fibroblast cells
    • COI: 1:CAS:528:DyaK2MXls1alu78%3D, PID: 775380
    • Chen Q, et al. Oxidative DNA damage and senescence of human diploid fibroblast cells. Proc Natl Acad Sci USA. 1995;92(10):4337–41.
    • (1995) Proc Natl Acad Sci USA , vol.92 , Issue.10 , pp. 4337-4341
    • Chen, Q.1
  • 41
    • 0028345161 scopus 로고
    • Senescence-like growth arrest induced by hydrogen peroxide in human diploid fibroblast F65 cells
    • COI: 1:CAS:528:DyaK2cXktVarurY%3D, PID: 818388
    • Chen Q, Ames BN. Senescence-like growth arrest induced by hydrogen peroxide in human diploid fibroblast F65 cells. Proc Natl Acad Sci USA. 1994;91(10):4130–4.
    • (1994) Proc Natl Acad Sci USA , vol.91 , Issue.10 , pp. 4130-4134
    • Chen, Q.1    Ames, B.N.2
  • 42
    • 0035951791 scopus 로고    scopus 로고
    • Subcytotoxic H2O2 stress triggers a release of transforming growth factor-beta 1, which induces biomarkers of cellular senescence of human diploid fibroblasts
    • COI: 1:CAS:528:DC%2BD3MXhtVGjsLw%3D, PID: 1106029
    • Frippiat C, et al. Subcytotoxic H2O2 stress triggers a release of transforming growth factor-beta 1, which induces biomarkers of cellular senescence of human diploid fibroblasts. J Biol Chem. 2001;276(4):2531–7.
    • (2001) J Biol Chem , vol.276 , Issue.4 , pp. 2531-2537
    • Frippiat, C.1
  • 43
    • 84858794576 scopus 로고    scopus 로고
    • Ginsenoside Rb1 reverses H2O2-induced senescence in human umbilical endothelial cells: involvement of eNOS pathway
    • COI: 1:CAS:528:DC%2BC38XjsVCnsLk%3D, PID: 2203089
    • Liu DH, et al. Ginsenoside Rb1 reverses H2O2-induced senescence in human umbilical endothelial cells: involvement of eNOS pathway. J Cardiovasc Pharmacol. 2012;59(3):222–30.
    • (2012) J Cardiovasc Pharmacol , vol.59 , Issue.3 , pp. 222-230
    • Liu, D.H.1
  • 44
    • 79955613626 scopus 로고    scopus 로고
    • Proteomic and metabolomic analysis of H2O2-induced premature senescent human mesenchymal stem cells
    • COI: 1:CAS:528:DC%2BC3MXlvFSrurg%3D, PID: 2138246
    • Kim JS, et al. Proteomic and metabolomic analysis of H2O2-induced premature senescent human mesenchymal stem cells. Exp Gerontol. 2011;46(6):500–10.
    • (2011) Exp Gerontol , vol.46 , Issue.6 , pp. 500-510
    • Kim, J.S.1
  • 45
    • 0033913425 scopus 로고    scopus 로고
    • Replicative senescence and oxidant-induced premature senescence. Beyond the control of cell cycle checkpoints
    • COI: 1:CAS:528:DC%2BD3cXlsVOjs7k%3D, PID: 1091195
    • Chen QM. Replicative senescence and oxidant-induced premature senescence. Beyond the control of cell cycle checkpoints. Ann N Y Acad Sci. 2000;908:111–25.
    • (2000) Ann N Y Acad Sci , vol.908 , pp. 111-125
    • Chen, Q.M.1
  • 46
    • 0034472888 scopus 로고    scopus 로고
    • Involvement of Rb family proteins, focal adhesion proteins and protein synthesis in senescent morphogenesis induced by hydrogen peroxide
    • COI: 1:CAS:528:DC%2BD3MXjt1Sg, PID: 1105809
    • Chen QM, et al. Involvement of Rb family proteins, focal adhesion proteins and protein synthesis in senescent morphogenesis induced by hydrogen peroxide. J Cell Sci. 2000;113(Pt 22):4087–97.
    • (2000) J Cell Sci , vol.113 , pp. 4087-4097
    • Chen, Q.M.1
  • 47
    • 84949116585 scopus 로고    scopus 로고
    • The senescent hepatocyte gene signature in chronic liver disease
    • COI: 1:CAS:528:DC%2BC2cXhsFyrt7fE, PID: 2524068
    • Aravinthan A, et al. The senescent hepatocyte gene signature in chronic liver disease. Exp Gerontol. 2014;60:37–45.
    • (2014) Exp Gerontol , vol.60 , pp. 37-45
    • Aravinthan, A.1
  • 48
    • 0000726641 scopus 로고    scopus 로고
    • Sequence-specific DNA cleavage by Fe2+-mediated fenton reactions has possible biological implications
    • COI: 1:CAS:528:DyaK1MXmt1aqtg%3D%3D, PID: 987303
    • Henle ES, et al. Sequence-specific DNA cleavage by Fe2+-mediated fenton reactions has possible biological implications. J Biol Chem. 1999;274(2):962–71.
    • (1999) J Biol Chem , vol.274 , Issue.2 , pp. 962-971
    • Henle, E.S.1
  • 49
    • 0032772792 scopus 로고    scopus 로고
    • Site-specific DNA damage at GGG sequence by oxidative stress may accelerate telomere shortening
    • COI: 1:CAS:528:DyaK1MXkt1ags7o%3D, PID: 1040517
    • Oikawa S, Kawanishi S. Site-specific DNA damage at GGG sequence by oxidative stress may accelerate telomere shortening. FEBS Lett. 1999;453(3):365–8.
    • (1999) FEBS Lett , vol.453 , Issue.3 , pp. 365-368
    • Oikawa, S.1    Kawanishi, S.2
  • 50
    • 0035901505 scopus 로고    scopus 로고
    • Site-specific DNA damage at the GGG sequence by UVA involves acceleration of telomere shortening
    • COI: 1:CAS:528:DC%2BD3MXitVyrsrY%3D, PID: 1129464
    • Oikawa S, Tada-Oikawa S, Kawanishi S. Site-specific DNA damage at the GGG sequence by UVA involves acceleration of telomere shortening. Biochemistry. 2001;40(15):4763–8.
    • (2001) Biochemistry , vol.40 , Issue.15 , pp. 4763-4768
    • Oikawa, S.1    Tada-Oikawa, S.2    Kawanishi, S.3
  • 51
    • 84859352385 scopus 로고    scopus 로고
    • Telomeres are favoured targets of a persistent DNA damage response in ageing and stress-induced senescence
    • PID: 2242622
    • Hewitt G, et al. Telomeres are favoured targets of a persistent DNA damage response in ageing and stress-induced senescence. Nat Commun. 2012;3:708.
    • (2012) Nat Commun , vol.3 , pp. 708
    • Hewitt, G.1
  • 52
    • 0033954684 scopus 로고    scopus 로고
    • Accumulation of single-strand breaks is the major cause of telomere shortening in human fibroblasts
    • von Zglinicki T, Pilger R, Sitte N. Accumulation of single-strand breaks is the major cause of telomere shortening in human fibroblasts. Free Radic Biol Med. 2000;28(1):64–74.
    • (2000) Free Radic Biol Med , vol.28 , Issue.1 , pp. 64-74
    • von Zglinicki, T.1    Pilger, R.2    Sitte, N.3
  • 53
    • 0031821009 scopus 로고    scopus 로고
    • Preferential accumulation of single-stranded regions in telomeres of human fibroblasts
    • COI: 1:CAS:528:DyaK1cXhs1yqt7s%3D, PID: 951173
    • Petersen S, Saretzki G, von Zglinicki T. Preferential accumulation of single-stranded regions in telomeres of human fibroblasts. Exp Cell Res. 1998;239(1):152–60.
    • (1998) Exp Cell Res , vol.239 , Issue.1 , pp. 152-160
    • Petersen, S.1    Saretzki, G.2    von Zglinicki, T.3
  • 54
    • 3242696917 scopus 로고    scopus 로고
    • Mechanism of telomere shortening by oxidative stress
    • COI: 1:CAS:528:DC%2BD2cXms12gu74%3D, PID: 1524702
    • Kawanishi S, Oikawa S. Mechanism of telomere shortening by oxidative stress. Ann N Y Acad Sci. 2004;1019:278–84.
    • (2004) Ann N Y Acad Sci , vol.1019 , pp. 278-284
    • Kawanishi, S.1    Oikawa, S.2
  • 55
    • 0036236630 scopus 로고    scopus 로고
    • Replicative aging, telomeres, and oxidative stress
    • COI: 1:CAS:528:DC%2BD38XktFWqtLc%3D, PID: 1197618
    • Saretzki G, Von Zglinicki T. Replicative aging, telomeres, and oxidative stress. Ann N Y Acad Sci. 2002;959:24–9.
    • (2002) Ann N Y Acad Sci , vol.959 , pp. 24-29
    • Saretzki, G.1    Von Zglinicki, T.2
  • 56
    • 0036629255 scopus 로고    scopus 로고
    • Oxidative stress shortens telomeres
    • von Zglinicki T. Oxidative stress shortens telomeres. Trends Biochem Sci. 2002;27(7):339–44.
    • (2002) Trends Biochem Sci , vol.27 , Issue.7 , pp. 339-344
    • von Zglinicki, T.1
  • 57
    • 84859372986 scopus 로고    scopus 로고
    • Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation
    • COI: 1:CAS:528:DC%2BC38XktVars7g%3D, PID: 2242607
    • Fumagalli M, et al. Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat Cell Biol. 2012;14(4):355–65.
    • (2012) Nat Cell Biol , vol.14 , Issue.4 , pp. 355-365
    • Fumagalli, M.1
  • 58
    • 0038209424 scopus 로고    scopus 로고
    • BRCA1 shifts p53-mediated cellular outcomes towards irreversible growth arrest
    • COI: 1:CAS:528:DC%2BD3sXksV2jtbk%3D, PID: 1280228
    • Ongusaha PP, et al. BRCA1 shifts p53-mediated cellular outcomes towards irreversible growth arrest. Oncogene. 2003;22(24):3749–58.
    • (2003) Oncogene , vol.22 , Issue.24 , pp. 3749-3758
    • Ongusaha, P.P.1
  • 59
    • 0034690249 scopus 로고    scopus 로고
    • Interplay of p53 and DNA-repair protein XRCC4 in tumorigenesis, genomic stability and development
    • COI: 1:CAS:528:DC%2BD3cXjtVCrurw%3D, PID: 1078679
    • Gao Y, et al. Interplay of p53 and DNA-repair protein XRCC4 in tumorigenesis, genomic stability and development. Nature. 2000;404(6780):897–900.
    • (2000) Nature , vol.404 , Issue.6780 , pp. 897-900
    • Gao, Y.1
  • 60
    • 0034280093 scopus 로고    scopus 로고
    • Mammalian Ku86 protein prevents telomeric fusions independently of the length of TTAGGG repeats and the G-strand overhang
    • COI: 1:CAS:528:DC%2BD3cXntVaqsbs%3D, PID: 1125660
    • Samper E, et al. Mammalian Ku86 protein prevents telomeric fusions independently of the length of TTAGGG repeats and the G-strand overhang. EMBO Rep. 2000;1(3):244–52.
    • (2000) EMBO Rep , vol.1 , Issue.3 , pp. 244-252
    • Samper, E.1
  • 61
    • 0030944985 scopus 로고    scopus 로고
    • Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a
    • COI: 1:CAS:528:DyaK2sXhvVSit7k%3D, PID: 905449
    • Serrano M, et al. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell. 1997;88(5):593–602.
    • (1997) Cell , vol.88 , Issue.5 , pp. 593-602
    • Serrano, M.1
  • 62
    • 4644266046 scopus 로고    scopus 로고
    • Ha-Ras(G12 V) induces senescence in primary and immortalized human esophageal keratinocytes with p53 dysfunction
    • COI: 1:CAS:528:DC%2BD2cXntFGnu7g%3D, PID: 1527372
    • Takaoka M, et al. Ha-Ras(G12 V) induces senescence in primary and immortalized human esophageal keratinocytes with p53 dysfunction. Oncogene. 2004;23(40):6760–8.
    • (2004) Oncogene , vol.23 , Issue.40 , pp. 6760-6768
    • Takaoka, M.1
  • 63
    • 0032192153 scopus 로고    scopus 로고
    • Senescence of human fibroblasts induced by oncogenic Raf
    • COI: 1:CAS:528:DyaK1cXmsleju7w%3D, PID: 976520
    • Zhu J, et al. Senescence of human fibroblasts induced by oncogenic Raf. Genes Dev. 1998;12(19):2997–3007.
    • (1998) Genes Dev , vol.12 , Issue.19 , pp. 2997-3007
    • Zhu, J.1
  • 64
    • 23244461708 scopus 로고    scopus 로고
    • Oncogene-induced senescence as an initial barrier in lymphoma development
    • COI: 1:CAS:528:DC%2BD2MXmvFentbY%3D, PID: 1607983
    • Braig M, et al. Oncogene-induced senescence as an initial barrier in lymphoma development. Nature. 2005;436(7051):660–5.
    • (2005) Nature , vol.436 , Issue.7051 , pp. 660-665
    • Braig, M.1
  • 65
    • 23244460597 scopus 로고    scopus 로고
    • Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis
    • COI: 1:CAS:528:DC%2BD2MXmvFenu78%3D, PID: 1607985
    • Chen Z, et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature. 2005;436(7051):725–30.
    • (2005) Nature , vol.436 , Issue.7051 , pp. 725-730
    • Chen, Z.1
  • 66
    • 23244447893 scopus 로고    scopus 로고
    • Tumour biology: senescence in premalignant tumours
    • COI: 1:CAS:528:DC%2BD2MXmvFentLc%3D, PID: 1607983
    • Collado M, et al. Tumour biology: senescence in premalignant tumours. Nature. 2005;436(7051):642.
    • (2005) Nature , vol.436 , Issue.7051 , pp. 642
    • Collado, M.1
  • 67
    • 0033552813 scopus 로고    scopus 로고
    • The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus
    • COI: 1:CAS:528:DyaK1MXntVygtQ%3D%3D, PID: 992367
    • Jacobs JJ, et al. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature. 1999;397(6715):164–8.
    • (1999) Nature , vol.397 , Issue.6715 , pp. 164-168
    • Jacobs, J.J.1
  • 68
    • 0029778901 scopus 로고    scopus 로고
    • Human fibroblast commitment to a senescence-like state in response to histone deacetylase inhibitors is cell cycle dependent
    • COI: 1:CAS:528:DyaK28XltFKjtbw%3D, PID: 875667
    • Ogryzko VV, et al. Human fibroblast commitment to a senescence-like state in response to histone deacetylase inhibitors is cell cycle dependent. Mol Cell Biol. 1996;16(9):5210–8.
    • (1996) Mol Cell Biol , vol.16 , Issue.9 , pp. 5210-5218
    • Ogryzko, V.V.1
  • 69
    • 0001510491 scopus 로고    scopus 로고
    • The RB and p53 pathways in cancer
    • COI: 1:CAS:528:DC%2BD38XmslSks7s%3D, PID: 1220453
    • Sherr CJ, McCormick F. The RB and p53 pathways in cancer. Cancer Cell. 2002;2(2):103–12.
    • (2002) Cancer Cell , vol.2 , Issue.2 , pp. 103-112
    • Sherr, C.J.1    McCormick, F.2
  • 70
    • 0035195557 scopus 로고    scopus 로고
    • The evolution of diverse biological responses to DNA damage: insights from yeast and p53
    • COI: 1:CAS:528:DC%2BD3MXpt1Ggu74%3D, PID: 1178158
    • Wahl GM, Carr AM. The evolution of diverse biological responses to DNA damage: insights from yeast and p53. Nat Cell Biol. 2001;3(12):E277–86.
    • (2001) Nat Cell Biol , vol.3 , Issue.12 , pp. E277-E286
    • Wahl, G.M.1    Carr, A.M.2
  • 71
    • 0032549711 scopus 로고    scopus 로고
    • ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways
    • COI: 1:CAS:528:DyaK1cXit1KlsLw%3D, PID: 952924
    • Zhang Y, Xiong Y, Yarbrough WG. ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways. Cell. 1998;92(6):725–34.
    • (1998) Cell , vol.92 , Issue.6 , pp. 725-734
    • Zhang, Y.1    Xiong, Y.2    Yarbrough, W.G.3
  • 72
    • 0027359827 scopus 로고
    • WAF1, a potential mediator of p53 tumor suppression
    • COI: 1:CAS:528:DyaK2cXpslGguw%3D%3D, PID: 824275
    • el-Deiry WS, et al. WAF1, a potential mediator of p53 tumor suppression. Cell. 1993;75(4):817–25.
    • (1993) Cell , vol.75 , Issue.4 , pp. 817-825
    • el-Deiry, W.S.1
  • 73
    • 0028220204 scopus 로고
    • p53-dependent inhibition of cyclin-dependent kinase activities in human fibroblasts during radiation-induced G1 arrest
    • COI: 1:CAS:528:DyaK2cXitl2hs7k%3D, PID: 813742
    • Dulic V, et al. p53-dependent inhibition of cyclin-dependent kinase activities in human fibroblasts during radiation-induced G1 arrest. Cell. 1994;76(6):1013–23.
    • (1994) Cell , vol.76 , Issue.6 , pp. 1013-1023
    • Dulic, V.1
  • 74
    • 0027496935 scopus 로고
    • The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases
    • COI: 1:CAS:528:DyaK2cXhtVShsLk%3D, PID: 824275
    • Harper JW, et al. The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell. 1993;75(4):805–16.
    • (1993) Cell , vol.75 , Issue.4 , pp. 805-816
    • Harper, J.W.1
  • 75
    • 0028177401 scopus 로고
    • p53-dependent G1 arrest involves pRB-related proteins and is disrupted by the human papillomavirus 16 E7 oncoprotein
    • COI: 1:CAS:528:DyaK2cXksFCqs70%3D, PID: 820248
    • Slebos RJ, et al. p53-dependent G1 arrest involves pRB-related proteins and is disrupted by the human papillomavirus 16 E7 oncoprotein. Proc Natl Acad Sci USA. 1994;91(12):5320–4.
    • (1994) Proc Natl Acad Sci USA , vol.91 , Issue.12 , pp. 5320-5324
    • Slebos, R.J.1
  • 76
    • 0037667702 scopus 로고    scopus 로고
    • Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence
    • COI: 1:CAS:528:DC%2BD3sXkvVekt7g%3D, PID: 1280960
    • Narita M, et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003;113(6):703–16.
    • (2003) Cell , vol.113 , Issue.6 , pp. 703-716
    • Narita, M.1
  • 77
    • 0033635597 scopus 로고    scopus 로고
    • Ablation of the retinoblastoma gene family deregulates G(1) control causing immortalization and increased cell turnover under growth-restricting conditions
    • COI: 1:CAS:528:DC%2BD3cXptVWlt70%3D, PID: 1111489
    • Dannenberg JH, et al. Ablation of the retinoblastoma gene family deregulates G(1) control causing immortalization and increased cell turnover under growth-restricting conditions. Genes Dev. 2000;14(23):3051–64.
    • (2000) Genes Dev , vol.14 , Issue.23 , pp. 3051-3064
    • Dannenberg, J.H.1
  • 78
    • 0033637086 scopus 로고    scopus 로고
    • Targeted disruption of the three Rb-related genes leads to loss of G(1) control and immortalization
    • COI: 1:CAS:528:DC%2BD3cXptVWlt7w%3D, PID: 1111489
    • Sage J, et al. Targeted disruption of the three Rb-related genes leads to loss of G(1) control and immortalization. Genes Dev. 2000;14(23):3037–50.
    • (2000) Genes Dev , vol.14 , Issue.23 , pp. 3037-3050
    • Sage, J.1
  • 79
    • 0042420304 scopus 로고    scopus 로고
    • DNA damage foci at dysfunctional telomeres
    • COI: 1:CAS:528:DC%2BD3sXntVKhsrc%3D, PID: 1295695
    • Takai H, Smogorzewska A, de Lange T. DNA damage foci at dysfunctional telomeres. Curr Biol. 2003;13(17):1549–56.
    • (2003) Curr Biol , vol.13 , Issue.17 , pp. 1549-1556
    • Takai, H.1    Smogorzewska, A.2    de Lange, T.3
  • 80
    • 2442511752 scopus 로고    scopus 로고
    • Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a)
    • COI: 1:CAS:528:DC%2BD2cXksl2jsrk%3D, PID: 1514959
    • Herbig U, et al. Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Mol Cell. 2004;14(4):501–13.
    • (2004) Mol Cell , vol.14 , Issue.4 , pp. 501-513
    • Herbig, U.1
  • 81
    • 0037102234 scopus 로고    scopus 로고
    • Different telomere damage signaling pathways in human and mouse cells
    • COI: 1:CAS:528:DC%2BD38Xms1Kntbk%3D, PID: 1216963
    • Smogorzewska A, de Lange T. Different telomere damage signaling pathways in human and mouse cells. EMBO J. 2002;21(16):4338–48.
    • (2002) EMBO J , vol.21 , Issue.16 , pp. 4338-4348
    • Smogorzewska, A.1    de Lange, T.2
  • 82
    • 0346873027 scopus 로고    scopus 로고
    • Loss of retinoblastoma but not p16 function allows bypass of replicative senescence in human fibroblasts
    • COI: 1:CAS:528:DC%2BD3sXosFajt7c%3D, PID: 1456632
    • Wei W, et al. Loss of retinoblastoma but not p16 function allows bypass of replicative senescence in human fibroblasts. EMBO Rep. 2003;4(11):1061–6.
    • (2003) EMBO Rep , vol.4 , Issue.11 , pp. 1061-1066
    • Wei, W.1
  • 83
    • 33845892109 scopus 로고    scopus 로고
    • Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation
    • COI: 1:CAS:528:DC%2BD28XhtlGktLjP, PID: 1714328
    • Choudhury AR, et al. Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation. Nat Genet. 2007;39(1):99–105.
    • (2007) Nat Genet , vol.39 , Issue.1 , pp. 99-105
    • Choudhury, A.R.1
  • 84
    • 4444278440 scopus 로고    scopus 로고
    • p14ARF is a component of the p53 response following ionizing irradiation of normal human fibroblasts
    • COI: 1:CAS:528:DC%2BD2cXmsVKltbk%3D, PID: 1519514
    • Khan S, et al. p14ARF is a component of the p53 response following ionizing irradiation of normal human fibroblasts. Oncogene. 2004;23(36):6040–6.
    • (2004) Oncogene , vol.23 , Issue.36 , pp. 6040-6046
    • Khan, S.1
  • 85
    • 0032485213 scopus 로고    scopus 로고
    • Agents that cause DNA double strand breaks lead to p16INK4a enrichment and the premature senescence of normal fibroblasts
    • COI: 1:CAS:528:DyaK1cXhvV2lt7k%3D, PID: 952885
    • Robles SJ, Adami GR. Agents that cause DNA double strand breaks lead to p16INK4a enrichment and the premature senescence of normal fibroblasts. Oncogene. 1998;16(9):1113–23.
    • (1998) Oncogene , vol.16 , Issue.9 , pp. 1113-1123
    • Robles, S.J.1    Adami, G.R.2
  • 86
    • 0031594707 scopus 로고    scopus 로고
    • p16INK4A participates in a G1 arrest checkpoint in response to DNA damage
    • COI: 1:CAS:528:DyaK1cXivFymuw%3D%3D, PID: 941888
    • Shapiro GI, et al. p16INK4A participates in a G1 arrest checkpoint in response to DNA damage. Mol Cell Biol. 1998;18(1):378–87.
    • (1998) Mol Cell Biol , vol.18 , Issue.1 , pp. 378-387
    • Shapiro, G.I.1
  • 87
    • 0345932931 scopus 로고    scopus 로고
    • Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway
    • COI: 1:CAS:528:DC%2BD3sXht1amsrg%3D, PID: 1258115
    • Iwasa H, Han J, Ishikawa F. Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway. Genes Cells. 2003;8(2):131–44.
    • (2003) Genes Cells , vol.8 , Issue.2 , pp. 131-144
    • Iwasa, H.1    Han, J.2    Ishikawa, F.3
  • 88
    • 1942469306 scopus 로고    scopus 로고
    • Mechanisms of cell-cycle arrest in Spitz nevi with constitutive activation of the MAP-kinase pathway
    • COI: 1:CAS:528:DC%2BD2cXns1Gqu7c%3D, PID: 1511132
    • Maldonado JL, et al. Mechanisms of cell-cycle arrest in Spitz nevi with constitutive activation of the MAP-kinase pathway. Am J Pathol. 2004;164(5):1783–7.
    • (2004) Am J Pathol , vol.164 , Issue.5 , pp. 1783-1787
    • Maldonado, J.L.1
  • 89
    • 0038457477 scopus 로고    scopus 로고
    • Human melanocyte senescence and melanoma susceptibility genes
    • COI: 1:CAS:528:DC%2BD3sXkt12qtbk%3D, PID: 1278928
    • Bennett DC. Human melanocyte senescence and melanoma susceptibility genes. Oncogene. 2003;22(20):3063–9.
    • (2003) Oncogene , vol.22 , Issue.20 , pp. 3063-3069
    • Bennett, D.C.1
  • 90
    • 0036242247 scopus 로고    scopus 로고
    • Sequential activation of the MEK-extracellular signal-regulated kinase and MKK3/6-p38 mitogen-activated protein kinase pathways mediates oncogenic ras-induced premature senescence
    • PID: 1197197
    • Wang W, et al. Sequential activation of the MEK-extracellular signal-regulated kinase and MKK3/6-p38 mitogen-activated protein kinase pathways mediates oncogenic ras-induced premature senescence. Mol Cell Biol. 2002;22(10):3389–403.
    • (2002) Mol Cell Biol , vol.22 , Issue.10 , pp. 3389-3403
    • Wang, W.1
  • 91
    • 77149164811 scopus 로고    scopus 로고
    • Feedback between p21 and reactive oxygen production is necessary for cell senescence
    • PID: 2016070
    • Passos JF, et al. Feedback between p21 and reactive oxygen production is necessary for cell senescence. Mol Syst Biol. 2010;6:347.
    • (2010) Mol Syst Biol , vol.6 , pp. 347
    • Passos, J.F.1
  • 92
    • 33845269825 scopus 로고    scopus 로고
    • Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication
    • PID: 1713609
    • Di Micco R, et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature. 2006;444(7119):638–42.
    • (2006) Nature , vol.444 , Issue.7119 , pp. 638-642
    • Di Micco, R.1
  • 93
    • 84889600900 scopus 로고    scopus 로고
    • Programmed cell senescence during mammalian embryonic development
    • COI: 1:CAS:528:DC%2BC3sXhslyqu7rN, PID: 2423896
    • Munoz-Espin D, et al. Programmed cell senescence during mammalian embryonic development. Cell. 2013;155(5):1104–18.
    • (2013) Cell , vol.155 , Issue.5 , pp. 1104-1118
    • Munoz-Espin, D.1
  • 94
    • 84889565689 scopus 로고    scopus 로고
    • Senescence is a developmental mechanism that contributes to embryonic growth and patterning
    • COI: 1:CAS:528:DC%2BC3sXhsl2jsrzE, PID: 2423896
    • Storer M, et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell. 2013;155(5):1119–30.
    • (2013) Cell , vol.155 , Issue.5 , pp. 1119-1130
    • Storer, M.1
  • 95
    • 33745823159 scopus 로고    scopus 로고
    • The ATM-mediated DNA-damage response: taking shape
    • COI: 1:CAS:528:DC%2BD28XntFWrsLo%3D, PID: 1677483
    • Shiloh Y. The ATM-mediated DNA-damage response: taking shape. Trends Biochem Sci. 2006;31(7):402–10.
    • (2006) Trends Biochem Sci , vol.31 , Issue.7 , pp. 402-410
    • Shiloh, Y.1
  • 96
    • 70350504453 scopus 로고    scopus 로고
    • The DNA-damage response in human biology and disease
    • COI: 1:CAS:528:DC%2BD1MXhtlGktLrK, PID: 1984725
    • Jackson SP, Bartek J. The DNA-damage response in human biology and disease. Nature. 2009;461(7267):1071–8.
    • (2009) Nature , vol.461 , Issue.7267 , pp. 1071-1078
    • Jackson, S.P.1    Bartek, J.2
  • 97
    • 45849099050 scopus 로고    scopus 로고
    • Living on a break: cellular senescence as a DNA-damage response
    • PID: 1857446
    • d’Adda di Fagagna F. Living on a break: cellular senescence as a DNA-damage response. Nat Rev Cancer. 2008;8(7):512–22.
    • (2008) Nat Rev Cancer , vol.8 , Issue.7 , pp. 512-522
    • d’Adda di Fagagna, F.1
  • 98
    • 34247251276 scopus 로고    scopus 로고
    • Single- and double-stranded DNA: building a trigger of ATR-mediated DNA damage response
    • COI: 1:CAS:528:DC%2BD2sXkslKjtL0%3D, PID: 1743799
    • Zou L. Single- and double-stranded DNA: building a trigger of ATR-mediated DNA damage response. Genes Dev. 2007;21(8):879–85.
    • (2007) Genes Dev , vol.21 , Issue.8 , pp. 879-885
    • Zou, L.1
  • 99
    • 0037012845 scopus 로고    scopus 로고
    • Genomic instability in mice lacking histone H2AX
    • COI: 1:CAS:528:DC%2BD38XjsFyms7o%3D, PID: 1193498
    • Celeste A, et al. Genomic instability in mice lacking histone H2AX. Science. 2002;296(5569):922–7.
    • (2002) Science , vol.296 , Issue.5569 , pp. 922-927
    • Celeste, A.1
  • 100
    • 0037468192 scopus 로고    scopus 로고
    • MDC1 is a mediator of the mammalian DNA damage checkpoint
    • COI: 1:CAS:528:DC%2BD3sXhsVKgtbo%3D, PID: 1260700
    • Stewart GS, et al. MDC1 is a mediator of the mammalian DNA damage checkpoint. Nature. 2003;421(6926):961–6.
    • (2003) Nature , vol.421 , Issue.6926 , pp. 961-966
    • Stewart, G.S.1
  • 101
    • 0036906303 scopus 로고    scopus 로고
    • Checkpoint signalling: focusing on 53BP1
    • COI: 1:CAS:528:DC%2BD38XptFKmtrw%3D, PID: 1246152
    • Abraham RT. Checkpoint signalling: focusing on 53BP1. Nat Cell Biol. 2002;4(12):E277–9.
    • (2002) Nat Cell Biol , vol.4 , Issue.12 , pp. E277-E279
    • Abraham, R.T.1
  • 102
    • 0035941021 scopus 로고    scopus 로고
    • ATR and ATRIP: partners in checkpoint signaling
    • COI: 1:CAS:528:DC%2BD3MXoslCrsLg%3D, PID: 1172105
    • Cortez D, et al. ATR and ATRIP: partners in checkpoint signaling. Science. 2001;294(5547):1713–6.
    • (2001) Science , vol.294 , Issue.5547 , pp. 1713-1716
    • Cortez, D.1
  • 103
    • 33847779345 scopus 로고    scopus 로고
    • ATM and ATR: components of an integrated circuit
    • COI: 1:CAS:528:DC%2BD2sXnvFCltrw%3D, PID: 1731239
    • Hurley PJ, Bunz F. ATM and ATR: components of an integrated circuit. Cell Cycle. 2007;6(4):414–7.
    • (2007) Cell Cycle , vol.6 , Issue.4 , pp. 414-417
    • Hurley, P.J.1    Bunz, F.2
  • 104
    • 33344464960 scopus 로고    scopus 로고
    • ATM regulates ATR chromatin loading in response to DNA double-strand breaks
    • COI: 1:CAS:528:DC%2BD28XhslCltrw%3D, PID: 1646133
    • Cuadrado M, et al. ATM regulates ATR chromatin loading in response to DNA double-strand breaks. J Exp Med. 2006;203(2):297–303.
    • (2006) J Exp Med , vol.203 , Issue.2 , pp. 297-303
    • Cuadrado, M.1
  • 105
    • 30344463835 scopus 로고    scopus 로고
    • ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks
    • COI: 1:CAS:528:DC%2BD28Xmt1al, PID: 1632778
    • Jazayeri A, et al. ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks. Nat Cell Biol. 2006;8(1):37–45.
    • (2006) Nat Cell Biol , vol.8 , Issue.1 , pp. 37-45
    • Jazayeri, A.1
  • 106
    • 33845715977 scopus 로고    scopus 로고
    • ATR-dependent phosphorylation and activation of ATM in response to UV treatment or replication fork stalling
    • COI: 1:CAS:528:DC%2BD28XhtlShsb3J, PID: 1712449
    • Stiff T, et al. ATR-dependent phosphorylation and activation of ATM in response to UV treatment or replication fork stalling. EMBO J. 2006;25(24):5775–82.
    • (2006) EMBO J , vol.25 , Issue.24 , pp. 5775-5782
    • Stiff, T.1
  • 107
    • 0037341599 scopus 로고    scopus 로고
    • Distinct spatiotemporal dynamics of mammalian checkpoint regulators induced by DNA damage
    • COI: 1:CAS:528:DC%2BD3sXitFKmtr4%3D, PID: 1259890
    • Lukas C, et al. Distinct spatiotemporal dynamics of mammalian checkpoint regulators induced by DNA damage. Nat Cell Biol. 2003;5(3):255–60.
    • (2003) Nat Cell Biol , vol.5 , Issue.3 , pp. 255-260
    • Lukas, C.1
  • 108
    • 33646117239 scopus 로고    scopus 로고
    • Spatial organization of the mammalian genome surveillance machinery in response to DNA strand breaks
    • COI: 1:CAS:528:DC%2BD28XktVCgu70%3D, PID: 1661881
    • Bekker-Jensen S, et al. Spatial organization of the mammalian genome surveillance machinery in response to DNA strand breaks. J Cell Biol. 2006;173(2):195–206.
    • (2006) J Cell Biol , vol.173 , Issue.2 , pp. 195-206
    • Bekker-Jensen, S.1
  • 109
    • 0034717309 scopus 로고    scopus 로고
    • Rapid destruction of human Cdc25A in response to DNA damage
    • COI: 1:CAS:528:DC%2BD3cXjslGrsbs%3D, PID: 1082795
    • Mailand N, et al. Rapid destruction of human Cdc25A in response to DNA damage. Science. 2000;288(5470):1425–9.
    • (2000) Science , vol.288 , Issue.5470 , pp. 1425-1429
    • Mailand, N.1
  • 110
    • 0035939844 scopus 로고    scopus 로고
    • Activation of p53 transcriptional activity requires ATM’s kinase domain and multiple N-terminal serine residues of p53
    • COI: 1:CAS:528:DC%2BD3MXmslGmurY%3D, PID: 1152649
    • Turenne GA, et al. Activation of p53 transcriptional activity requires ATM’s kinase domain and multiple N-terminal serine residues of p53. Oncogene. 2001;20(37):5100–10.
    • (2001) Oncogene , vol.20 , Issue.37 , pp. 5100-5110
    • Turenne, G.A.1
  • 111
    • 33746618049 scopus 로고    scopus 로고
    • gammaH2AX as a checkpoint maintenance signal
    • COI: 1:CAS:528:DC%2BD28XpvF2ru7s%3D, PID: 1685538
    • Downey M, Durocher D. gammaH2AX as a checkpoint maintenance signal. Cell Cycle. 2006;5(13):1376–81.
    • (2006) Cell Cycle , vol.5 , Issue.13 , pp. 1376-1381
    • Downey, M.1    Durocher, D.2
  • 112
    • 66149167336 scopus 로고    scopus 로고
    • DNA damage response and cellular senescence in tissues of aging mice
    • COI: 1:CAS:528:DC%2BD1MXntFGqu7w%3D, PID: 1962727
    • Wang C, et al. DNA damage response and cellular senescence in tissues of aging mice. Aging Cell. 2009;8(3):311–23.
    • (2009) Aging Cell , vol.8 , Issue.3 , pp. 311-323
    • Wang, C.1
  • 113
    • 0038052805 scopus 로고    scopus 로고
    • The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer
    • COI: 1:CAS:528:DC%2BD3sXmtVOitr4%3D, PID: 1281443
    • Vermeulen K, Van Bockstaele DR, Berneman ZN. The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer. Cell Prolif. 2003;36(3):131–49.
    • (2003) Cell Prolif , vol.36 , Issue.3 , pp. 131-149
    • Vermeulen, K.1    Van Bockstaele, D.R.2    Berneman, Z.N.3
  • 114
    • 0033588882 scopus 로고    scopus 로고
    • The INK4 family of cell cycle inhibitors in cancer
    • COI: 1:CAS:528:DyaK1MXms1Onsbk%3D, PID: 1049888
    • Roussel MF. The INK4 family of cell cycle inhibitors in cancer. Oncogene. 1999;18(38):5311–7.
    • (1999) Oncogene , vol.18 , Issue.38 , pp. 5311-5317
    • Roussel, M.F.1
  • 115
    • 0028988585 scopus 로고
    • Inhibitors of mammalian G1 cyclin-dependent kinases
    • COI: 1:CAS:528:DyaK2MXlvFShs7Y%3D, PID: 775894
    • Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 1995;9(10):1149–63.
    • (1995) Genes Dev , vol.9 , Issue.10 , pp. 1149-1163
    • Sherr, C.J.1    Roberts, J.M.2
  • 116
    • 0029102829 scopus 로고
    • Inhibition of nucleotide excision repair by the cyclin-dependent kinase inhibitor p21
    • COI: 1:CAS:528:DyaK2MXotFait78%3D, PID: 766562
    • Pan ZQ, et al. Inhibition of nucleotide excision repair by the cyclin-dependent kinase inhibitor p21. J Biol Chem. 1995;270(37):22008–16.
    • (1995) J Biol Chem , vol.270 , Issue.37 , pp. 22008-22016
    • Pan, Z.Q.1
  • 117
    • 0019394338 scopus 로고
    • Gene required in G1 for commitment to cell cycle and in G2 for control of mitosis in fission yeast
    • COI: 1:CAS:528:DyaL38XjsVyr, PID: 725435
    • Nurse P, Bissett Y. Gene required in G1 for commitment to cell cycle and in G2 for control of mitosis in fission yeast. Nature. 1981;292(5823):558–60.
    • (1981) Nature , vol.292 , Issue.5823 , pp. 558-560
    • Nurse, P.1    Bissett, Y.2
  • 118
    • 33847703331 scopus 로고    scopus 로고
    • Cellular senescence and chromatin organisation
    • COI: 1:CAS:528:DC%2BD2sXisVGis7g%3D, PID: 1731101
    • Narita M. Cellular senescence and chromatin organisation. Br J Cancer. 2007;96(5):686–91.
    • (2007) Br J Cancer , vol.96 , Issue.5 , pp. 686-691
    • Narita, M.1
  • 119
    • 34249725997 scopus 로고    scopus 로고
    • Alterations to nuclear architecture and genome behavior in senescent cells
    • COI: 1:CAS:528:DC%2BD2sXlslOitrc%3D, PID: 1746018
    • Mehta IS, et al. Alterations to nuclear architecture and genome behavior in senescent cells. Ann N Y Acad Sci. 2007;1100:250–63.
    • (2007) Ann N Y Acad Sci , vol.1100 , pp. 250-263
    • Mehta, I.S.1
  • 120
    • 0029047362 scopus 로고
    • A biomarker that identifies senescent human cells in culture and in aging skin in vivo
    • COI: 1:CAS:528:DyaK2MXosVWlur0%3D, PID: 756813
    • Dimri GP, et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA. 1995;92(20):9363–7.
    • (1995) Proc Natl Acad Sci USA , vol.92 , Issue.20 , pp. 9363-9367
    • Dimri, G.P.1
  • 121
    • 34548799705 scopus 로고    scopus 로고
    • Methods to detect biomarkers of cellular senescence: the senescence-associated beta-galactosidase assay
    • COI: 1:CAS:528:DC%2BD2sXls1Knu7s%3D, PID: 1763457
    • Itahana K, Campisi J, Dimri GP. Methods to detect biomarkers of cellular senescence: the senescence-associated beta-galactosidase assay. Methods Mol Biol. 2007;371:21–31.
    • (2007) Methods Mol Biol , vol.371 , pp. 21-31
    • Itahana, K.1    Campisi, J.2    Dimri, G.P.3
  • 122
    • 33746752125 scopus 로고    scopus 로고
    • A novel role for high-mobility group a proteins in cellular senescence and heterochromatin formation
    • COI: 1:CAS:528:DC%2BD28XosFGjtbg%3D, PID: 1690178
    • Narita M, et al. A novel role for high-mobility group a proteins in cellular senescence and heterochromatin formation. Cell. 2006;126(3):503–14.
    • (2006) Cell , vol.126 , Issue.3 , pp. 503-514
    • Narita, M.1
  • 123
    • 33746601439 scopus 로고    scopus 로고
    • Chromatin relaxation in response to DNA double-strand breaks is modulated by a novel ATM- and KAP-1 dependent pathway
    • COI: 1:CAS:528:DC%2BD28XnsFWqurg%3D, PID: 1686214
    • Ziv Y, et al. Chromatin relaxation in response to DNA double-strand breaks is modulated by a novel ATM- and KAP-1 dependent pathway. Nat Cell Biol. 2006;8(8):870–6.
    • (2006) Nat Cell Biol , vol.8 , Issue.8 , pp. 870-876
    • Ziv, Y.1
  • 124
    • 30344444484 scopus 로고    scopus 로고
    • Histone acetylation by Trrap-Tip60 modulates loading of repair proteins and repair of DNA double-strand breaks
    • COI: 1:CAS:528:DC%2BD28Xmt1eh, PID: 1634120
    • Murr R, et al. Histone acetylation by Trrap-Tip60 modulates loading of repair proteins and repair of DNA double-strand breaks. Nat Cell Biol. 2006;8(1):91–9.
    • (2006) Nat Cell Biol , vol.8 , Issue.1 , pp. 91-99
    • Murr, R.1
  • 125
    • 33644905252 scopus 로고    scopus 로고
    • Changes in chromatin structure and mobility in living cells at sites of DNA double-strand breaks
    • COI: 1:CAS:528:DC%2BD28XisFKis7k%3D, PID: 1652038
    • Kruhlak MJ, et al. Changes in chromatin structure and mobility in living cells at sites of DNA double-strand breaks. J Cell Biol. 2006;172(6):823–34.
    • (2006) J Cell Biol , vol.172 , Issue.6 , pp. 823-834
    • Kruhlak, M.J.1
  • 126
    • 84891713034 scopus 로고    scopus 로고
    • Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor
    • COI: 1:CAS:528:DC%2BD1MXlsFGhsQ%3D%3D, PID: 1905317
    • Coppe JP, et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008;6(12):2853–68.
    • (2008) PLoS Biol , vol.6 , Issue.12 , pp. 2853-2868
    • Coppe, J.P.1
  • 127
    • 77954563804 scopus 로고    scopus 로고
    • Senescent cells as a source of inflammatory factors for tumor progression
    • PID: 2039032
    • Davalos AR, et al. Senescent cells as a source of inflammatory factors for tumor progression. Cancer Metastasis Rev. 2010;29(2):273–83.
    • (2010) Cancer Metastasis Rev , vol.29 , Issue.2 , pp. 273-283
    • Davalos, A.R.1
  • 128
    • 58849154235 scopus 로고    scopus 로고
    • Senescence-messaging secretome: SMS-ing cellular stress
    • COI: 1:CAS:528:DC%2BD1MXjt1SmsA%3D%3D, PID: 1913200
    • Kuilman T, Peeper DS. Senescence-messaging secretome: SMS-ing cellular stress. Nat Rev Cancer. 2009;9(2):81–94.
    • (2009) Nat Rev Cancer , vol.9 , Issue.2 , pp. 81-94
    • Kuilman, T.1    Peeper, D.S.2
  • 129
    • 38049057905 scopus 로고    scopus 로고
    • DNA damage in telomeres and mitochondria during cellular senescence: is there a connection?
    • COI: 1:CAS:528:DC%2BD1cXisFyqsA%3D%3D, PID: 1798646
    • Passos JF, Saretzki G, von Zglinicki T. DNA damage in telomeres and mitochondria during cellular senescence: is there a connection? Nucleic Acids Res. 2007;35(22):7505–13.
    • (2007) Nucleic Acids Res , vol.35 , Issue.22 , pp. 7505-7513
    • Passos, J.F.1    Saretzki, G.2    von Zglinicki, T.3
  • 130
    • 34548790754 scopus 로고    scopus 로고
    • Mitochondria and ageing: winning and losing in the numbers game
    • COI: 1:CAS:528:DC%2BD2sXhtFWgsbzL, PID: 1768823
    • Passos JF, von Zglinicki T, Kirkwood TB. Mitochondria and ageing: winning and losing in the numbers game. BioEssays. 2007;29(9):908–17.
    • (2007) BioEssays , vol.29 , Issue.9 , pp. 908-917
    • Passos, J.F.1    von Zglinicki, T.2    Kirkwood, T.B.3
  • 131
    • 82755189388 scopus 로고    scopus 로고
    • Autophagy in cancer: having your cake and eating it
    • COI: 1:CAS:528:DC%2BC3MXhsFOhtb%2FJ, PID: 2194534
    • Hoare M, Young AR, Narita M. Autophagy in cancer: having your cake and eating it. Semin Cancer Biol. 2011;21(6):397–404.
    • (2011) Semin Cancer Biol , vol.21 , Issue.6 , pp. 397-404
    • Hoare, M.1    Young, A.R.2    Narita, M.3
  • 132
    • 79956325949 scopus 로고    scopus 로고
    • Spatial coupling of mTOR and autophagy augments secretory phenotypes
    • COI: 1:CAS:528:DC%2BC3MXmtFSgurw%3D, PID: 2151200
    • Narita M, et al. Spatial coupling of mTOR and autophagy augments secretory phenotypes. Science. 2011;332(6032):966–70.
    • (2011) Science , vol.332 , Issue.6032 , pp. 966-970
    • Narita, M.1
  • 133
    • 64349123107 scopus 로고    scopus 로고
    • Autophagy mediates the mitotic senescence transition
    • COI: 1:CAS:528:DC%2BD1MXks1SrtbY%3D, PID: 1927932
    • Young AR, et al. Autophagy mediates the mitotic senescence transition. Genes Dev. 2009;23(7):798–803.
    • (2009) Genes Dev , vol.23 , Issue.7 , pp. 798-803
    • Young, A.R.1
  • 134
    • 84869214214 scopus 로고    scopus 로고
    • The autophagy-senescence connection in chemotherapy: must tumor cells (self) eat before they sleep?
    • COI: 1:CAS:528:DC%2BC38Xhslyjs77E, PID: 2292754
    • Goehe RW, et al. The autophagy-senescence connection in chemotherapy: must tumor cells (self) eat before they sleep? J Pharmacol Exp Ther. 2012;343(3):763–78.
    • (2012) J Pharmacol Exp Ther , vol.343 , Issue.3 , pp. 763-778
    • Goehe, R.W.1
  • 135
    • 84858142662 scopus 로고    scopus 로고
    • MicroRNAs and their roles in aging
    • COI: 1:CAS:528:DC%2BC38XjslKjsrY%3D, PID: 2229461
    • Smith-Vikos T, Slack FJ. MicroRNAs and their roles in aging. J Cell Sci. 2012;125(Pt 1):7–17.
    • (2012) J Cell Sci , vol.125 , pp. 7-17
    • Smith-Vikos, T.1    Slack, F.J.2
  • 136
    • 71849111080 scopus 로고    scopus 로고
    • Alterations in microRNA expression in stress-induced cellular senescence
    • COI: 1:CAS:528:DC%2BD1MXhsFGrt77F, PID: 1978269
    • Li G, et al. Alterations in microRNA expression in stress-induced cellular senescence. Mech Ageing Dev. 2009;130(11–12):731–41.
    • (2009) Mech Ageing Dev , vol.130 , Issue.11-12 , pp. 731-741
    • Li, G.1
  • 137
    • 84877834704 scopus 로고    scopus 로고
    • From cellular senescence to age-associated diseases: the miRNA connection
    • PID: 2447223
    • Schraml E, Grillari J. From cellular senescence to age-associated diseases: the miRNA connection. Longev Healthspan. 2012;1(1):10.
    • (2012) Longev Healthspan , vol.1 , Issue.1 , pp. 10
    • Schraml, E.1    Grillari, J.2
  • 138
    • 84866516513 scopus 로고    scopus 로고
    • Regulation of senescence by microRNA biogenesis factors
    • COI: 1:CAS:528:DC%2BC38XhsVSlur%2FO, PID: 2230679
    • Abdelmohsen K, et al. Regulation of senescence by microRNA biogenesis factors. Ageing Res Rev. 2012;11(4):491–500.
    • (2012) Ageing Res Rev , vol.11 , Issue.4 , pp. 491-500
    • Abdelmohsen, K.1
  • 139
    • 79956315291 scopus 로고    scopus 로고
    • MicroRegulators come of age in senescence
    • COI: 1:CAS:528:DC%2BC3MXmvVaqurs%3D, PID: 2159261
    • Gorospe M, Abdelmohsen K. MicroRegulators come of age in senescence. Trends Genet. 2011;27(6):233–41.
    • (2011) Trends Genet , vol.27 , Issue.6 , pp. 233-241
    • Gorospe, M.1    Abdelmohsen, K.2
  • 140
    • 84934439522 scopus 로고    scopus 로고
    • Markers of cellular senescence
    • COI: 1:CAS:528:DC%2BC2MXitFertrs%3D, PID: 2329665
    • Carnero A. Markers of cellular senescence. Methods Mol Biol. 2013;965:63–81.
    • (2013) Methods Mol Biol , vol.965 , pp. 63-81
    • Carnero, A.1
  • 141
    • 0034713329 scopus 로고    scopus 로고
    • Is beta-galactosidase staining a marker of senescence in vitro and in vivo?
    • COI: 1:CAS:528:DC%2BD3cXjvFCrs7w%3D, PID: 1085406
    • Severino J, et al. Is beta-galactosidase staining a marker of senescence in vitro and in vivo? Exp Cell Res. 2000;257(1):162–71.
    • (2000) Exp Cell Res , vol.257 , Issue.1 , pp. 162-171
    • Severino, J.1
  • 142
    • 0032566278 scopus 로고    scopus 로고
    • Endogenous beta-galactosidase activity in continuously nonproliferating cells
    • COI: 1:CAS:528:DyaK1cXls1Sht7o%3D, PID: 971646
    • Yegorov YE, et al. Endogenous beta-galactosidase activity in continuously nonproliferating cells. Exp Cell Res. 1998;243(1):207–11.
    • (1998) Exp Cell Res , vol.243 , Issue.1 , pp. 207-211
    • Yegorov, Y.E.1
  • 143
    • 26244445013 scopus 로고    scopus 로고
    • The limitations and validities of senescence associated-beta-galactosidase activity as an aging marker for human foreskin fibroblast Hs68 cells
    • COI: 1:CAS:528:DC%2BD2MXhtVyms7zK, PID: 1615430
    • Yang NC, Hu ML. The limitations and validities of senescence associated-beta-galactosidase activity as an aging marker for human foreskin fibroblast Hs68 cells. Exp Gerontol. 2005;40(10):813–9.
    • (2005) Exp Gerontol , vol.40 , Issue.10 , pp. 813-819
    • Yang, N.C.1    Hu, M.L.2
  • 144
    • 79953292680 scopus 로고    scopus 로고
    • Oxidative damage increases and antioxidant gene expression decreases with aging in the mouse ovary
    • COI: 1:CAS:528:DC%2BC3MXjvFanu7s%3D, PID: 2114810
    • Lim J, Luderer U. Oxidative damage increases and antioxidant gene expression decreases with aging in the mouse ovary. Biol Reprod. 2011;84(4):775–82.
    • (2011) Biol Reprod , vol.84 , Issue.4 , pp. 775-782
    • Lim, J.1    Luderer, U.2
  • 145
    • 77949881221 scopus 로고    scopus 로고
    • The senescence-associated secretory phenotype: the dark side of tumor suppression
    • COI: 1:CAS:528:DC%2BC3cXivFekt70%3
    • Coppe JP, et al. The senescence-associated secretory phenotype: the dark side of tumor suppression. Ann Rev Pathol. 2010;5:99–118.
    • (2010) Ann Rev Pathol , vol.5 , pp. 99-118
    • Coppe, J.P.1
  • 146
    • 0033539169 scopus 로고    scopus 로고
    • Microarray analysis of replicative senescence
    • COI: 1:CAS:528:DyaK1MXlvFWlurw%3D, PID: 1050858
    • Shelton DN, et al. Microarray analysis of replicative senescence. Curr Biol. 1999;9(17):939–45.
    • (1999) Curr Biol , vol.9 , Issue.17 , pp. 939-945
    • Shelton, D.N.1
  • 147
    • 68249096784 scopus 로고    scopus 로고
    • Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion
    • COI: 1:CAS:528:DC%2BD1MXpsVaktb0%3D, PID: 1959748
    • Rodier F, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009;11(8):973–9.
    • (2009) Nat Cell Biol , vol.11 , Issue.8 , pp. 973-979
    • Rodier, F.1
  • 148
    • 79955017452 scopus 로고    scopus 로고
    • p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype
    • COI: 1:CAS:528:DC%2BC3MXjtVGmsr4%3D, PID: 2139961
    • Freund A, Patil CK, Campisi J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J. 2011;30(8):1536–48.
    • (2011) EMBO J , vol.30 , Issue.8 , pp. 1536-1548
    • Freund, A.1    Patil, C.K.2    Campisi, J.3
  • 149
    • 84856209016 scopus 로고    scopus 로고
    • Emerging role of NF-kappaB signaling in the induction of senescence-associated secretory phenotype (SASP)
    • COI: 1:CAS:528:DC%2BC38XhsVeksLo%3D, PID: 2218250
    • Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-kappaB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal. 2012;24(4):835–45.
    • (2012) Cell Signal , vol.24 , Issue.4 , pp. 835-845
    • Salminen, A.1    Kauppinen, A.2    Kaarniranta, K.3
  • 150
    • 84887866778 scopus 로고    scopus 로고
    • Cellular senescence or EGFR signaling induces Interleukin 6 (IL-6) receptor expression controlled by mammalian target of rapamycin (mTOR)
    • COI: 1:CAS:528:DC%2BC2cXhs1Wlu7k%3D, PID: 2404769
    • Garbers C, et al. Cellular senescence or EGFR signaling induces Interleukin 6 (IL-6) receptor expression controlled by mammalian target of rapamycin (mTOR). Cell Cycle. 2013;12(21):3421.
    • (2013) Cell Cycle , vol.12 , Issue.21 , pp. 3421
    • Garbers, C.1
  • 151
    • 84862794254 scopus 로고    scopus 로고
    • A senescent cell bystander effect: senescence-induced senescence
    • COI: 1:CAS:528:DC%2BC38XltFSisbc%3D, PID: 2232166
    • Nelson G, et al. A senescent cell bystander effect: senescence-induced senescence. Aging Cell. 2012;11:345.
    • (2012) Aging Cell , vol.11 , pp. 345
    • Nelson, G.1
  • 152
    • 44649120132 scopus 로고    scopus 로고
    • Chemokine signaling via the CXCR2 receptor reinforces senescence
    • COI: 1:CAS:528:DC%2BD1cXnsF2gtbo%3D, PID: 1855577
    • Acosta JC, et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008;133(6):1006–18.
    • (2008) Cell , vol.133 , Issue.6 , pp. 1006-1018
    • Acosta, J.C.1
  • 153
    • 44649101304 scopus 로고    scopus 로고
    • Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network
    • COI: 1:CAS:528:DC%2BD1cXnsF2gtbs%3D, PID: 1855577
    • Kuilman T, et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008;133(6):1019–31.
    • (2008) Cell , vol.133 , Issue.6 , pp. 1019-1031
    • Kuilman, T.1
  • 154
    • 0034704916 scopus 로고    scopus 로고
    • Defects in TGF-beta signaling overcome senescence of mouse keratinocytes expressing v-Ha-ras
    • COI: 1:CAS:528:DC%2BD3cXivVartLc%3D, PID: 1076382
    • Tremain R, et al. Defects in TGF-beta signaling overcome senescence of mouse keratinocytes expressing v-Ha-ras. Oncogene. 2000;19(13):1698–709.
    • (2000) Oncogene , vol.19 , Issue.13 , pp. 1698-1709
    • Tremain, R.1
  • 155
    • 16244377705 scopus 로고    scopus 로고
    • TGF beta1 induces prolonged mitochondrial ROS generation through decreased complex IV activity with senescent arrest in Mv1Lu cells
    • COI: 1:CAS:528:DC%2BD2MXitV2nsro%3D, PID: 1568803
    • Yoon YS, et al. TGF beta1 induces prolonged mitochondrial ROS generation through decreased complex IV activity with senescent arrest in Mv1Lu cells. Oncogene. 2005;24(11):1895–903.
    • (2005) Oncogene , vol.24 , Issue.11 , pp. 1895-1903
    • Yoon, Y.S.1
  • 156
    • 38849168162 scopus 로고    scopus 로고
    • Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7
    • COI: 1:CAS:528:DC%2BD1cXivVahtbc%3D, PID: 1826706
    • Wajapeyee N, et al. Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell. 2008;132(3):363–74.
    • (2008) Cell , vol.132 , Issue.3 , pp. 363-374
    • Wajapeyee, N.1
  • 157
    • 34447645303 scopus 로고    scopus 로고
    • Regulation of replicative senescence by insulin-like growth factor-binding protein 3 in human umbilical vein endothelial cells
    • COI: 1:CAS:528:DC%2BD2sXptlyks7s%3D, PID: 1763541
    • Kim KS, et al. Regulation of replicative senescence by insulin-like growth factor-binding protein 3 in human umbilical vein endothelial cells. Aging Cell. 2007;6(4):535–45.
    • (2007) Aging Cell , vol.6 , Issue.4 , pp. 535-545
    • Kim, K.S.1
  • 158
    • 35848947951 scopus 로고    scopus 로고
    • Induction of cellular senescence by insulin-like growth factor binding protein-5 through a p53-dependent mechanism
    • COI: 1:CAS:528:DC%2BD2sXht12js7vK, PID: 1780481
    • Kim KS, et al. Induction of cellular senescence by insulin-like growth factor binding protein-5 through a p53-dependent mechanism. Mol Biol Cell. 2007;18(11):4543–52.
    • (2007) Mol Biol Cell , vol.18 , Issue.11 , pp. 4543-4552
    • Kim, K.S.1
  • 159
    • 33746616991 scopus 로고    scopus 로고
    • Plasminogen activator inhibitor-1 is a critical downstream target of p53 in the induction of replicative senescence
    • COI: 1:CAS:528:DC%2BD28XnsFWqurY%3D, PID: 1686214
    • Kortlever RM, Higgins PJ, Bernards R. Plasminogen activator inhibitor-1 is a critical downstream target of p53 in the induction of replicative senescence. Nat Cell Biol. 2006;8(8):877–84.
    • (2006) Nat Cell Biol , vol.8 , Issue.8 , pp. 877-884
    • Kortlever, R.M.1    Higgins, P.J.2    Bernards, R.3
  • 160
    • 0035834121 scopus 로고    scopus 로고
    • Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging
    • COI: 1:CAS:528:DC%2BD3MXns1CjsLw%3D, PID: 1159301
    • Krtolica A, et al. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc Natl Acad Sci USA. 2001;98(21):12072–7.
    • (2001) Proc Natl Acad Sci USA , vol.98 , Issue.21 , pp. 12072-12077
    • Krtolica, A.1
  • 161
    • 13944260728 scopus 로고    scopus 로고
    • Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation
    • COI: 1:CAS:528:DC%2BD2MXhvFKntb0%3D, PID: 1565708
    • Parrinello S, et al. Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation. J Cell Sci. 2005;118(Pt 3):485–96.
    • (2005) J Cell Sci , vol.118 , pp. 485-496
    • Parrinello, S.1
  • 162
    • 36849039660 scopus 로고    scopus 로고
    • IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland
    • COI: 1:CAS:528:DC%2BD2sXhsValsLzK, PID: 1806003
    • Sansone P, et al. IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. J Clin Invest. 2007;117(12):3988–4002.
    • (2007) J Clin Invest , vol.117 , Issue.12 , pp. 3988-4002
    • Sansone, P.1
  • 163
    • 33749549246 scopus 로고    scopus 로고
    • Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence
    • COI: 1:CAS:528:DC%2BD28XhtVahsLfJ, PID: 1688020
    • Coppe JP, et al. Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. J Biol Chem. 2006;281(40):29568–74.
    • (2006) J Biol Chem , vol.281 , Issue.40 , pp. 29568-29574
    • Coppe, J.P.1
  • 164
    • 33846937033 scopus 로고    scopus 로고
    • Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas
    • COI: 1:CAS:528:DC%2BD2sXhtlOrtbo%3D, PID: 1725193
    • Xue W, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007;445(7128):656–60.
    • (2007) Nature , vol.445 , Issue.7128 , pp. 656-660
    • Xue, W.1
  • 165
    • 49749145668 scopus 로고    scopus 로고
    • Senescence of activated stellate cells limits liver fibrosis
    • COI: 1:CAS:528:DC%2BD1cXhtVGqtrbI, PID: 1872493
    • Krizhanovsky V, et al. Senescence of activated stellate cells limits liver fibrosis. Cell. 2008;134(4):657–67.
    • (2008) Cell , vol.134 , Issue.4 , pp. 657-667
    • Krizhanovsky, V.1
  • 166
    • 81855169964 scopus 로고    scopus 로고
    • Senescence surveillance of pre-malignant hepatocytes limits liver cancer development
    • COI: 1:CAS:528:DC%2BC3MXhsVKjsLvO, PID: 2208094
    • Kang TW, et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature. 2011;479(7374):547–51.
    • (2011) Nature , vol.479 , Issue.7374 , pp. 547-551
    • Kang, T.W.1
  • 167
    • 84900969225 scopus 로고    scopus 로고
    • Ink4a/Arf expression is a biomarker of aging
    • COI: 1:CAS:528:DC%2BD2cXpsFCgtrs%3D, PID: 1552086
    • Krishnamurthy J, et al. Ink4a/Arf expression is a biomarker of aging. J Clin Invest. 2004;114(9):1299–307.
    • (2004) J Clin Invest , vol.114 , Issue.9 , pp. 1299-1307
    • Krishnamurthy, J.1
  • 168
    • 33749172559 scopus 로고    scopus 로고
    • Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a
    • COI: 1:CAS:528:DC%2BD28XhtVSns7fE, PID: 1695773
    • Janzen V, et al. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature. 2006;443(7110):421–6.
    • (2006) Nature , vol.443 , Issue.7110 , pp. 421-426
    • Janzen, V.1
  • 169
    • 33749171885 scopus 로고    scopus 로고
    • Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing
    • COI: 1:CAS:528:DC%2BD28XhtVSns7bL, PID: 1695773
    • Molofsky AV, et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006;443(7110):448–52.
    • (2006) Nature , vol.443 , Issue.7110 , pp. 448-452
    • Molofsky, A.V.1
  • 170
    • 5444253109 scopus 로고    scopus 로고
    • Exploration of replicative senescence-associated genes in human dermal fibroblasts by cDNA microarray technology
    • COI: 1:CAS:528:DC%2BD2cXos1eqtLs%3D, PID: 1548906
    • Yoon IK, et al. Exploration of replicative senescence-associated genes in human dermal fibroblasts by cDNA microarray technology. Exp Gerontol. 2004;39(9):1369–78.
    • (2004) Exp Gerontol , vol.39 , Issue.9 , pp. 1369-1378
    • Yoon, I.K.1
  • 171
    • 33646470845 scopus 로고    scopus 로고
    • Expression profiling of aging in the human skin
    • COI: 1:CAS:528:DC%2BD28XksVOhurw%3D, PID: 1653036
    • Lener T, et al. Expression profiling of aging in the human skin. Exp Gerontol. 2006;41(4):387–97.
    • (2006) Exp Gerontol , vol.41 , Issue.4 , pp. 387-397
    • Lener, T.1
  • 172
    • 53649109801 scopus 로고    scopus 로고
    • Critical pathways in cellular senescence and immortalization revealed by gene expression profiling
    • COI: 1:CAS:528:DC%2BD1cXht1WmtLvE, PID: 1871140
    • Fridman AL, Tainsky MA. Critical pathways in cellular senescence and immortalization revealed by gene expression profiling. Oncogene. 2008;27(46):5975–87.
    • (2008) Oncogene , vol.27 , Issue.46 , pp. 5975-5987
    • Fridman, A.L.1    Tainsky, M.A.2
  • 173
    • 84864070781 scopus 로고    scopus 로고
    • Microarray-based identification of age-dependent differences in gene expression of human dermal fibroblasts
    • COI: 1:CAS:528:DC%2BC38XhtV2nsrfL, PID: 2272168
    • Dekker P, et al. Microarray-based identification of age-dependent differences in gene expression of human dermal fibroblasts. Mech Ageing Dev. 2012;133(7):498–507.
    • (2012) Mech Ageing Dev , vol.133 , Issue.7 , pp. 498-507
    • Dekker, P.1
  • 174
    • 84939935728 scopus 로고    scopus 로고
    • Selective insulin resistance in hepatocyte senescence, Exp Cell Res
    • Aravinthan A, et al. Selective insulin resistance in hepatocyte senescence. Exp Cell Res. 2014.
    • (2014) et al
    • Aravinthan, A.1
  • 175
    • 0036829090 scopus 로고    scopus 로고
    • Cancer and aging: a model for the cancer promoting effects of the aging stroma
    • COI: 1:CAS:528:DC%2BD38XmsVersbY%3D, PID: 1220003
    • Krtolica A, Campisi J. Cancer and aging: a model for the cancer promoting effects of the aging stroma. Int J Biochem Cell Biol. 2002;34(11):1401–14.
    • (2002) Int J Biochem Cell Biol , vol.34 , Issue.11 , pp. 1401-1414
    • Krtolica, A.1    Campisi, J.2
  • 176
    • 0028856794 scopus 로고
    • Telomere length and replicative aging in human vascular tissues
    • COI: 1:CAS:528:DyaK2MXps1altrs%3D, PID: 747996
    • Chang E, Harley CB. Telomere length and replicative aging in human vascular tissues. Proc Natl Acad Sci USA. 1995;92(24):11190–4.
    • (1995) Proc Natl Acad Sci USA , vol.92 , Issue.24 , pp. 11190-11194
    • Chang, E.1    Harley, C.B.2
  • 177
    • 0035130153 scopus 로고    scopus 로고
    • Differential expression of thymosin beta-10 by early passage and senescent vascular endothelium is modulated by VPF/VEGF: evidence for senescent endothelial cells in vivo at sites of atherosclerosis
    • COI: 1:CAS:528:DC%2BD3MXhsFemsrs%3D, PID: 1115696
    • Vasile E, et al. Differential expression of thymosin beta-10 by early passage and senescent vascular endothelium is modulated by VPF/VEGF: evidence for senescent endothelial cells in vivo at sites of atherosclerosis. FASEB J. 2001;15(2):458–66.
    • (2001) FASEB J , vol.15 , Issue.2 , pp. 458-466
    • Vasile, E.1
  • 178
    • 0042734423 scopus 로고    scopus 로고
    • The role of chondrocyte senescence in osteoarthritis
    • COI: 1:CAS:528:DC%2BD3sXkt1Kqtrw%3D, PID: 1288235
    • Price JS, et al. The role of chondrocyte senescence in osteoarthritis. Aging Cell. 2002;1(1):57–65.
    • (2002) Aging Cell , vol.1 , Issue.1 , pp. 57-65
    • Price, J.S.1
  • 179
    • 0037007075 scopus 로고    scopus 로고
    • Endothelial cell senescence in human atherosclerosis: role of telomere in endothelial dysfunction
    • COI: 1:CAS:528:DC%2BD38XjtlCmtLg%3D, PID: 1192751
    • Minamino T, et al. Endothelial cell senescence in human atherosclerosis: role of telomere in endothelial dysfunction. Circulation. 2002;105(13):1541–4.
    • (2002) Circulation , vol.105 , Issue.13 , pp. 1541-1544
    • Minamino, T.1
  • 180
    • 80855138775 scopus 로고    scopus 로고
    • Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders
    • COI: 1:CAS:528:DC%2BC3MXhsVSktb7N, PID: 2204831
    • Baker DJ, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479(7372):232–6.
    • (2011) Nature , vol.479 , Issue.7372 , pp. 232-236
    • Baker, D.J.1
  • 181
    • 84914815749 scopus 로고    scopus 로고
    • Are there roles for brain cell senescence in aging and neurodegenerative disorders?
    • COI: 1:CAS:528:DC%2BC2cXhslOgtrfO, PID: 2530505
    • Tan FC, et al. Are there roles for brain cell senescence in aging and neurodegenerative disorders? Biogerontology. 2014;15:643.
    • (2014) Biogerontology , vol.15 , pp. 643
    • Tan, F.C.1
  • 182
    • 84885676831 scopus 로고    scopus 로고
    • Cellular senescence in normal and premature lung aging
    • COI: 1:STN:280:DC%2BC3sbivVClsg%3D%3D, PID: 2398244
    • Bartling B. Cellular senescence in normal and premature lung aging. Z Gerontol Geriatr. 2013;46(7):613–22.
    • (2013) Z Gerontol Geriatr , vol.46 , Issue.7 , pp. 613-622
    • Bartling, B.1
  • 183
    • 84874115065 scopus 로고    scopus 로고
    • Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease
    • COI: 1:CAS:528:DC%2BC3sXnvFSqsQ%3D%3D, PID: 2314262
    • Aravinthan A, et al. Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease. J Hepatol. 2013;58(3):549–56.
    • (2013) J Hepatol , vol.58 , Issue.3 , pp. 549-556
    • Aravinthan, A.1
  • 184
    • 84861651694 scopus 로고    scopus 로고
    • Accelerated senescence of renal tubular epithelial cells is associated with disease progression of patients with immunoglobulin A (IgA) nephropathy
    • COI: 1:CAS:528:DC%2BC38Xnsl2lsr4%3D, PID: 2263309
    • Liu J, et al. Accelerated senescence of renal tubular epithelial cells is associated with disease progression of patients with immunoglobulin A (IgA) nephropathy. Transl Res. 2012;159(6):454–63.
    • (2012) Transl Res , vol.159 , Issue.6 , pp. 454-463
    • Liu, J.1
  • 185
    • 33748164460 scopus 로고    scopus 로고
    • Vascular smooth muscle cell senescence in atherosclerosis
    • COI: 1:CAS:528:DC%2BD28XpsVWqt7c%3D, PID: 1682449
    • Gorenne I, et al. Vascular smooth muscle cell senescence in atherosclerosis. Cardiovasc Res. 2006;72(1):9–17.
    • (2006) Cardiovasc Res , vol.72 , Issue.1 , pp. 9-17
    • Gorenne, I.1
  • 186
    • 0031755297 scopus 로고    scopus 로고
    • The role of cellular senescence in skin aging
    • COI: 1:STN:280:DyaK1cvgtVOqtQ%3D%3D, PID: 973204
    • Campisi J. The role of cellular senescence in skin aging. J Investig Dermatol Symp Proc. 1998;3(1):1–5.
    • (1998) J Investig Dermatol Symp Proc , vol.3 , Issue.1 , pp. 1-5
    • Campisi, J.1
  • 187
    • 84884570112 scopus 로고    scopus 로고
    • Hepatocyte expression of the senescence marker p21 is linked to fibrosis and an adverse liver-related outcome in alcohol-related liver disease
    • COI: 1:CAS:528:DC%2BC3sXhsFKisbnO, PID: 2408626
    • Aravinthan A, et al. Hepatocyte expression of the senescence marker p21 is linked to fibrosis and an adverse liver-related outcome in alcohol-related liver disease. PLoS One. 2013;8(9):e72904.
    • (2013) PLoS One , vol.8 , Issue.9 , pp. e72904
    • Aravinthan, A.1
  • 188
    • 84901733241 scopus 로고    scopus 로고
    • Senescent cardiac fibroblast is critical for cardiac fibrosis after myocardial infarction
    • COI: 1:CAS:528:DC%2BC3sXhsV2rtbjO, PID: 2404027
    • Zhu F, et al. Senescent cardiac fibroblast is critical for cardiac fibrosis after myocardial infarction. PLoS One. 2013;8(9):e74535.
    • (2013) PLoS One , vol.8 , Issue.9 , pp. e74535
    • Zhu, F.1
  • 189
    • 0034003333 scopus 로고    scopus 로고
    • A critical role for telomeres in suppressing and facilitating carcinogenesis
    • COI: 1:CAS:528:DC%2BD3cXhslOksrg%3D, PID: 1067939
    • Artandi SE, DePinho RA. A critical role for telomeres in suppressing and facilitating carcinogenesis. Curr Opin Genet Dev. 2000;10(1):39–46.
    • (2000) Curr Opin Genet Dev , vol.10 , Issue.1 , pp. 39-46
    • Artandi, S.E.1    DePinho, R.A.2
  • 190
    • 0035500487 scopus 로고    scopus 로고
    • Cellular senescence as a tumor-suppressor mechanism
    • COI: 1:CAS:528:DC%2BD3MXnvFGrsLY%3D, PID: 1168443
    • Campisi J. Cellular senescence as a tumor-suppressor mechanism. Trends Cell Biol. 2001;11(11):S27–31.
    • (2001) Trends Cell Biol , vol.11 , Issue.11 , pp. S27-S31
    • Campisi, J.1
  • 191
    • 34247860915 scopus 로고    scopus 로고
    • Telomere dysfunction suppresses spontaneous tumorigenesis in vivo by initiating p53-dependent cellular senescence
    • COI: 1:CAS:528:DC%2BD2sXkslCksrs%3D, PID: 1739613
    • Cosme-Blanco W, et al. Telomere dysfunction suppresses spontaneous tumorigenesis in vivo by initiating p53-dependent cellular senescence. EMBO Rep. 2007;8(5):497–503.
    • (2007) EMBO Rep , vol.8 , Issue.5 , pp. 497-503
    • Cosme-Blanco, W.1
  • 192
    • 0035835818 scopus 로고    scopus 로고
    • p16(MTS-1/CDKN2/INK4a) in cancer progression
    • COI: 1:CAS:528:DC%2BD3MXhsFOhu78%3D, PID: 1123752
    • Rocco JW, Sidransky D. p16(MTS-1/CDKN2/INK4a) in cancer progression. Exp Cell Res. 2001;264(1):42–55.
    • (2001) Exp Cell Res , vol.264 , Issue.1 , pp. 42-55
    • Rocco, J.W.1    Sidransky, D.2
  • 193
    • 0842346437 scopus 로고    scopus 로고
    • Principles of tumor suppression
    • COI: 1:CAS:528:DC%2BD2cXhtVals74%3D, PID: 1474443
    • Sherr CJ. Principles of tumor suppression. Cell. 2004;116(2):235–46.
    • (2004) Cell , vol.116 , Issue.2 , pp. 235-246
    • Sherr, C.J.1
  • 194
    • 33846899456 scopus 로고    scopus 로고
    • Restoration of p53 function leads to tumour regression in vivo
    • COI: 1:CAS:528:DC%2BD2sXhtlOrtbg%3D, PID: 1725193
    • Ventura A, et al. Restoration of p53 function leads to tumour regression in vivo. Nature. 2007;445(7128):661–5.
    • (2007) Nature , vol.445 , Issue.7128 , pp. 661-665
    • Ventura, A.1
  • 195
    • 0037470488 scopus 로고    scopus 로고
    • Evolutionary medicine: from dwarf model systems to healthy centenarians?
    • PID: 1261029
    • Longo VD, Finch CE. Evolutionary medicine: from dwarf model systems to healthy centenarians? Science. 2003;299(5611):1342–6.
    • (2003) Science , vol.299 , Issue.5611 , pp. 1342-1346
    • Longo, V.D.1    Finch, C.E.2
  • 196
    • 13944253348 scopus 로고    scopus 로고
    • Calorie restriction–the SIR2 connection
    • COI: 1:CAS:528:DC%2BD2MXitVWnuro%3D, PID: 1573468
    • Guarente L, Picard F. Calorie restriction–the SIR2 connection. Cell. 2005;120(4):473–82.
    • (2005) Cell , vol.120 , Issue.4 , pp. 473-482
    • Guarente, L.1    Picard, F.2
  • 197
    • 84866459450 scopus 로고    scopus 로고
    • Dietary restriction attenuates age-associated muscle atrophy by lowering oxidative stress in mice even in complete absence of CuZnSOD
    • COI: 1:CAS:528:DC%2BC38XhsFCls7%2FF, PID: 2267261
    • Jang YC, et al. Dietary restriction attenuates age-associated muscle atrophy by lowering oxidative stress in mice even in complete absence of CuZnSOD. Aging Cell. 2012;11(5):770–82.
    • (2012) Aging Cell , vol.11 , Issue.5 , pp. 770-782
    • Jang, Y.C.1
  • 198
    • 79952118049 scopus 로고    scopus 로고
    • Adult-onset, short-term dietary restriction reduces cell senescence in mice
    • COI: 1:CAS:528:DC%2BC3cXht1KlsLv
    • Wang C, et al. Adult-onset, short-term dietary restriction reduces cell senescence in mice. Aging (Albany NY). 2010;2(9):555–66.
    • (2010) Aging (Albany NY) , vol.2 , Issue.9 , pp. 555-566
    • Wang, C.1
  • 199
    • 3042648746 scopus 로고    scopus 로고
    • Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway
    • COI: 1:CAS:528:DC%2BD2cXkvV2gtb4%3D, PID: 1518674
    • Kapahi P, et al. Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Curr Biol. 2004;14(10):885–90.
    • (2004) Curr Biol , vol.14 , Issue.10 , pp. 885-890
    • Kapahi, P.1
  • 200
    • 0016724057 scopus 로고
    • Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle
    • COI: 1:CAS:528:DyaE28Xis1eruw%3D%3
    • Vezina C, Kudelski A, Sehgal SN. Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J Antibiot (Tokyo). 1975;28(10):721–6.
    • (1975) J Antibiot (Tokyo) , vol.28 , Issue.10 , pp. 721-726
    • Vezina, C.1    Kudelski, A.2    Sehgal, S.N.3
  • 201
    • 67650944993 scopus 로고    scopus 로고
    • Rapamycin fed late in life extends lifespan in genetically heterogeneous mice
    • COI: 1:CAS:528:DC%2BD1MXotlSgtLg%3D, PID: 1958768
    • Harrison DE, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392.
    • (2009) Nature , vol.460 , pp. 392
    • Harrison, D.E.1
  • 202
    • 79951794971 scopus 로고    scopus 로고
    • Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice
    • PID: 2097473
    • Miller RA, et al. Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice. J Gerontol A Biol Sci Med Sci. 2011;66(2):191–201.
    • (2011) J Gerontol A Biol Sci Med Sci , vol.66 , Issue.2 , pp. 191-201
    • Miller, R.A.1
  • 203
    • 83755165458 scopus 로고    scopus 로고
    • Rapamycin increases lifespan and inhibits spontaneous tumorigenesis in inbred female mice
    • COI: 1:CAS:528:DC%2BC38XhvVKlur0%3D, PID: 2210796
    • Anisimov VN, et al. Rapamycin increases lifespan and inhibits spontaneous tumorigenesis in inbred female mice. Cell Cycle. 2011;10(24):4230–6.
    • (2011) Cell Cycle , vol.10 , Issue.24 , pp. 4230-4236
    • Anisimov, V.N.1
  • 204
    • 70349433524 scopus 로고    scopus 로고
    • Expression and mechanism of mammalian target of rapamycin in age-related renal cell senescence and organ aging
    • COI: 1:CAS:528:DC%2BD1MXht1WrsbjE, PID: 1969873
    • Zhuo L, et al. Expression and mechanism of mammalian target of rapamycin in age-related renal cell senescence and organ aging. Mech Ageing Dev. 2009;130(10):700–8.
    • (2009) Mech Ageing Dev , vol.130 , Issue.10 , pp. 700-708
    • Zhuo, L.1
  • 205
    • 84874925174 scopus 로고    scopus 로고
    • Potential anti-aging agents suppress the level of constitutive mTOR- and DNA damage- signaling
    • COI: 1:CAS:528:DC%2BC3sXjsVSitLs%3
    • Halicka HD, et al. Potential anti-aging agents suppress the level of constitutive mTOR- and DNA damage- signaling. Aging (Albany NY). 2012;4(12):952–65.
    • (2012) Aging (Albany NY) , vol.4 , Issue.12 , pp. 952-965
    • Halicka, H.D.1
  • 206
    • 33847226657 scopus 로고    scopus 로고
    • An anti-aging drug today: from senescence-promoting genes to anti-aging pill
    • COI: 1:CAS:528:DC%2BD2sXitl2htL4%3D, PID: 1733188
    • Blagosklonny MV. An anti-aging drug today: from senescence-promoting genes to anti-aging pill. Drug Discov Today. 2007;12(5–6):218–24.
    • (2007) Drug Discov Today , vol.12 , Issue.5-6 , pp. 218-224
    • Blagosklonny, M.V.1
  • 207
    • 80855165212 scopus 로고    scopus 로고
    • Genome protective effect of metformin as revealed by reduced level of constitutive DNA damage signaling
    • COI: 1:CAS:528:DC%2BC3MXhs1ags7b
    • Halicka HD, et al. Genome protective effect of metformin as revealed by reduced level of constitutive DNA damage signaling. Aging (Albany NY). 2011;3(10):1028–38.
    • (2011) Aging (Albany NY) , vol.3 , Issue.10 , pp. 1028-1038
    • Halicka, H.D.1
  • 208
    • 84885861971 scopus 로고    scopus 로고
    • Mechanism of metformin: Inhibition of DNA damage and proliferative activity in Drosophila midgut stem cell
    • COI: 1:CAS:528:DC%2BC3sXht1Ggt7vL, PID: 2389175
    • Na HJ, et al. Mechanism of metformin: Inhibition of DNA damage and proliferative activity in Drosophila midgut stem cell. Mech Ageing Dev. 2013;134:381.
    • (2013) Mech Ageing Dev , vol.134 , pp. 381
    • Na, H.J.1
  • 209
    • 84888082787 scopus 로고    scopus 로고
    • Dual CDK4/CDK6 inhibition induces cell-cycle arrest and senescence in neuroblastoma
    • COI: 1:CAS:528:DC%2BC3sXhslyku7rL, PID: 2404517
    • Rader J, et al. Dual CDK4/CDK6 inhibition induces cell-cycle arrest and senescence in neuroblastoma. Clin Cancer Res. 2013;19(22):6173–82.
    • (2013) Clin Cancer Res , vol.19 , Issue.22 , pp. 6173-6182
    • Rader, J.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.