648
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Cellular Senescence - its role in cancer and the response to ionizing radiation

      review-article
      Bookmark

            Abstract

            Cellular senescence is a normal biological process that is initiated in response to a range of intrinsic and extrinsic factors that functions to remove irreparable damage and therefore potentially harmful cells, from the proliferative pool. Senescence can therefore be thought of in beneficial terms as a tumour suppressor. In contrast to this, there is a growing body of evidence suggesting that senescence is also associated with the disruption of the tissue microenvironment and development of a pro-oncogenic environment, principally via the secretion of senescence-associated pro-inflammatory factors. The fraction of cells in a senescent state is known to increase with cellular age and from exposure to various stressors including ionising radiation therefore, the implications of the detrimental effects of the senescent phenotype are important to understand within the context of the increasing human exposure to ionising radiation. This review will discuss what is currently understood about senescence, highlighting possible associations between senescence and cancer and, how exposure to ionising radiation may modify this.

            Main article text

            Cellular Senescence

            Cellular senescence is a metabolically active form of irreversible growth arrest that halts the proliferation of ageing and/or damaged cells and as a consequence, prevents the transmission of damage to daughter cells. This complicated cellular event is initiated in response to a variety of intrinsic and extrinsic genotoxic stimuli [1-4] and mediated through tumour suppressor pathways involving p53, and p16INK4A/pRb [5,6]. This ultimately leads to the inhibition of cyclin-dependant kinases [7,8]. Accordingly, cellular senescence can be thought of as a tumour suppressor mechanism. Indeed the majority of cancers have mutations in p53 and/or the pRb/p16 pathways, while germ-line mutations in these pathways result in a cell-specific ability to overcome senescence-inducing signals, thereby greatly increasing their susceptibility to cellular transformation [9-11]. The importance of cellular senescence as a tumour suppressor is further demonstrated by cell fusion experiments [12] that provide evidence that growth arrest observed in senescent cells has a strong influence over the growth in proliferating cells and cellular oncogenes of tumour cells. When proliferating cells were fused with senescent cells, DNA replication was inhibited even in the presence of mitogens, and when senescent cells were fused with tumour cells, DNA replication was similarly inhibited. These fusion experiments led to the assumption that senescent cells contained control elements capable of exerting a dominant effect over proliferating pre-senescent cells. Importantly, this tumour suppressive mechanism of cellular senescence has been supported in both mice and human studies [13].

            As well as possessing tumour suppressive mechanisms, senescence has been found to play an important role in wound healing and tissue repair and/or communication to surrounding tissues/cells of damage crisis to assist healing [14,15]. For instance, senescent cells have been shown in in vivo mouse models to play a role in the resolution of fibrosis by matrix metalloproteinases (MMPs) after acute liver injury. Under normal conditions, proliferating hepatic stellate cells triggered in response to acute liver injury, produce fibrotic scars in advance of entering into a senescent state, followed by secretion of MMPs and scar dissolution. However in cells deficient in either p53/pRb pathways, liver injury results in severe, irresolvable fibrosis [16]. Similarly, the matricellular protein CCN1, which is expressed at sites of cutaneous wound repair, has been shown to initiate DNA damage response pathways and reactive-oxygen species dependent activation of p16INK4A/pRb pathway, resulting in senescence and the expression of anti-fibrotic genes in wild-type mice. Mutant mice that express a senescence-defective CCN1 protein however show increased fibrosis at sites of wound repair [17]. Cellular senescence has been also shown to be important in the prevention of epithelial-mesenchymal transition (EMT) whereby the metastatic dissemination of cancerous cells is prevented [18,19]. Thus, the functional significance of cellular senescence includes a diverse range of roles which are essentially beneficial to the organism.

            Phenotype

            The major phenotype of senescence that characteristically distinguishes senescence from quiescence is irreversible growth arrest that cannot be reversed by any known physiological stimuli, associated with resistance to apoptosis and increased sensitivity to cellular injury [20-26]. Other characteristic changes include altered gene expression with increased expression of proteins including p53, p16, p19 and p21 [27,28], an increase in senescence-associated beta galactosidase (SA-β-gal) activity at pH 6.0 [29], the presence of persistent telomere and non-telomere DNA damage foci [30,31], senescence-associated heterochromatic foci (SAHF) [6] and a senescence-associated secretory phenotype (SASP) [32,33]. Accordingly, identification of senescent cells can be achieved by assaying for a combination of the above characteristics. The application of proliferation cell-cycle specific markers, such as Ki-67, can also be used [34-36]. For example Kill et al (1996) showed that 56% of human dermal fibroblasts were Ki-67 positive at early passage 4 [37] compared to only 30% at passage 38 [38] with the decrease in fraction of Ki-67 positive cells reflecting an increase in senescence.

            A range of morphological changes have also been documented with senescent fibroblasts showing enlarged and flattened morphology accompanied by the loss of elongated, spindle-like properties, when compared to normal proliferating fibroblasts. Specifically, the mean nuclear area of fibroblasts was shown to be 255 μm2 at early passage, compared to 293 μm2 at later passage [39]. Interestingly, the sub-nuclear organisation of chromosomes has also been shown to be different in senescent and proliferating mammalian somatic cells, whereby gene poor chromosomes such as chromosomes 13 and 18 are thought to alter their preferential nuclear position from near the nuclear periphery and relocate to the nuclear interior when induced to senesce. Thus, the interphase organisation of particular chromosome territories changes such that their position correlates according to the size of the chromosome, rather than the density [40,41].

            Replicative Senescence

            Seminal work carried out by Hayflick and Moorhead (1961) demonstrated that normal cells grown in culture dishes are only able to undergo a finite number of cellular divisions before their growth is irreversibly arrested [42-44]. This 'Hayflick Limit' was the first demonstration of a senescence phenotype and described the replicative capacity of diploid cells in culture before the cells ceased to divide [45] and has since been demonstrated for many different types of cells both in vitro and in vivo [46,47]. Importantly, diploid cells within cell populations do not all reach senescence at the same time, rather there is a progressive decrease in the fraction of proliferating cells that are capable of undergoing cellular division with each round of replication [48,49]. Also, different cell types and lineages will vary in the rate at which they enter a senescent state [50]. For example, in vitro studies comparing the growth rates and passage number of fibroblast and keratinocyte cell types observed that the decline in cell growth rate was notably higher for keratinocytes which had senesced by P6, compared to fibroblasts that were passaged beyond P10 in all donor age groups above and below 40 years of age; suggesting that the growth rate of the two cell types is independent of donor age [51]. Indeed a 'memory' for the number of completed population doublings was observed when WI-38 fibroblasts were found to enter senescence with respect to their remaining replicative capacity, even after cryopreservation for a period of 23 years [44].

            The mechanism for replicative senescence is believed to be associated with a progressive shortening of telomeres that occurs with each DNA replication cycle [52,53]. Functional telomeres protect the ends of chromosomes however, approximately 100-300 bp of these repeat sequences are lost as a result of incomplete replication of the extending 3' overhang of nucleotides [54,55]. Support for this comes from studies that show critically short telomeres trigger a DNA damage response which results in cellular senescence [30,56], while immortal cancer and germ cell lines overcome the action of telomeric shortening by the expression of the telomerase enzyme [57]. Telomerase synthesizes and maintains telomeric end sequences, preventing the exposure of uncapped ends [58] thereby permitting continued cellular proliferation [59-63].

            Stress-induced Premature Senescence (SIPS)

            Stress-induced premature senescence (SIPS), also known as premature senescence, culture shock and STASIS (stress or aberrant signalling-induced senescence) [64-66], occurs rapidly in response to a variety of intrinsic or extrinsic stressors, including DNA damage from ionising and non-ionising radiation, cytotoxic DNA damaging agents, oxidative stress and as a consequence of oncogenic activation [3,4,8,67]. Serrano et al (1997) were among the first to identify a form of SIPS that was not attributable to telomere attrition that they described as oncogene-induced senescence (OIS). The group showed that oncogenic ras expression permanently arrested primary human and rodent cells in G1 and that the cells displayed features similar to those of replicative senescence, including the accumulation of p53 and p16 [65]. Importantly the expression of the catalytic subunit of the telomerase enzyme hTERT has been shown not to abrogate SIPS, demonstrating that cellular senescence can be triggered prematurely independently of telomere attrition [68,69]. Thus, oncogenic activation and stressors that lead to DNA damage but which are independent of loss or dysfunction of telomeres, can initiate a response that results in a cellular phenotype indistinct to that observed for replicative senescence [63,70].

            Molecular Pathways of Senescence

            The initiating event for both replicative senescence and SIPS involves the recognition of DNA damage and the activation of the DNA damage response (DDR) pathway [3,56,71-74]. The key mediator in this process, ATM, phosphorylates important sensors and effectors of the DDR including H2AX, 53BP1 [31,75-77] and p53 leading to the up-regulation of cyclin-dependant kinase inhibitor p21, which in-turn acts to inhibit the action of CDK2 kinase activity arresting the cell cycle in G1 [76] (Figure 1). In addition, p21 also activates pRb through the inhibition of cyclin E/CDK2 [1]. Where SIPS differs from replicative senescence is in the formers dependence on the P16INK4 family of tumour suppressor proteins, which are activated upstream to pRb [10,78]. Accordingly, increased P16INK4A expression is considered as another useful marker of senescence in vitro, and indeed elevated protein levels have been detected in ageing baboon fibroblasts along with markers of telomere damage and SAHF [79]. The hypophosphorylated state of pRb results in inhibition of the transcription factor gene E2F and this acts to bring about G1 cell cycle arrest. For this reason, the p53 and p16/pRB dependent senescent pathways are not completely separable and as well as the common link through p21, pRB has been shown to regulate the activity of MDM2 which acts to control the stability of p53 [80]. Thus increased expression of p21 is important for senescence [81]. Consequently, the DNA damage response, apoptotic and senescence pathways share common molecular mediators through p53 and p21. What directs a cell to senesce or apoptose remains unclear, but cell type, the type of damaging agent and the dose administered may be important; as well as the post-translational modifications that p53 undergoes [82]. For instance in normal cells, senescence has been shown to be more favourable than apoptosis to deal with low levels of DNA damage, perhaps as the cell makes the decision to attempt to repair instead of removal from the cell pool [3,76,83]. By contrast, adult human dental pulp stem cells (DPSCs) were found to enter premature senescence in the G2 phase of the cell cycle after exposure to much higher doses (2-20 Gy) of ionising radiation, as detectable by phosphorylated p53 and increasing p16 expression observed over 13 days and SA-β-gal activity from day 3 after irradiation [84]. Possible mechanisms that may be involved in determining cellular fate include the status of the tumour suppressor phosphate and tensin homolog (PTEN). For instance, Lee J, et al (2010) showed that PTEN-deficient glioma cells preferentially entered senescence, while PTEN-proficient glioma cells generally apoptosed in response to ionising radiation. The authors concluded that SIPS may be a compensatory mechanism in place of apoptosis when PTEN tumour suppressor protein is absent [85].

            Figure 1

            Scheme highlighting initiating and molecular mediators of cellular senescence. The senescent phenotype includes expression of SA-β-galactosidase (SA-β-gal), increased expression of p16INK4a leading to cell cycle arrest and an increase in the secretion of pro-inflammatory factors termed as senescence-associated secretory phenotype (SASP). Senescent cells have been observed in normal ageing cells and in cells/tissues of various age-related pathologies.

            What is clear is that the convergence of multiple pathways through p53 and pRb are required to establish and maintain the senescent state and removal of either of these has been shown to prevent senescence in mouse embryonic fibroblasts [86]. In humans, it is thought that both p53 and pRb pathways must be inactivated in order to prevent the onset of cellular senescence [87], consistent with the majority if findings that show ~50% of all tumours show evidence of mutated/non-functional p53 and/or pRb [88,89]. Interestingly though, more recent experiments in humans and mice have shown that senescence can be prevented, or significantly delayed as a result of inactivation of either p53 or Rb alone while some cell types exhibit delayed onset of senescence upon p16 inactivation [90]. Therefore the relationship between these two pathways and the potential for redundancy in either pathway may provide further protection against senescence bypass in different cell types [3,91].

            Senescence and Cancer

            Senescence, in addition and in contrast to the previously noted beneficial tumour suppression and tissue repair effects, has also been linked to reduced tissue functionality and is increasingly thought to play a role in age-related pathologies such as cancer, Alzheimer's disease, diabetes and obesity [54,92-96]. For instance, senescent cells have been observed in many ageing mice tissues [20,97], baboon skin fibroblasts [79] and human tissues [29] indicating that senescence may have a causal role in ageing in vivo as well as in vitro. Senescent endothelial cells have been shown to increase in atherosclerosis, thrombosis and at sites of inflamed vascular endothelium [18,98], demonstrating possible links with pathology. Therefore, the accumulation of senescent haemopoietic stem cells has been suggested as a possible mediator for the decline in tissue regeneration and repair with age.

            Markers of DNA damage are known to accumulate in ageing stem cells [64,99-101] and other senescing human and mouse cell types [102-104]. For instance, studies employing the DSB marker, γ-H2AX, reveal γ-H2AX foci to accumulate in normal human fibroblasts, WI38 fibroblasts and PrEC prostate epithelial cells with increasing passage in a manner that correlates with an increasing fraction of SA-β gal positive cells. Specifically, early passage cultures show 0.2-0.3 γ-H2AX foci/cell increasing to 2.2-4.1 foci/cell in senescent cultures [31]. Further to this, radiation-induced γ-H2AX foci have both been shown to increase in both murine and human senescent cells in vitro [56,73,74] while, in vivo studies have shown the long-term expression of senescence markers, including an increased expression of p16INK4a to be coupled with the persistence of DNA damage foci 45 weeks post irradiation to a sub-lethal dose of radiation [105]. Interestingly, γ-H2AX foci (along with other DNA damage markers such as 53BP1) have been shown to localise both at telomeres as telomere-dysfunction-induced-foci (TIFs) in both early and late passage fibroblasts [106] and also throughout the genome as a consequence of ionising radiation exposure. Therefore γ-H2AX foci seen in senescent cells are not necessarily telomere-associated foci, representative of replicative senescence, but may represent SIPS-induced sites of DSB, highlighting that mediators of SIPS may contribute to age-related pathologies, including cancer. Accordingly, there is a relationship between cellular ageing and the accumulation of residual DNA damage both in vitro and in vivo, however as yet there is no evidence to determine whether senescence is a resultant part of ageing and age-related pathologies or whether it is a state that contributes to the development of ageing tissues and tissue pathology. Interestingly though, mouse models of accelerated ageing that are deficient for p16INK4a show delayed onset of age-related phenotypes, highlighting the role of increasing p16INK4a in maintaining the senescent state and its role in age-related decline of tissue regeneration and repair [107]. Further studies will hopefully decipher the evidential link between increasing populations of senescent cells and the contribution they have in the development of age-related pathologies such as cancer [108,109].

            Senescence-Associated Secretory Phenotype (SASP)

            It is well established that senescent cells secrete factors such as interleukins, chemokines, growth factors and proteases, encompassing what is known as the senescence-associated secretory phenotype (SASP) [15,32,110]. The function of SASP is to mediate the characteristic growth arrest of senescence via the autocrine activities of pro-inflammatory cytokines (including IL-6 and IL-8), in addition to pro-apoptotic protein insulin growth-factor binding protein 7 (IGFBP7), epithelial growth factors (heregulin and VEGF), matrix metalloproteinases including MMP-3 and plasminogen activator inhibitor 1 (PAI-1) [111-113]. Interestingly the name 'senescence-messaging secretome' (SMS) was proposed to highlight that the associated factors of the secretory phenotype were not only essential for initiating the senescent state but also for its maintenance and communication of this state to the local microenvironment [114,115]. A study that highlights this communication shows that re-activation of endogenous p53 in p53-deficient tumours in a mosaic mouse model of hepatocellular carcinoma led to tumour regression. This was proposed to occur through the induction of cellular senescence and up-regulation of inflammatory cytokines, triggering an innate immune response in vivo that ultimately led to tumour clearance [116]. Thus, inflammatory cytokines are necessary for both the establishment and maintenance of senescence, suggesting SASP/SMS products are important for the suppression of malignancy [111,114]. However, SASP is also known to influence the proliferation of neighbouring cells and disrupt tissue architecture [117], principally through these pro-inflammatory influences. For instance, an increase in VEGF, as a result of senescent fibroblasts, has been seen to stimulate tumour vascularisation and invasion of basement membranes [118]. Further, inflammation is thought of as a key mediator in cancer development and inflammatory cytokines and MMPs are being increasingly implicated as a contributing factor in this multistep process.

            What this suggests is that senescent cells can actually promote, in addition to preventing, the progression of malignancy; a relationship that is described as antagonistically pleiotrophic [21,118-121]. For instance, senescent human fibroblasts have been shown to stimulate pre-malignant and malignant fibroblasts to hyperproliferate and form tumours in mouse models when senescent cells comprised ~10% of the fibroblast population [122]. Close proximity of senescent fibroblasts to pre-neoplastic cells are thought to be the trigger for this change. Additionally, after exposure to the DNA-damaging agent bleomycin, human SIPS fibroblasts co-transplanted into xenografts of immunodeficient mice were seen to stimulate nearby cancer cells to proliferate, either directly or through local tissue damage and inflammation mediated by MMPs [123]. These findings support SASP as being an important mediator of the transformation process of pre-neoplatic cells. In addition, studies carried out by Zhou et al (2011) have shown SIPS in normal airway epithelial cells to result in an impairment of repair of drug-induced damage initiating a p38 MAPK dependant increase of pro-inflammatory cytokines that was subsequently seen to exacerbate the airway injury [124]. Interestingly this cytokine secretion, which primarily involves IL-6 and IL-8, is only established as a result of persistent DNA damage response signalling (DDR) and not as a result of transient signalling [125] suggesting the presence of long-lived, irreparable DNA lesions are important in this process.

            Thus alteration of the tissue microenvironment that results in the promotion of cell growth as a consequence of the senescence phenotype, through inflammation and persistent tissue damage [4,15,126] may provide a mechanism whereby senescent cells may also contribute to cancer promoting effects in otherwise normal tissues [127]. If demonstrated then senescence may functionally protect young animals from cancer via tumour suppression, whilst contributing to the deleterious effects in aged organisms through persistent inflammation and tissue injury [122,126].

            Ionising radiation and Senescence

            Ionising radiation is known to induce SIPS in both normal and cancer cell types after exposure to relatively high doses (10Gy) of radiation [125,128-131]. Thus, an important implication is what contribution, if any, senescence plays as a possible mediator of tumour recurrence after radiotherapy, given the effects of SASP in stimulating pre-neoplastic cells as discussed earlier. SIPS is also induced after exposure to lower doses of radiation [125,130,132,133] which similarly has consequences for understanding human cancer risk to radiation exposure, but this time within the context of SIPS in normal tissue after e.g. diagnostic exposures. For instance it is well established that radiation induces damage in cells that are not directly irradiated but which are in communication with irradiated cells. This radiation-induced non-targeted bystander (NTE) phenomenon is known to dominate at low radiation doses and to mediate a range of cellular effects such as DNA damage [134,135], cell death [136], cell proliferation, adaptive protective effects and malignant transformation [75,137-141]. To date, such NTE have been observed in microbeam-irradiated human tissue [141,142], in vivo animal models [143-145] and interestingly, in cells cultured in both non-irradiated tumour and senescent cell conditioned medium [14,75].

            Thus, it is reasonable to ask if there is a possible concordance between radiation-induced SIPS and SASP, and candidate mediators of NTE effects. Reactive oxygen species (ROS) are known to be important damaging agents involved in NTE [143,146,147], but additionally, activated macrophage, NO, IL-6, IL-8, IFN-γ and TGF-β have all also been implicated [143,144,148]. For instance, one study used radiation-induced AML susceptible and resistant mouse strains, CBA/ca and C57BL/6 respectively, to correlate radiation-induced up-regulation of gene expression of a M1 pro-inflamatory macrophage profile with more NTE in CBA/ca and an M2 anti-inflammatory macrophage profile, with less NTE in C57BL/6 strains [144]. Thus, candidate sources for the mediation of radiation-induced NTE include inflammation-associated cytokines and chemokines secreted from irradiated (or otherwise stressed) cells. Whether the irradiated cells that contribute to this plethora of inflammatory signals remain within the proliferative pool upon repair of damage or whether they become senescent is unknown however it is clear that even low doses of radiation induce SIPS and these cells subsequently secrete inflammatory cytokines, including IL-6 and IL-8 [27,110,133,149-153]. Interestingly Tsai et al (2009) showed that stromal fibroblasts that were induced to senesce after low dose radiation exposure stimulated the proliferation of breast-carcinoma cells when co-cultured in the same medium [125,130,132,133].

            This potential relationship between exposure to radiation, cellular age and deleterious inflammatory (NTE) responses is further demonstrated by human and animal studies which show a correlation exists between the immunological imbalances caused as a result of exposure to radiation and, those effects which are seen in normal aged immune cells, implying ionising radiation may accelerate immunological ageing [154]. For instance, the normal age-related decrease of total CD4+ T-cells was found to be ~4% per 10 years, compared to a radiation-induced decrease of ~2% Gy-1, equivalent to a 5 year age increase per 1 Gy [155,156]. This group also demonstrated a dose-dependent increase in CD25+/CD127- regulatory T-cells and attributed T-cell immunosenescence to a higher level of inflammatory markers in A-bomb survivors. For instance, changes in the immunological profiles of cytokines, known to be involved in the coordination of the inflammatory response (TNF-α, IFN-γ, IL-6 and IL-10) were seen in both A-bomb survivors and liquidators which may contribute to the persistent subclinical inflammatory status that is seen in these individuals [157-160]. There is the suggestion therefore that radiation-induced enhancement of inflammatory reactions might contribute to the development of radiation-induced disorders and premature ageing [155,161]. Indeed, it is also well known that A-bomb survivors show increased cardiovascular and respiratory diseases associated with persistent inflammation [162,163].

            Taken all together it is tempting to speculate radiation-induced SIPS and SASP as important mediators and, or amplifiers of radiation-induced NTE, which in turn may perpetuate inflammatory signals that subsequently, also contribute to increasing SIPS. In elucidating the importance of any such relationship in contributing to cancer risk, particularly at low doses, future work needs to understand the relevance of radiation quality, dose and dose rate in initiating SIPS and the long term tissue damage and pathological alterations that may arise as a consequence.

            Conclusion

            The beneficial tumour suppressive role of senescence whereby damage is prevented from being transmitted to daughter cells is well established. What is only recently becoming apparent is that pro-inflammatory factors such as those encompassing the senescence-associated secretory phenotype (SASP) are linked to cellular proliferation, a persistent low grade inflammation, elevated DNA damage foci and transformation of pre-neoplastic cells. Human populations are increasingly being exposed to ionising radiation from a range of diagnostic, treatment and occupational sources highlighting the potential risks of SASP whereby stress-induced premature senescence (SIPS) is initiated instead of apoptosis. The potential effects of this are two-fold; accelerated cellular ageing and an amplification of any detrimental effects produced by SASP. Thus, further research is required to understand the relationship between exposures to radiation, SIPS and how, in turn, SIPS may modify the biological effect of radiation exposure.

            Competing interests

            The authors declare that they have no competing interests.

            Authors' contributions

            RJS drafted the manuscript. RA participated in its design and helped to draft the manuscript. Both authors read and approved the final manuscript.

            References

            1. CampisiJCellular senescence as a tumor-suppressor mechanismTrends Cell Biol20011111S27S31[Cross Ref] [PubMed]

            2. WrightWEShayJWCellular senescence as a tumor-protection mechanism: the essential role of countingCurr Opin Genet Dev200111198103[Cross Ref] [PubMed]

            3. Ben-PorathIWeinbergRAThe signals and pathways activating cellular senescenceInt J Biochem Cell Biol2005375961976[Cross Ref] [PubMed]

            4. DavalosARCoppeJPCampisiJDesprezPSenescent cells as a source of inflammatory factors for tumour progressionCancer Metastasis Rev201029273283[Cross Ref] [PubMed]

            5. KuljuKSLehmanJMIncreased p53 Protein Associated with Aging in Human Diploid FibroblastsExp Cell Res19952172336345[Cross Ref] [PubMed]

            6. NaritaMNunezSHeardENaritaMLinAWHearnSASpectorDLHannonGJLoweSWRb-Mediated Heterochromatin Formation and Silencing of E2F Target Genes during Cellular SenescenceCell20031136703716[Cross Ref] [PubMed]

            7. MehtaISFiggitMClementsCSKillIRBridgerJMAlterations to Nuclear Architecture and Genome Behaviour in Senescent CellsAnn N Y Acad Sci200711001250263[Cross Ref] [PubMed]

            8. BlagosklonnyMVCell senescence and hypermitogenic arrestEMBO Rep200344358362[Cross Ref] [PubMed]

            9. OhtaniNYamakoshiKTakahashiAHaraEThe p16INK4a-RB pathway: molecular link between cellular senescence and tumour supressionJ Med Invest2004513-4146153[PubMed]

            10. GilJPetersGRegulation of the INK4b-ARF-INK4a tumour supressor locus: all for one and one for allNat Rev Mol Cell Biol200679667677[PubMed]

            11. CollinsCJSedivyJMInvolvement of the INK4a/Arf gene locus in senescenceAging Cell200323145150[Cross Ref] [PubMed]

            12. SmithJRPereira-SmithOMReplicative Senescence: Implications for in Vivo Aging and Tumor SuppressionScience199627352716367[Cross Ref] [PubMed]

            13. SerranoMColladoMSenescence in tumours: evidence from mice and humansNat Rev Cancer2010105157[Cross Ref] [PubMed]

            14. CoppeJ-PPatilCKRodierFSunYMunozDPGoldsteinJNelsonPSDesprezPYCampisiJSenescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the P53 tumour supressorPLoS Biol200861228532868[PubMed]

            15. CoppeJ-PDesprezP-YKrtolicaACampisiJThe Senescence-Associated Secretory Phenotype: The Dark Side of Tumor SuppressionAnnu Rev Pathol20105199118[Cross Ref] [PubMed]

            16. KrizhanovskyVYonMDickinsRAHearnSSimonJMiethingCYeeHZenderLLoweSWSenescence of Activated Stellate Cells Limits Liver FibrosisCell20081344657667[Cross Ref] [PubMed]

            17. JunJILauLFThe matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healingNat Cell Biol2010127676685[Cross Ref] [PubMed]

            18. EvanGId' Adda di FagagnaFCellular senescence: hot or what?Curr Opin Genet Dev20091912531[Cross Ref] [PubMed]

            19. BirchmeierWBirchmeierCEpithelial-mesenchymal transitions in development and tumour progressionEXS199574115[PubMed]

            20. WangESenescent Human Fibroblasts Resist Programmed Cell Death, and Failure to Suppress bcl2 Is InvolvedCancer Res1995551122842292[PubMed]

            21. CampisiJCancer, aging and cellular senescenceIn Vivo2000141183188[PubMed]

            22. RyuSJOhYSParkSCFailure of stress-induced down regulation of Bcl-2 contributes to apoptosis resistance in senescent human diploid fibroblastsCell Death Differ200714510201028[PubMed]

            23. RochettePJBrashDEProgressive apoptosis resistance prior to senescence and control by the anti-apoptotic protein BCL-xLMech Ageing Dev20081294207214[Cross Ref] [PubMed]

            24. ChaturvediVBaconPBodnarBNickoloffBJProliferating cultured human keratinocytes are more susceptible to apoptosis compared with mouse keratinocytesJ Invest Dermatol2004123612001203[Cross Ref] [PubMed]

            25. HampelBMalisanFNiedereggerHTestiRJansen-DurrPDifferential regulation of apoptotic cell death in senescent human cellsExp Gerontol20043911-1217131721[Cross Ref] [PubMed]

            26. LabinskyyNCsiszarAOroszZSmithKRiveraABuffensteinRUngvariZComparison of endothelial function, superoxide and H2O2 production, and vascular oxidative stress resistance between the longest-living rodent, the naked mole rat, and miceAm J Physiol Heart Circ Physiol20062916H2698H2704[Cross Ref] [PubMed]

            27. SheltonDNChangEWhittierPSChoiDFunkWDMicroarray analysis of replicative senescenceCurr Biol1999917939945[Cross Ref] [PubMed]

            28. KristnamurthyJp16INK4a induces an age-dependent decline in islet regenerative potentialNature2006443443457

            29. DimriGPA biomarker that identifies senescent human cells in culture and in ageing skin in vivoProc Natl Acad Sci USA19959293639367[PubMed]

            30. HerbigUJoblingWAChenBPCChenDJSedivyJMTelomere Shortening Triggers Senescence of Human Cells through a Pathway Involving ATM, p53, and p21CIP1, but Not p16INK4aMol Cell2004144501513[Cross Ref] [PubMed]

            31. SedelnikovaOASenescing human cells and ageing mice accumulate DNA lesions with unrepairable double-strand breaksNat Cell Biol200462168170[Cross Ref] [PubMed]

            32. CampisiJSenescent Cells, Tumor Suppression, and Organismal Aging: Good Citizens, Bad NeighborsCell20051204513522[Cross Ref] [PubMed]

            33. KuilmanTPeeperTSSenescence-messaging secretome: SMS-ing cellular stressNat Rev Cancer2009928134[Cross Ref] [PubMed]

            34. BridgerJMKillIRLichterPAssociation of pKi-67 with satellite DNA of the human genome in early G1 cellsChromosome Res199861324[Cross Ref] [PubMed]

            35. GerdesJLemkeHBaischHWackerHHSchwabUSteinHCell cycle analysis of a cell proliferation-associated human nuclear antigen defined by the monoclonal antibody Ki-67J Immunol1984133417101715[PubMed]

            36. GerdesJKi-67 and other proliferation markers useful for immunohistological diagnostic and prognostic evaluations in human malignanciesSemin Cancer Biol199013199206[PubMed]

            37. KillIRLocalisation of the Ki-67 antigen within the nucleolus. Evidence for a fibrillarin-deficient region of the dense fibrillar componentJ Cell Sci1996109612531263[PubMed]

            38. BridgerJMBoyleSKillIRBickmoreWARe-modelling of nuclear architecture in quiescent and senescent human fibroblastsCurr Biol2000103149152[Cross Ref] [PubMed]

            39. MehtaISFiggittMClementsCSKillIRBridgerJMAlterations to nuclear architecture and genome behaviour in senescent cellsAnn N Y Acad Sci20071100250263[Cross Ref] [PubMed]

            40. BridgerJMBoyleSKillIRBickmoreWARe-modelling of nuclear architecture in quiescent and senescent human fibroblastsCurr Biol2000103149152[Cross Ref] [PubMed]

            41. MeaburnKJCabuyEBonneGLevyNMorrisGENovelliGKillIRBridgerJMPrimary laminopathy fibroblasts display altered genome organization and apoptosisAging Cell200762139153[Cross Ref] [PubMed]

            42. HayflickLMoorheadPSThe serial cultivation of human diploid cell strainsExp Cell Res196125585621[Cross Ref] [PubMed]

            43. HayflickLHow and why we ageExp Gerontol1998337639653[Cross Ref] [PubMed]

            44. HayflickLThe cell biology of agingClin Geriatr Med1985121527[PubMed]

            45. ShayJWWrightWEHayflick, his limit, and cellular ageingNat Rev Mol Cell Biol2000117276[Cross Ref] [PubMed]

            46. HayflickLThe cell biology of agingJ Invest Dermatol1979731814[Cross Ref] [PubMed]

            47. ItahanaKCampisiJDimriGPMethods to detect biomarkers of cellular senescence: the senescence-associated beta-galactosidase assayMethods Mol Biol20073712131[Cross Ref] [PubMed]

            48. CristofaloVJSharfBBCellular senescence and DNA synthesis: Thymidine incorporation as a measure of population age in human diploid cellsExp Cell Res1973762419427[Cross Ref] [PubMed]

            49. MerzGSRossJDViability of human diploid cells as a function of in vitro ageJ Cell Physiol1969743219222[Cross Ref] [PubMed]

            50. ThomasEAl-BakerEDropcovaSDenyerSOstadNLloydAKillIRFaragherRGADifferent Kinetics of Senescence in Human Fibroblasts and Peritoneal Mesothelial CellsExp Cell Res19972361355358[Cross Ref] [PubMed]

            51. NgMHAminuddinBSHamizahSLynetteCMazlyzamALRuszymahBHICorrelation of donor age and telomerase activity with in vitro cell growth and replicative potential for dermal fibroblasts and keratinocytesJ Tissue Viability2009184109116[Cross Ref] [PubMed]

            52. HarleyCBFutcherABGreiderCWTelomeres shorten during ageing of human fibroblastsNature19903456274458460[Cross Ref] [PubMed]

            53. VaziriHBenchimolSReconstitution of telomerase activity in normal human cells leads to elongation of telomeres and extended replicative life spanCurr Biol199885279282[Cross Ref] [PubMed]

            54. ArtandiSEDePinhoRATelomeres and telomerase in cancerCarcinogenesis2010311918[Cross Ref] [PubMed]

            55. MakarovVLHiroseYLangmoreJPLong G Tails at Both Ends of Human Chromosomes Suggest a C Strand Degradation Mechanism for Telomere ShorteningCell1997885657666[Cross Ref] [PubMed]

            56. d' Adda di FagagnaFReaperPMClay-FarrenceLFieglerHCarrPVon ZglinickiTSaretzkiGCarterNPJSPA DNA damage checkpoint response in telomere-initiated senescenceNature20034266963194198[Cross Ref] [PubMed]

            57. ShayJWBacchettiSA survey of telomerase activity in human cancerEur J Cancer1997335787791[Cross Ref] [PubMed]

            58. HayflickLMortalilty and immortality at the cellular level. A reviewBiochemistry (Mosc)1997621111801190[PubMed]

            59. van SteenselBSmogorzewskaAde LangeTTRF2 Protects Human Telomeres from End-to-End FusionsCell1998923401413[Cross Ref] [PubMed]

            60. ShayJWWrightWETelomerase therapeutics for cancer: challenges and new directionsNat Rev Drug Discov200657577584[Cross Ref] [PubMed]

            61. BodnarAGOuelletteMFrolkisMHoltSEChiuCPMorinGBHarleyCBShayJWLichtsteinerSWrightWEExtension of life-span by introduction of telomerase into normal human cellsScience19982795349349352[Cross Ref] [PubMed]

            62. HayflickLThe illusion of cell immortalityBr J Cancer2000837841846[Cross Ref] [PubMed]

            63. PazolliEStewartSASenescence: the good the bad and the dysfunctionalCurr Opin Genet Dev20081814247[Cross Ref] [PubMed]

            64. ChenJSenescence and functional failure in hematopoietic stem cellsExp Haematol2004321110251032[Cross Ref]

            65. SerranoMThe Tumor Suppressor Protein p16INK4aExp Cell Res19972371713[Cross Ref] [PubMed]

            66. RamirezRDMoralesCPHerbertB-SRohdeJMPassonsCShayJWWrightWEPutative telomere-independent mechanisms of replicative aging reflect inadequate growth conditionsGenes Dev2001154398403[Cross Ref] [PubMed]

            67. SerranoMBlascoMPutting the stress on senescenceCurr Opin Cell Biol2001136748753[Cross Ref] [PubMed]

            68. RoblesSJAdamiGRAgents that cause DNA double strand breaks lead to P16INK4a enrichment and the premature senescence of normal fibroblastsOncogene199816911131125[Cross Ref] [PubMed]

            69. WeiSWeiWSedivyJMExpression of Catalytically Active Telomerase Does Not Prevent Premature Senescence Caused by Overexpression of Oncogenic Ha-Ras in Normal Human FibroblastsCancer Res199959715391543[PubMed]

            70. ZglinickiTVSaretzkiGLadhoffJFagagnaFdAdJacksonSPHuman cell senescence as a DNA damage responseMech Ageing Dev20051261111117[Cross Ref] [PubMed]

            71. RodierFKimS-HNijjarTYaswenPCampisiJCancer and aging: the importance of telomeres in genome maintenanceIn J Biochem Cell Biol2005375977990[Cross Ref]

            72. Ben-PorathIWeinbergRAWhen cells get stressed: an integrative view of cellular senescenceJ Clin Invest2004113813[PubMed]

            73. NakamuraAJChiangYJHathcockKSHorikawaISedelnikovaOAHodesRJBonnerWMBoth telomeric and non-telomeric DNA damage are determinants of mammalian cellular senescenceEpigenetics Chromatin2008116[Cross Ref] [PubMed]

            74. KastanMBZhanQEl-DeiryWSCarrierFJacksTWalshWVPlunkettBSVogelsteinBFornaceAJA mammalian cell cycle checkpoint pathway utilizing p53 and GADD45 is defective in ataxia-telangiectasiaCell1992714587597[Cross Ref] [PubMed]

            75. DickeyJSBairdBJRedonCESokolovMVSedelnikovaOABonnerWMIntercellular communication of cellular stress monitored by gamma-H2AX inductionCarcinogenesis2009301016861695[Cross Ref] [PubMed]

            76. AbrahamRTCheckpoint signalling: focusing on 53BP1Nat Cell Biol2002412277279[Cross Ref]

            77. LeeHKwakH-JChoI-TParkSHLeeC-HS1219 residue of 53BP1 is phosphorylated by ATM kinase upon DNA damage and required for proper execution of DNA damage responseBiochem Biophys Res Commun200937813236[Cross Ref] [PubMed]

            78. FinkelTSerranoMBlascoMAThe common biology of cancer and ageingNature20074487155767774[Cross Ref] [PubMed]

            79. JeyapalanJCFerreiraMSedivyJMHerbigUAccumulation of senescent cells in mitotic tissue of aging primatesMech Ageing Dev200712813644[Cross Ref] [PubMed]

            80. YapDBHsiehJKChanFSLuXmdm2: a bridge over the two tumour suppressors, p53 and RbOncogene1999185376817689[Cross Ref] [PubMed]

            81. ZhangJLiuWLTangDCChenLWangMPackSDZhuangZRodgersGPIdentification and characterization of a novel member of olfactomedin-related protein family, hGC-1, expressed during myeloid lineage developmentGene20022831-28393[Cross Ref] [PubMed]

            82. WebleyKBondJAJonesCJBlaydesJPCraigAHuppTWynford-ThomasDPosttranslational Modifications of p53 in Replicative Senescence Overlapping but Distinct from Those Induced by DNA DamageMol Cell Biol200020828032808[Cross Ref] [PubMed]

            83. SuzukiKMoriINakayamaYMiyakodaMKodamaSWatanabeMRadiation-induced senescence-like growth arrest requires TP53 function but not telomere shorteningRadiat Res20011551 Pt 2248253[PubMed]

            84. MuthnaDSoukupTVavrovaJMokryJCmielovaJVisekBJiroutovaAHavelekRSuchanekJFilipSEnglishDRezacovaMIrradiation of adult human dental pulp stem cells provokes activation of p53, cell cycle arrest, and senescence but not apoptosisStem Cells Dev2010191218551862[Cross Ref] [PubMed]

            85. LeeJJKimBCParkMJLeeYSKimYNLeeBLLeeJSPTEN status switches cell fate between premature senescence and apoptosis in glioma exposed to ionising radiationCell Death Differ2010184666677[PubMed]

            86. DiracAMGBernardsRReversal of Senescence in Mouse Fibroblasts through Lentiviral Suppression of p53J Biol Chem2003278141173111734[Cross Ref] [PubMed]

            87. SmogorzewskaAde LangeTDifferent telomere damage signalling pathways in human and mouse cellsEMBO2002211643384348[Cross Ref]

            88. KaulSCYaguchiTTairaKReddelRRWadhwaROverexpressed mortalin (mot-2)/mthsp70/GRP75 and hTERT cooperate to extend the in vitro lifespan of human fibroblastsExp Cell Res2003286196101[Cross Ref] [PubMed]

            89. ShayJWPereira-SmithOMWrightWEA role for both RB and p53 in the regulation of human cellular senescenceExp Cell Res199119613339[Cross Ref] [PubMed]

            90. AnsieauSBastidJDoreauAMorelA-PBouchetBPThomasCFauvetFPuisieuxIDoglioniCPiccininSMaestroRVoeltzelTSelmiAValsesia-WittmannSCaron de FromentelCPuisieuxAInduction of EMT by Twist Proteins as a Collateral Effect of Tumor-Promoting Inactivation of Premature SenescenceCancer Cell20081417989[Cross Ref] [PubMed]

            91. WeiWHerbigUWeiSDutriauxASedivyJMLoss of retinoblastoma but not p16 function allows bypass of replicative senescence in human fibroblastsEMBO200341110611066[Cross Ref]

            92. ErusalimskyJDKurzDJCellular senescence in vivo: Its relevance in ageing and cardiovascular diseaseExp Gerontol2005408-9634642[Cross Ref] [PubMed]

            93. PriceJSWatersJGDarrahCPenningtonCEdwardsDRDonellSTClarkIMThe role of chondrocyte senescence in osteoarthritisAging Cell2002115765[Cross Ref] [PubMed]

            94. DePinhoRAThe age of cancerNature20004086809248254[Cross Ref] [PubMed]

            95. BegheCBalducciLBiological basis of cancer in the older personCancer Treat Res2005124189221[Cross Ref] [PubMed]

            96. WuXPandolfiPPMouse models for multistep tumorigenesisTrends Cell Biol2001111129[Cross Ref] [PubMed]

            97. WangCJurkDMaddickMNelsonGMartin-RuizCVon ZglinickiTDNA damage response and cellular senescence in tissues of aging miceAging Cell200983311323[Cross Ref] [PubMed]

            98. ErusalimskyJDVascular endothelial senescence: from mechanisms to pathophysiologyJ Appl Physiol20091061326332[PubMed]

            99. RossiDJBryderDSeitaJNussenzweigAHoeijmakersJWeissmanILDeficiencies in DNA damage repair limit the function of haematopoietic stem cells with ageNature20074497160288291[Cross Ref] [PubMed]

            100. NijnikAWoodbineLMarchettiCDawsonSLambeTLiuCRodriguesNPCrockfordTLCabuyEVindigniAEnverTBellJISlijepcevicPGoodnowCCJeggoPACornallRJDNA repair is limiting for haematopoietic stem cells during ageingNature20074477145686690[Cross Ref] [PubMed]

            101. JanzenVForkertRFlemingHESaitoYWaringMTDombkowskiDMChengTDePinhoRASharplessNEScaddenDTStem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4aNature20064437110421426[PubMed]

            102. PaullTTRogakouEPYamazakiVKirchgessnerCUGellertMBonnerWMA critical role for histone H2AX in recruitment of repair factors to nuclear foci after DNA damageCurr Biol20001015886895[Cross Ref] [PubMed]

            103. RothkammKLobrichMEvidence for a lack of DNA double-strand break repair in human cells exposed to very low x-ray dosesProc Natl Acad Sci USA2003100950575062[Cross Ref] [PubMed]

            104. SedelnikovaOARogakouEPPanyutinIGBonnerWMQuantitative detection of (125)IdU-induced DNA double-strand breaks with gamma-H2AX antibodyRadiat Res20021584486492[Cross Ref] [PubMed]

            105. LeONRodierFFontaineFCoppeJ-PCampisiJDeGregoriJLaverdiereCKoktaVHaddadEBeausejourCMIonizing radiation-induced long-term expression of senescence markers in mice is independent of p53 and immune statusAging Cell201093398409[Cross Ref] [PubMed]

            106. BakkenistCJDrissiRWuJKastanMBDomeJSDisappearance of the Telomere Dysfunction-Induced Stress Response in Fully Senescent CellsCancer Res2004641137483752[Cross Ref] [PubMed]

            107. BakerDJJinFvan DeursenJMThe yin and yang of the Cdkn2a locus in senescence and agingCell Cycle200871827952802[Cross Ref] [PubMed]

            108. SikoraEArendtTBennettMNaritaMImpact of cellular senescence signature on ageing researchAgeing Res Rev2011101146152[Cross Ref] [PubMed]

            109. VijgJCampisiJPuzzles, promises and a cure for ageingNature2008454720810651071[Cross Ref] [PubMed]

            110. FreundAOrjaloAVDesprezP-YCampisiJInflammatory networks during cellular senescence: causes and consequencesTrends Mol Med2010165238246[Cross Ref] [PubMed]

            111. AcostaJCO'LoghlenABanitoARaguzSGilJControl of senescence by CXCR2 and its ligandsCell Cycle200871929562959[Cross Ref] [PubMed]

            112. WajapeyeeNSerraRWZhuXMahalingamMGreenMROncogenic BRAF Induces Senescence and Apoptosis through Pathways Mediated by the Secreted Protein IGFBP7Cell20081323363374[Cross Ref] [PubMed]

            113. KortleverRMHigginsPJBernardsRPlasminogen activator inhibitor-1 is a critical downstream target of p53 in the induction of replicative senescenceNat Cell Biol200688877884[PubMed]

            114. KuilmanTMichaloglouCVredeveldLCWDoumaSvan DoornRDesmetCJAardenLAMooiWJPeeperDSOncogene-Induced Senescence Relayed by an Interleukin-Dependent Inflammatory NetworkCell2008133610191031[Cross Ref] [PubMed]

            115. KuilmanTMichaloglouCMooiWJPeeperDSThe essence of senescenceGenes Dev2010242224632479[Cross Ref] [PubMed]

            116. XueWZenderLMiethingCDickinsRAHernandoEKrizhanovskyVCordon-CardoCLoweSWSenescence and tumour clearance is triggered by p53 restoration in murine liver carcinomasNature20074457128656660[Cross Ref] [PubMed]

            117. ParrinelloSCoppeJ-PKrtolicaACampisiJStromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiationJ Cell Sci20051183485496[Cross Ref] [PubMed]

            118. CoppeJ-PKauserKCampisiJBeausejourCMSecretion of Vascular Endothelial Growth Factor by Primary Human Fibroblasts at SenescenceJ Biol Chem2006281402956829574[Cross Ref] [PubMed]

            119. YeagerTRDeVriesSJarrardDFKaoCNakadaSYMoonTDBruskewitzRStadlerWMMeisnerLFGilchristKWNewtonMAWaldmanFMReznikoffCAOvercoming cellular senescence in human cancer pathogenesisGenes Dev1998122163174[Cross Ref] [PubMed]

            120. ReddelRRThe role of senescence and immortalization in carcinogenesisCarcinogenesis2000213477484[Cross Ref] [PubMed]

            121. CampisiJd'Adda di FagagnaFCellular senescence: when bad things happen to good cellsNature Reviews Mol Cell Biol200789729740[Cross Ref]

            122. KrtolicaAParrinelloSLockettSDesprezP-YCampisiJSenescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and agingProc Natl Acad Sci USA200198211207212077[Cross Ref] [PubMed]

            123. LiuDHornsbyPJSenescent Human Fibroblasts Increase the Early Growth of Xenograft Tumors via Matrix Metalloproteinase SecretionCancer Res200767731173126[Cross Ref] [PubMed]

            124. ZhouFOnizawaSNagaiAAoshibaKEpithelial cell senescence impairs repair process and exacerbates inflammation after airway injuryRespir Res20111278118[PubMed]

            125. RodierFCoppeJ-PParilCKHoeijmakersWAMMunozDPRazaSRFreundACampeauEDavalosARCampisiJPersistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretionNat Cell Biol2009118973979[Cross Ref] [PubMed]

            126. BassiPSaccoECancer and aging: The molecular pathwaysUrol Oncol2009276620627[Cross Ref] [PubMed]

            127. KrtolicaAOrtiz de SolorzanoCLockettSCampisiJQuantification of epithelial cells in coculture with fibroblasts by fluorescence image analysisCytometry20024927382[Cross Ref] [PubMed]

            128. SuzukiMBoothmanDAStress-induced premature senescence (SIPS) - influence of SIPS on radiotherapyJ Radiat Res200848105112[PubMed]

            129. RaffettoJDLeverkusMParkHYMenzoianJOSynopsis on cellular senescence and apoptosisJ Vasc Surg200134173177[Cross Ref] [PubMed]

            130. TsaiKKStuartJChuangYYLittleJBYuanZMLow-dose radiation-induced senescent stromal fibroblasts render nearby breast cancer cells radioresistantRadiat Res20091723306313[Cross Ref] [PubMed]

            131. MirzayansRScottACameronMMurrayDInduction of accelerated senescence by gamma radiation in human solid tumor-derived cell lines expressing wild-type TP53Radiat Res200516315362[Cross Ref] [PubMed]

            132. TsaiKKCChuangEY-YLittleJBYuanZ-MCellular Mechanisms for Low-Dose Ionizing Radiation-Induced Perturbation of the Breast Tissue MicroenvironmentCancer Res2005651567346744[Cross Ref] [PubMed]

            133. RodierFMunozDPTeachenorRChuVLeOBhaumikDCoppeJ-PCampeauEBeausejourCMKimSHDavalosARCampisiJDNA-SCARS: distinct nuclear structures that sustain damage-induced senescence growth arrest and inflammatory cytokine secretionJ Cell Sci20101246881[PubMed]

            134. AzzamEIde ToledoSMLittleJBDirect evidence for the participation of gap junction-mediated intercellular communication in the transmission of damage signals from alpha-particle irradiated to nonirradiated cellsProc Natl Acad Sci USA2001982473478[Cross Ref] [PubMed]

            135. LittleJBNagasawaHLiGCChenDJInvolvement of the nonhomologous end joining DNA repair pathway in the bystander effect for chromosomal aberrationsRadiat Res20031592262267[Cross Ref] [PubMed]

            136. Lyng FMSeymour CBMothersillCInitiation of apoptosis in cells exposed to medium from the progeny of irradiated cells: a possible mechanism for bystander-induced genomic instability?Radiat Res20021574365370[Cross Ref] [PubMed]

            137. NagasawaHEffect of dose rate on the survival of irradiated human skin fibroblastsRadiat Res19921323375379[Cross Ref] [PubMed]

            138. MothersillCSeymourCMedium from irradiated human epithelial cells but not human fibroblasts reduces the clonogenic survival of unirradiated cellsInt J Radiat Biol1997714421427[Cross Ref] [PubMed]

            139. LorimoreSAKadhimMAPocockDAPapworthDStevensDLGoodheadDTWrightEGChromosomal instability in the descendants of unirradiated surviving cells after alpha-particle irradiationProc Natl Acad Sci USA1998951057305733[Cross Ref] [PubMed]

            140. SawantSGRanders-PehrsonGMettingNFHallEJAdaptive response and the bystander effect induced by radiation in C3H 10T(1/2) cells in cultureRadiat Res20011562177180[Cross Ref] [PubMed]

            141. BelyakovOVMitchellSAParikhDRanders-PehrsonGMarinoSAAmundsonSAGeardCRBrennerDJBiological effects in unirradiated human tissue induced by radiation damage up to 1 mm awayProc Natl Acad Sci USA2005102401420314208[Cross Ref] [PubMed]

            142. SedelnikovaOANakamuraAKovalchukOKoturbashIMitchellSAMarinoSABrennerDJBonnerWMDNA Double-Strand Breaks Form in Bystander Cells after Microbeam Irradiation of Three-dimensional Human Tissue ModelsCancer Res200767942954302[Cross Ref] [PubMed]

            143. CoatesPJLorimoreSAWrightEGDamaging and protective cell signalling in the untargeted effects of ionizing radiationMutat Res20045681520[PubMed]

            144. CoatesPJRobinsonJILorimoreSAWrightEGOngoing activation of p53 pathway responses is a long-term consequence of radiation exposure in vivo and associates with altered macrophage activitiesJ Pathol20082145610616[Cross Ref] [PubMed]

            145. IlnytskyyYKoturbashIKovalchukORadiation-induced bystander effects in vivo are epigenetically regulated in a tissue-specific mannerEnviron Mol Mutagen2009502105113[Cross Ref] [PubMed]

            146. Rzeszowska-WolnyJPrzybyszewskiWMWidelMIonizing radiation-induced bystander effects, potential targets for modulation of radiotherapyEur J Pharmacol20096251-3156164[Cross Ref] [PubMed]

            147. BuonannoMde ToledoSMPainDAzzamEILong-term consequences of radiation-induced bystander effects depend on radiation quality and dose and correlate with oxidative stressRadiat Res20111754405415[Cross Ref] [PubMed]

            148. DieriksBDe VosWHDerradjiHBaatoutSVan OostveldtPMedium-mediated DNA repair response after ionizing radiation is correlated with the increase of specific cytokines in human fibroblastsMutat Res20106871-24048[PubMed]

            149. MilisAJTHoyleMMcCueHMMartiniHDifferential expression of metalloproteinases and tissue inhibitor of metalloproteinase genes in aged human fibroblastsExp Cell Res1992201373379[Cross Ref] [PubMed]

            150. WestMDPereira-SmithOMSmithJRReplicative senescence of human skin fibroblasts correlates with a loss of regulation and overexpression of collagenase activityExp Cell Res19891841138147[Cross Ref] [PubMed]

            151. PasiFFacoettiANanoRIL-8 and IL-6 bystander signalling in human glioblastoma cells exposed to gamma radiationAnticancer Res201030727692772[PubMed]

            152. McBrideWHPajonkFChiangCSSunJRNF-kappa B, cytokines, proteasomes, and low-dose radiation exposureMil Med20021676667[PubMed]

            153. SchaueDMcBrideWHLinks between Innate Immunity and Normal Tissue RadiobiologyRadiat Res20101734406417[Cross Ref] [PubMed]

            154. ParkH-RJoS-KLasting effects of an impairment of Th1-like immune response in gamma-irradiated mice: A resemblance between irradiated mice and aged miceCell Immunol2011267118[Cross Ref] [PubMed]

            155. KusunokiYYamaokaMKuboYHayashiTKasagiFDoupleEBNakachiKT-cell immunosenescence and inflammatory response in atomic bomb survivorsRadiat Res20101746870876[Cross Ref] [PubMed]

            156. KusunokiYKyoizumiSHiraiYSuzukiTNakashimaEKodamaKSeyamaTFlow cytometry measurements of subsets of T, B and NK cells in peripheral blood lymphocytes of atomic bomb survivorsRadiat Res19981502227236[Cross Ref] [PubMed]

            157. HayashiTKusunokiYHakodaMMorishitaYKuboYMakiMKasagiFKodamaKMacpheeDGKyoizumiSRadiation dose-dependent increases in inflammatory response markers in A-bomb survivorsInt J Radiat Biol2003792129136[PubMed]

            158. HayashiTMorishitaYKuboYKusunokiYHayashiIKasagiFHakodaMKyoizumiSNakachiKLong-term effects of radiation dose on inflammatory markers in atomic bomb survivorsAm J Med200511818386[Cross Ref] [PubMed]

            159. KusunokiYYamaokaMKasagiFHayashiTMacPheeDGKyoizumiSLong-last changes in the T-cell receptor V beta repertoires of CD4 memory T-cell populations in the peripheral blood of radiation-exposed peopleBr J Haematol20031226975984[Cross Ref] [PubMed]

            160. NakachiKHayashiTImaiKKusunokiYPerspectives on cancer immuno-epidemiologyCancer Science20049512921929[Cross Ref] [PubMed]

            161. YamadaMWongFLFujiwaraSAkahoshiMSuzukiGNoncancer disease incidence in atomic bomb survivors, 1958-1998Radiat Res20041616622632[Cross Ref] [PubMed]

            162. Van der MeerenATourdesFGremyOGrillonGAbramMCPoncyJLGriffithsNActivation of alveolar macrophages after plutonium oxide inhalation in rats: involvement in the early inflammatory responseRadiat Res20081705591603[Cross Ref] [PubMed]

            163. NeriishiENPersistent subclinical inflammation among A-bomb survivorsInt J Radiat Biol2001774475482[Cross Ref] [PubMed]

            Author and article information

            Contributors
            Journal
            Genome Integr
            Genome Integr
            Genome Integrity
            BioMed Central
            2041-9414
            2011
            11 August 2011
            : 2
            : 7
            Affiliations
            [1 ]Centre for Cell and Chromosome Biology and Centre for Infection, Immunity and Disease Mechanisms, Division of Biosciences, Brunel University, West London. UB8 3PH, UK
            Article
            2041-9414-2-7
            10.1186/2041-9414-2-7
            3169443
            21834983
            41785005-24b9-430a-ba96-2f947dac2c40
            Copyright ©2011 Sabin and Anderson; licensee BioMed Central Ltd.

            This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

            History
            : 12 April 2011
            : 11 August 2011
            Categories
            Review

            Genetics
            ionising radiation,premature senescence,sasp,inflammation,age-related pathologies

            Comments

            Comment on this article