Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Feb 1.
Published in final edited form as: Curr Opin Genet Dev. 2010 Nov 17;21(1):107–112. doi: 10.1016/j.gde.2010.10.005

Cellular senescence: putting the paradoxes in perspective

Judith Campisi *
PMCID: PMC3073609  NIHMSID: NIHMS254508  PMID: 21093253

Summary

Cellular senescence arrests the proliferation of potential cancer cells, and so is a potent tumor suppressive mechanism, akin to apoptosis. Or is it? Why did cells evolve an anti-cancer mechanism that arrests, rather than kills, would-be tumor cells? Recent discoveries that senescent cells secrete growth factors, proteases and cytokines provide a shifting view – from senescence as a cell autonomous suppressor of tumorigenesis to senescence as a means to mobilize the systemic and local tissue milieu for repair. In some instances, this mobilization benefits the organism, but in others it can be detrimental. These discoveries provide potential mechanisms by which cellular senescence might contribute to the diverse, and seemingly incongruent, processes of tumor suppression, tumor promotion, tissue repair and aging.

Keywords: Aging, Cancer, Inflammation, Tumor suppression, Wound Healing

Introduction

Cellular senescence was first formally described nearly 5 decades ago as the process that limits the proliferation (growth) of normal human cells in culture [1]. This early study included two prescient -- but seemingly contradictory -- speculations. First, it speculated that the cellular changes associated with the growth arrest might reflect degenerative changes that are hallmarks of organismal aging. Second, it speculated that the senescence growth arrest might suppress the development of cancer in vivo.

In the ensuing five decades, remarkable progress has been made in understanding the causes and nature of cellular senescence. There is now substantial evidence that the senescence response suppresses malignant tumorigenesis, and mounting, albeit still indirect, evidence that it can promote aging. Further, recent findings paint an increasingly complex picture of cellular senescence. Under some circumstances, senescent cells can promote tumor progression, in obvious and apparent contradiction to its being tumor suppressive. And under other circumstances, senescent cells appear to aid tissue repair. Emerging data now provide a framework for understanding the strikingly different consequences of cellular senescence, and suggest strategies for harnessing the benefits of this process, while mitigating its potentially deleterious effects.

Senescent cells – in culture and in vivo

Cell culture experiments have identified several hallmarks of senescent cells, which have been used to identify these cells in mammalian tissues. However, there is as yet no single marker or characteristic that is unique to senescent cells. Further, not all senescent cells express all possible senescence markers. Nonetheless, senescent cells can be identified by an aggregate of phenotypes (Fig. 1). Some prominent senescence-associated characteristics are:

Figure 1. Characteristics and documented presence of senescent cells.

Figure 1

Potentially oncogenic stimuli induce cellular senescence via activation of the p53 and pRB tumor suppressor pathways. The senescent phenotype develops over several days in culture, and, depending on the type of cell and stimulus, includes expression of SA-Bgal activity and p16INK4a, development of DNA-SCARS that provide continuous DDR signals and SAHF, and the expression of a SASP. To varying extents, these senescence-associated characteristics have been used to identify senescent cells in vivo in mouse, non-human primate and human tissues. Conditions under which senescence cells have been identified in vivo include normal aging, damaged or wounded tissues, degenerative pathologies of aging, and hyperplastic, preneoplastic and early neoplastic lesions.

  1. The classic hallmark of cellular senescence is an essentially irreversible growth arrest, generally in response to potentially oncogenic stimuli [2]. The irreversibility of the arrest -- and fact that senescence is an oncogenic stress response -- distinguish senescent cells from quiescent or terminally differentiated cells.

  2. A potent inducer of senescence is (epi)genomic stress, which can result from direct DNA damage, dysfunctional telomeres, disrupted chromatin or strong mitogenic signals, such as those caused by certain oncogenes [2]. These stressors generate persistent DNA damage response (DDR) signals that emanate from stable, cytologically detectable nuclear foci [3] or DNA-SCARS (DNA segments with chromatin alterations reinforcing senescence) [4]. The persistent DDR foci sustain the activity of the p53 tumor suppressor [4,5], which maintains the senescence growth arrest by stimulating the transcription of genes that inhibit cell cycle progression [2].

  3. Senescent cells generally adopt an enlarged morphology [1], and express a senescence-associated beta-galactosidase (SA-Bgal) activity that is cytologically detectable in freshly fixed cells or tissues [6].

  4. Many senescent cells express p16INK4a, a cyclin-dependent kinase inhibitor and tumor suppressor [7]. p16INK4a is an upstream activator of the pRB tumor suppressor, which, in some cells, orchestrates the formation of cytologically detectable senescence-associated heterochromatin foci (SAHF) [8,9]. These foci silence the expression of genes that are needed for cell cycle progression.

  5. Senescent cells that harbor DNA-SCARS secrete numerous cytokines, growth factors and proteases that have wide-ranging autocrine and paracrine activities [10]. This senescence-associated secretory phenotype (SASP), as discussed below, is key to understanding many of the diverse effects attributed to senescent cells.

Most division-competent cells, including some tumor cells, can undergo senescence when appropriately stressed [2,11]. Further, the senescence-associated characteristics and markers described above (Fig. 1) have been used to identify senescent cells in a variety of mammalian tissues. So, when and where are these cells found?

Cellular senescence and aging

The first studies of cellular senescence in vivo showed that senescent cells increase with age (Fig. 1), support one of the two prescient early speculations. The increase in senescent cells occurs mainly in tissues that contain mitotically competent cells, and has now been documented in many rodent, non-human primate and human tissues [e.g., see 6,1214]. Moreover, senescent cells have been identified at sites of several degenerative age-related pathologies (Fig. 1) – examples include atherosclerosis, renal tubulointerstitial fibrosis and glomerulosclerosis, osteoarthritis, slow healing venous ulcers, and degenerating intervertebral discs [13,1518]. Do senescent cells reflect the aging process and/or the development of age-related pathology? Or do they play causative roles in aging and/or age-related disease? At present, there are no definitive answers to these questions. It has not yet been possible, for example, to eliminate senescent cells and demonstrate improved tissue function or amelioration of age-related pathology. Nonetheless, recent data suggest two mechanisms by which cellular senescence might actively drive aging and age-related pathology.

First, most adult stem cells are capable of undergoing senescence, and cellular senescence may account at least in part for the age-related decline in the number and/or function of certain stem cells in adult organisms [1923]. Thus, an accumulation of senescent stem cells may contribute to the decline in tissue repair and regeneration that is a hallmark of aging organisms. Second, the SASP includes factors that control many vital processes within tissues – cell growth, motility and differentiation, as well as tissue structure and vascularization [24,25] (Table 1). Factors secreted by senescent cells might therefore disrupt normal tissue structures and functions [26], including stem cell niches [27]. Finally, the SASP also includes many potent inflammatory cytokines and mediators of inflammatory reactions [28,29] (Table 1). This aspect of the SASP is noteworthy because a widespread feature of aging tissues is low-level chronic inflammation, without obvious microbial infection; furthermore, inflammation is an established cause or contributor to virtually every major age-related disease, both degenerative and hyperproliferative [30,31]. It is therefore possible that senescent cells are a source of the ‘sterile’ inflammation that is a hallmark of aging tissues and driver of multiple age-related pathologies.

Table 1.

Autocrine and paracrine activities of selected SASP factors

Factor Symbol Major Activities
Amphiregulin AREG Cell proliferation
Granulocyte-macrophage colony stimulating factor GM-CSF Hematopoietic stem cell differentiation; inflammation
Growth-related oncogene(s) (CXC chemokines) GROs
CXCLs
Cell proliferation; cell migration/invasion
Insulin-like growth factor binding protein-7 IGFBP-7 Apoptosis; autocrine growth arrest
Interleukin-6 IL-6 Epithelial-to-mesenchyme transition; cell migration/invasion; inflammation; autocrine growth arrest
Interleukin-8 IL-8
CXCL8
Epithelial-to-mesenchyme transition; cell migration/invasion; inflammation; autocrine growth arrest
Matrix metalloproteinase(s) MMPs Tissue remodeling; cell migration/invasion; wound healing (resolution of fibrobrosis)
Monocyte chemoattractant proteins (CCL chemokines) MCPs
CCLs
Inflammation; cell migration/invastion
Plasminogen activator inhibitor-1 PAI-1 Wound healing; autocrine growth arrest
Vascular endothelial growth factor VEGF Endothelial cell migration/invasion; angiogenesis

Cellular senescence and tumor suppression

Senescent cells have also been found in hyperplastic lesions such as benign prostatic hyperplasia [32], premalignant lesions such as melanocytic nevi and lung adenomas [33,34], and early stage malignancies such as non-invasive pancreatic and prostate cancers [34,35]. Of course, cancer is primarily an age-related disease, despite having characteristics that are quite distinct from the degenerative diseases of aging. Does the presence of senescent cells in pre/early-cancers have functional significance? In this case, the answer is clearly yes. As discussed below, mounting evidence strongly supports the early speculation that cellular senescence is a tumor suppressive mechanism.

First, it is firmly established that mutations that inactivate either the p53 or pRB pathway – both of which are critical for a proper senescence response (Fig. 1) – greatly increase the susceptibility to developing cancer in mice and humans [2,7,36]. Second, although senescent cells are often prominent in premalignant or early cancer lesions, they are rarely found in the aggressive cancers that eventually develop from these lesions [34,35]. In addition, genotoxic chemotherapy can induce a senescence response in some tumor cells, and this response is associated with a cessation of tumor growth and eventual regression [11,37]. Third, and perhaps most importantly, dismantling the senescence response – for example, by inactivating p53– markedly accelerates the development of malignant tumors from premalignant lesions [35].

It is generally assumed that the growth arrest is the major mechanism by which cellular senescence suppresses malignant tumorigenesis [2,7,36,38]. This is undoubtedly the case. However, there is also evidence that some of the factors secreted by senescent cells help reinforce the senescence growth arrest in an autocrine manner [3942]. These factors include the pro-inflammatory cytokines IL (interleukin)-6 and IL-8, but also factors such as the pro-apoptotic protein IGFBP (insulin-like growth factor binding protein)-7 and PAI (plasminogen activator inhibitor)-1 (Table 1).

There are now numerous examples of the senescence response acting to suppress cancer progression in both mice and humans [38]. It was somewhat surprising, then, when evidence emerged showing that senescent cells might also promote cancer progression.

Cellular senescence and tumor promotion

Cancer is among the pathologies that are fueled by inflammation [43]. Thus, the inflammatory cytokines that comprise the SASP could, in principle, also contribute to the development of age-related cancer by stimulating inflammation. In addition, the SASP includes factors that can induce -- in neighboring cells -- phenotypes that are associated with aggressive cancer cells [10,25] (Table 1). Examples of such SASP factors include amphiregulin and GRO (growth-related oncogene)-α, which stimulate cell proliferation; VEGF (vascular endothelial growth factor), which stimulates angiogenesis; and the pro-inflammatory cytokines IL-6 and IL-8, which can induce an epithelial-to-mesenchyme transitions and epithelial cell migration and invasion.

More direct evidence for tumor promoting effects of senescent cells comes from mouse xenograft experiments in which senescent stromal cells are co-injected with premalignant, or even frankly malignant, epithelial cells. In these studies, senescent cells, but not their non-senescent counterparts, stimulate tumor progression and tumor growth [24,4446]. In at least one case, the tumor promoting effects of senescence cells could be attributed to their secretion of matrix metalloproteinases (MMPs) [45], which are prominent SASP components [24]. It is not yet know whether senescent cells stimulate the progression of naturally occurring tumor in vivo, but at least in mouse xenografts they are clearly capable of promoting the malignant progression of precancerous cells, as well as established tumor cells.

Cellular senescence and tissue repair

A quick glance at the factors that comprise the SASP reveals many proteins that are important for wound healing, tissue repair or tissue regeneration [24,25]. Two recent studies show that senescent cells can indeed participate in repair or regenerative processes in vivo in mice.

In one case, acute liver injury stimulated hepatic stellate cells to proliferate and produce an extracellular matrix-rich fibrotic scar; the stellate cells subsequently underwent senescence, accompanied by the secretion of MMPs and dissolution of the fibrotic scar [47]. When stellate cells were rendered incapable of undergoing senescence (due to genetic deficiency in either the p53 or pRB pathways), the liver injury resulted in severe fibrosis that did not resolve. Thus, at least in this model, the senescence response was important for limiting the extent of fibrosis after tissue damage. In a second case, skin the extracellular matrix protein CCN1 was expressed in skin wounds and induced a senescence response in resident fibroblasts and myofibroblasts [48]. Mice engineered to express a CCN1 protein that fails to bind fibroblasts and induce senescence developed excessive fibrosis during wound healing, presumably due to the deficiency in MMPs at the site of the wound.

These findings suggest that senescent cells, and particularly the SASP, may function to communicate cellular damage or dysfunction to the surrounding tissue, and stimulate repair.

Reconciling the apparent paradoxes

Two processes to which cellular senescence contributes (tumor suppression and tissue repair) are clearly beneficial, whereas two others (aging and tumor promotion) are clearly detrimental (Fig. 2). How can these apparently paradoxical activities be reconciled?

Figure 2. Selected and unselected activities of senescent cells.

Figure 2

Cellular senescence contributes to four biological processes through the cell autonomous growth arrest and paracrine activities of the SASP. We hypothesize that the growth arrest and SASP are selected traits that ensure tumor suppression and facilitate tissue repair, respectively. However, the SASP also has unselected activities that can cause or contribute to tumor promotion and aging.

A teleological answer to this question is provided by theories that explain the evolution of late life phenotypes [49]. Most organisms evolve in environments that are rich in fatal extrinsic hazards (starvation, infection, predation, etc). In such environments, old individuals are rare. Consequently, selection against processes that promote late life disabilities is weak. Thus, evolutionary theory holds that aging phenotypes, including age-related disease, are a consequence of the declining force of natural selection with age. An extension of this theory is the concept of antagonistic pleiotropy [49]: a biological process that was selected to promote fitness in young organisms (for example, a tumor suppressive mechanism) can be deleterious in old organisms (for example, promote age-related disease, including late life cancer).

From the evolutionary arguments and documented biological activities of cellular senescence, we hypothesize that the senescence response evolved to both suppress the development of cancer and promote tissue repair in young organisms (Fig. 2). This idea suggests that cellular senescence is not simply a tumor suppressive failsafe mechanism, redundant to apoptosis. Rather, it may also be an important response for resolving tissue damage. A role for senescence in tissue repair would also explain the evolution of the SASP and the finding that senescence-associated inflammatory cytokine secretion is controlled by DDR signaling [5]. The dark side (unselected activities) of the senescence response, then, would be revealed only late in life, when the age-dependent accumulation of senescent cells could create local sites of chronic inflammation, tissue remodeling and other features of wound healing – which are also known to promote the development of cancer (Fig. 2).

Conclusions

There are now at least four major processes to which cellular senescence is thought to contribute– tumor suppression, aging, tumor promotion and tissue repair (Fig. 2). The apparent paradox -- that two of these processes benefit the organism, whereas two of the processes compromise organismal fitness – can be reconciled by understanding how aging phenotypes evolved. And the apparent redundancy to apoptosis as a tumor suppressive mechanism and secretory phenotype of senescent cells can be understood if the senescence response evolved to allow damaged cells to communicate their compromised state to the surrounding tissue and prepare the tissue for repair.

Acknowledgments

The author gratefully acknowledges the many laboratory members and colleagues whose lively discussions contributed to the ideas in this review. Research described in the review was funded by grants from the US National Institutes of Health and fellowships from the American Federation for Aging Research, Hillblom Foundation, Dutch Cancer Society and US National Science Foundation.

Abbreviations

DDR

DNA damage response

DNA-SCARS

DNA segments with chromatin alterations reinforcing senescence

MMP

Matrix metalloproteinase

SA-Bgal

Senescence-associated beta-galactosidase

SAHF

Senescence-associated heterochromatin foci

SASP

Senescence-associated secretory phenotype

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References and recommended reading

  • 1.Hayflick L. The limited in vitro lifetime of human diploid cell strains. Exp Cell Res. 1965;37:614–636. doi: 10.1016/0014-4827(65)90211-9. [DOI] [PubMed] [Google Scholar]
  • 2.Campisi J, d'Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nature Rev Molec Cell Biol. 2007;8:729–740. doi: 10.1038/nrm2233. [DOI] [PubMed] [Google Scholar]
  • *3.von Zglinicki T, Saretzki G, Ladhoff J, d'Adda di Fagagna F, Jackson SP. Human cell senescence as a DNA damage response. Mech Ageing Dev. 2005;126:111–117. doi: 10.1016/j.mad.2004.09.034. [DOI] [PubMed] [Google Scholar]
  • *4.Rodier F, Munoz DP, Teachenor R, Chu V, Le O, Bhaumik D, Coppe JP, Campeau E, Beausejour C, Kim SH, et al. DNA-SCARS: Distinct nuclear structures that sustain damage-induced senescence growth arrest and inflammatory cytokine secretion. J Cell Sci. doi: 10.1242/jcs.071340. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *5.Rodier F, Coppe JP, Patil CK, Hoeijmakers WAM, Munoz D, Raza SR, Freund A, Campeau E, Davalos AR, Campisi J. Persistent DNA damage signaling triggers senescence-associated inflammatory cytokine secretion. Nature Cell Biol. 2009;11:973–979. doi: 10.1038/ncb1909. These papers describe the intimate relationship between DDR signaling and senescence-associated phenotypes, including the secretory phenotypes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano EE, Linskens M, Rubelj I, Pereira-Smith OM, et al. A novel biomarker identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA. 1995;92:9363–9367. doi: 10.1073/pnas.92.20.9363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ohtani N, Yamakoshi K, Takahashi A, Hara E. The p16INK4a-RB pathway: molecular link between cellular senescence and tumor suppression. J Med Invest. 2004;51:146–153. doi: 10.2152/jmi.51.146. [DOI] [PubMed] [Google Scholar]
  • 8.Narita M, Nunez S, Heard E, Narita M, Lin AW, Hearn SA, Spector DL, Hannon GJ, Lowe SW. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003;113:703–716. doi: 10.1016/s0092-8674(03)00401-x. [DOI] [PubMed] [Google Scholar]
  • 9.Zhang R, Poustovoitov MV, Ye X, Santos HA, Chen W, Daganzo SM, Erzberger JP, Serebriiskii IG, Canutescu AA, Dunbrack RL, et al. Formation of macroH2A-containing senescence-associated heterochromatin foci and senescence driven by ASF1a and HIRA. Dev Cell. 2005;8:19–31. doi: 10.1016/j.devcel.2004.10.019. [DOI] [PubMed] [Google Scholar]
  • **10.Young AR, Narita M. SASP reflects senescence. EMBO Rep. 2009;10:228–230. doi: 10.1038/embor.2009.22. This review suscinctly but comprehensively summarizes the features and functions of senescence-associated secretory phenotypes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Shay JW, Roninson IB. Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene. 2004;23:2919–2933. doi: 10.1038/sj.onc.1207518. [DOI] [PubMed] [Google Scholar]
  • 12.Jeyapalan JC, Ferreira M, Sedivy JM, Herbig U. Accumulation of senescent cells in mitotic tissue of aging primates. Mech Ageing Dev. 2007;128:36–44. doi: 10.1016/j.mad.2006.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Erusalimsky JD, Kurz DJ. Cellular senescence in vivo: its relevance in ageing and cardiovascular disease. Exp Gerontol. 2005;40:634–642. doi: 10.1016/j.exger.2005.04.010. [DOI] [PubMed] [Google Scholar]
  • 14.Wang C, Jurk D, Maddick M, Nelson G, Martin-Ruiz C, von Zglinicki T. DNA damage response and cellular senescence in tissues of aging mice. Aging Cell. 2009;8:311–323. doi: 10.1111/j.1474-9726.2009.00481.x. [DOI] [PubMed] [Google Scholar]
  • 15.Ding G, Franki N, Kapasi AA, Reddy K, Gibbons N, Singhal PC. Tubular cell senescence and expression of TGF-beta1 and p21(WAF1/CIP1) in tubulointerstitial fibrosis of aging rats. Exp Mol Path. 2001;70:43–53. doi: 10.1006/exmp.2000.2346. [DOI] [PubMed] [Google Scholar]
  • 16.Price JS, Waters J, Darrah C, Pennington C, Edwards DR, Donell ST, Clark IM. The role of chondrocyte senescence in osteoarthritis. Aging Cell. 2002;1:57–65. doi: 10.1046/j.1474-9728.2002.00008.x. [DOI] [PubMed] [Google Scholar]
  • 17.Roberts S, Evans EH, Kletsas D, Jaffray DC, Eisenstein SM. Senescence in human intervertebral discs. Eur Spine J. 2006;15:312–316. doi: 10.1007/s00586-006-0126-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Stanley A, Osler T. Senescence and the healing rates of venous ulcers. J Vasc Surg. 2001;33:1206–1211. doi: 10.1067/mva.2001.115379. [DOI] [PubMed] [Google Scholar]
  • 19.Chen J. Senescence and functional failure in hematopoietic stem cells. Exp Hematol. 2004;32:1025–1032. doi: 10.1016/j.exphem.2004.08.001. [DOI] [PubMed] [Google Scholar]
  • 20.Zhou S, Greenberger JS, Epperly MW, Goff JP, Adler C, Leboff MS, Glowacki J. Age-related intrinsic changes in human bone-marrow-derived mesenchymal stem cells and their differentiation to osteoblasts. Aging Cell. 2008;7:335–343. doi: 10.1111/j.1474-9726.2008.00377.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Janzen V, Forkert R, Fleming H, Saito Y, Waring MT, Dombkowski DM, Cheng T, DePinho RA, Sharpless NE, Scadden DT. Stem cell aging modified by the cyclin-dependent kinase inhibitor, p16INK4a. Nature. 2006;443:421–426. doi: 10.1038/nature05159. [DOI] [PubMed] [Google Scholar]
  • 22.Krishnamurthy J, Ramsey MR, Ligon KL, Torrice C, Koh A, Bonner-Weir S, Sharpless NE. p16INK4a induces an age-dependent decline in islet regenerative potential. Nature. 2006;443:453–457. doi: 10.1038/nature05092. [DOI] [PubMed] [Google Scholar]
  • 23.Molofsky AV, Slutsky SG, Joseph NM, He S, Pardal R, Krishnamurthy J, Sharpless NE, Morrison SJ. Increasing Ink4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006;443:448–452. doi: 10.1038/nature05091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **24.Coppe JP, Patil CK, Rodier F, Krtolica A, Beausejour C, Parrinello S, Hodgson G, Chin K, Desprez PY, Campisi J. A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS ONE. 2010;5:e9188. doi: 10.1371/journal.pone.0009188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **25.Coppe JP, Patil CK, Rodier F, Sun Y, Munoz D, Goldstein J, Nelson PS, Desprez PY, Campisi J. Senescence-associated secretory phenotypes reveal cell non-automous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. doi: 10.1371/journal.pbio.0060301. These papers provide comprehensive analyses of the SASPs of human and mouse cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Campisi J. Senescent cells, tumor suppression and organismal aging: Good citizens, bad neighbors. Cell. 2005;120:513–522. doi: 10.1016/j.cell.2005.02.003. [DOI] [PubMed] [Google Scholar]
  • 27.Drummond-Barbosa D. Stem cells, their niches and the systemic environment: an aging network. Genetics. 2008;180:1787–1797. doi: 10.1534/genetics.108.098244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Coppé JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5 :99–118. doi: 10.1146/annurev-pathol-121808-102144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Freund A, Orjalo A, Desprez PY, Campisi J. Inflammatory networks during cellular senescence: causes and consequences. Trends Molec Med. 2010;16:238–248. doi: 10.1016/j.molmed.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chung HY, Cesari M, Anton S, Marzetti E, Giovannini S, Seo AY, Carter C, Yu BP, Leeuwenburgh C. Molecular inflammation: underpinnings of aging and age-related diseases. Ageing Res Rev. 2009;8:18–30. doi: 10.1016/j.arr.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Franceschi C, Capri M, Monti D, Giunta S, Olivieri F, Sevini F, Panourgia MP, Invidia L, Celani L, Scurti M, et al. Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mech Ageing Dev. 2007;128:92–105. doi: 10.1016/j.mad.2006.11.016. [DOI] [PubMed] [Google Scholar]
  • 32.Castro P, Giri D, Lamb D, Ittmann M. Cellular senescence in the pathogenesis of benign prostatic hyperplasia. Prostate. 2003;55:30–38. doi: 10.1002/pros.10204. [DOI] [PubMed] [Google Scholar]
  • **33.Michaloglou C, Vredeveld LCW, Soengas MS, Denoyelle C, Kuilman T, van der Horst CMAM, Majoor DM, Shay JW, Mooi WJ, Peeper DS. BRAFE600-associated senescence-like cell cycle arrest of human nevi. Nature. 2005;436:720–724. doi: 10.1038/nature03890. [DOI] [PubMed] [Google Scholar]
  • **34.Collado M, Gil J, Efeyan A, Guerra C, JSA, Barradas M, Benguria A, Zaballos A, Flores JM, Barbacid M, et al. Tumor biology: senescence in premalignant tumours. Nature. 2005;436:642. doi: 10.1038/436642a. [DOI] [PubMed] [Google Scholar]
  • **35.Chen Z, Trotman LC, Shaffer D, Lin H, Dotan ZA, Niki M, Koutcher JA, Scher HI, Ludwig T, Gerald W, et al. Critical role of p53 dependent cellular senescence in suppression of Pten deficient tumourigenesis. Nature. 2005;436:725–730. doi: 10.1038/nature03918. These papers show that preneoplastic or early neoplastic lesions are replete with senescent cells, whereas the cancers that emerge from these lesions are devoid of senescent cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rodier F, Campisi J, Bhaumik D. Two faces of p53: aging and tumor suppression. Nucleic Acids Res. 2007;35:7475–7584. doi: 10.1093/nar/gkm744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Schmitt CA. Cellular senescence and cancer treatment. Biochim Biophys Acta. 2007;1775:5–20. doi: 10.1016/j.bbcan.2006.08.005. [DOI] [PubMed] [Google Scholar]
  • 38.Collado M, Serrano M. Senescence in tumours: evidence from mice and humans. Nature Rev Cancer. 2010;10:51–57. doi: 10.1038/nrc2772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *39.Acosta JC, O'Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, Furnagalli M, DaCosta M, Brown C, Popov N, et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008;133:1006–1018. doi: 10.1016/j.cell.2008.03.038. [DOI] [PubMed] [Google Scholar]
  • *40.Kuilman T, Michaloglou C, Vredeveld LCW, Douma S, van Doorn R, Desmet CJ, AAL, Mooi WJ, Peeper DS. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008;133:1019–1031. doi: 10.1016/j.cell.2008.03.039. [DOI] [PubMed] [Google Scholar]
  • *41.Wajapeyee N, Serra RW, Zhu X, Mahalingam M, Green MR. Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell. 2008;132:363–374. doi: 10.1016/j.cell.2007.12.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *42.Kortlever RM, Higgins PJ, Bernards R. Plasminogen activator inhibitor-1 is a critical downstream target of p53 in the induction of replicative senescence. Nature Cell Biol. 2006;8:877–884. doi: 10.1038/ncb1448. Each of these papers describes how a protein factor (IL-8, IL-6, IGFBP-7 and PAI-1) that is secreted by senescent cells acts in an autocrine manner to reinforce the senescence growth arrest. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **44.Krtolica A, Parrinello S, Lockett S, Desprez P, Campisi J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proc Natl Acad Sci USA. 2001;98:12072–12077. doi: 10.1073/pnas.211053698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **45.Liu D, Hornsby PJ. Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Res. 2007;67:3117–3126. doi: 10.1158/0008-5472.CAN-06-3452. [DOI] [PubMed] [Google Scholar]
  • **46.Bartholomew JN, Volonte D, Galbiati F. Caveolin-1 regulates the antagonistic pleiotropic properties of cellular senescence through a novel Mdm2/p53-mediated pathway. Cancer Res. 2009;69:2878–2886. doi: 10.1158/0008-5472.CAN-08-2857. These papers provide evidence that, at least in mouse xenograft models, senescent cells can promote the progression of premalignant cells to malignancy, and stimulate the growth of tumors initiated by frankly malignant cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **47.Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, Yee H, Zender L, Lowe SW. Senescence of activated stellate cells limits liver fibrosis. Cell. 2008;134:657–667. doi: 10.1016/j.cell.2008.06.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **48.Jun JI, Lau LF. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nature Cell Biol. 2010;12:676–685. doi: 10.1038/ncb2070. These papers provide evidence that senescent cells are important for limiting the extent of fibrosis after tissue damage. Both papers show that the MMPs secreted by senescent cells are especially important allowing proper tissue repair. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Rauser CL, Mueller LD, Rose MR. The evolution of late life. Ageing Res Rev. 2006;5:14–32. doi: 10.1016/j.arr.2005.06.003. [DOI] [PubMed] [Google Scholar]

RESOURCES