Review
Cellular senescence and organismal aging

https://doi.org/10.1016/j.mad.2008.04.001Get rights and content

Abstract

Cellular senescence, first observed and defined using in vitro cell culture studies, is an irreversible cell cycle arrest which can be triggered by a variety of factors. Emerging evidence suggests that cellular senescence acts as an in vivo tumor suppression mechanism by limiting aberrant proliferation. It has also been postulated that cellular senescence can occur independently of cancer and contribute to the physiological processes of normal organismal aging. Recent data have demonstrated the in vivo accumulation of senescent cells with advancing age. Some characteristics of senescent cells, such as the ability to modify their extracellular environment, could play a role in aging and age-related pathology. In this review, we examine current evidence that links cellular senescence and organismal aging.

Introduction

Cellular senescence was first described by Hayflick and Moorfield in 1961 who observed that cultures of normal human fibroblasts had a limited replicative potential and eventually became irreversibly arrested (Hayflick and Moorhead, 1961, Campisi and d’Adda di Fagagna, 2007, Sedivy et al., 2007). The majority of senescent cells assume a characteristic flattened and enlarged morphology, and over the years a large number of molecular phenotypes have been described, such as changes in gene expression, protein processing and chromatin organization (Gonos et al., 1998, Shelton et al., 1999, Schwarze et al., 2002, Semov et al., 2002, Narita et al., 2003, Zhang et al., 2003, Zhang et al., 2005, Zhang et al., 2007, Yoon et al., 2004, Pascal et al., 2005, Xie et al., 2005, Cong et al., 2006, Funayama et al., 2006, Trougakos et al., 2006, Zdanov et al., 2006). The growth arrest occurs mostly in G1 phase (Pignolo et al., 1998). Although individual cells arrest rapidly, probably within the duration of a single cell cycle, cultures are typically quite asynchronous with increasing proportions of cells withdrawing into senescence over a period of several weeks (Herbig et al., 2003, Herbig et al., 2004). Senescent cells maintain metabolic activity and can remain viable essentially indefinitely (Matsumura et al., 1979, Pignolo et al., 1994). An important component of this stability in culture may be the capacity of senescent cells to resist apoptosis (Marcotte et al., 2004, Hampel et al., 2005).

Conceptually, there are two broad categories of replicative cellular senescence. The first is initiated by dysfunctional telomeres or other forms of genotoxic stress eliciting a DNA damage response mediated primarily by the p53 tumor suppressor pathway (d’Adda di Fagagna et al., 2003, Herbig et al., 2004). The second, much less understood response does not involve telomeres or DNA damage, and is characterized by the upregulation of the CDKN2A gene (cyclin-dependent kinase inhibitor p16INK4a). These basic distinctions are however complicated by the fact that p16 can be upregulated by a wide variety of stresses, including some forms of genotoxic damage.

The relationship between elapsed cell divisions and the onset of senescence was clearly apparent from early in vitro studies, and led to proposals that senescence may contribute to in vivo organismal aging phenotypes (Hayflick, 1985). This view was reinforced by findings that cells explanted from old donors were capable of fewer in vitro population doublings than those from young individuals (Martin et al., 1970, Le Guilly et al., 1973, Rheinwald and Green, 1975, Schneider and Mitsui, 1976, Bierman, 1978, Bruce et al., 1986). The gradual attrition of telomeres subsequently provided the molecular mechanism for the cell division clock (Harley et al., 1990, Bodnar et al., 1998). It has been proposed that the term “cellular senescence” be reserved for those phenomena based on an inherent counting mechanism, with other terms, such as “stasis” to be applied to the large variety of stress-induced arrests (Drayton and Peters, 2002, Wright and Shay, 2002). Recent usage has however favored irreversible cell cycle arrest as the defining feature, and “cellular senescence” is now commonly used to encompass states induced by stress and signaling imbalances (Collado et al., 2007).

It is important to note that the intrinsic cell division clock can be significantly affected by extrinsic influences, such as reactive oxygen species, which accelerate the rate of telomere shortening (von Zglinicki, 2002). Although senescent cells display a number of phenotypes that discriminate them from quiescent cells, it has also been suggested that senescence could be considered a form of terminal differentiation (Bayreuther et al., 1988, Seshadri and Campisi, 1990). This view has recently been given new life by observations that downregulation of Wnt signaling may be a factor in triggering the onset of senescence (Ye et al., 2007). Thus, in addition to being influenced by the environment, senescence may also respond to developmental or endocrine cues.

Given that senescence results in the arrest of proliferation, its potential for opposing cancer development was pointed out some time ago (Sager, 1991). This notion was strongly reinforced by the discovery that activation of oncogenes in normal cells could trigger senescence (Serrano et al., 1997). Recent data have indeed implicated cellular senescence as an important in vivo tumor suppressor mechanism in a variety of human and mouse tissues (Braig et al., 2005, Chen et al., 2005, Collado et al., 2005, Michaloglou et al., 2005, Courtois-Cox et al., 2006, Cosme-Blanco et al., 2007, Feldser and Greider, 2007, Ventura et al., 2007, Xue et al., 2007). In addition to being on sound experimental footing, the tumor suppressive function of senescence also provides a rational explanation for its evolution. The possible role of senescence in age-associated dysfunction is often justified by invoking the concept of antagonistic pleiotropy (Williams, 1957, Rose, 1991, Kirkwood and Austad, 2000, Campisi, 2005), namely, that beneficial traits (such as cancer suppression) under selection in reproductively active individuals may have unselected and unintended effects in more advanced age.

In contrast to tumor suppression, the connections between cellular senescence and the aging of organisms are significantly more tenuous. In this review we focus on these links and their implications. Elucidating these relationships is expected to advance our comprehension of the mechanisms involved in age-related diseases as well as normal aging processes.

Section snippets

Telomeres

Telomeres shorten with each round of genome duplication, an unavoidable consequence of the RNA priming mechanism of DNA replication (Olovnikov, 1973). The minimum rate of telomere shortening in human cells (30–50 bp per cell division) is slightly in excess of that predicted by the “end-replication problem”, and probably stems from further exonucleolytic processing of chromosome ends (Sfeir et al., 2005). Telomere attrition can be accelerated by a number of factors, such as oxidative damage (von

Accumulation of senescent cells in vivo

To the extent that senescent cells are considered to confer deleterious effects, including the promotion of organismal aging, it is crucial to distinguish them from the majority of healthy but quiescent cells found in normal tissues. The original definition of irreversible arrest is clearly not feasible in this context, a situation that has led to a concerted search for biomarkers to assess the increasingly numerous molecular phenotypes of senescent cells. Unfortunately, to date no biomarker

Senescent cells at sites of age-related pathology

The presence of senescence-associated markers at sites of age-related pathologies have provided further links between cellular senescence and aging. Telomere length as a function of donor age was found to decrease more rapidly in arterial than in venous endothelial cells, and telomere loss was greater in intimal than in medial cells (Chang and Harley, 1995). Other studies found that age-dependent telomere attrition is faster in the distal than in the proximal segment of the abdominal aorta,

CDKN2A (p16) and aging

The cyclin-dependent kinase inhibitor p16 has emerged as an important player in aging and age-related disease. Biochemically, p16 inactivates CDK4 and CDK6, which maintains pRB in its active, hypophosphorylated form and consequently blocks cell cycle progression in G1 (Sherr and Roberts, 1999). Genetically, p16 is an important and potent tumor suppressor and is frequently inactivated in several human cancers, such as melanoma. Because p16 expression can be upregulated by a wide variety of

Physiological relevance of senescent cells in vivo

Senescent cells are generated in response to a number of stimuli. Telomere attrition and oncogene activation are the best understood triggers, but a variety of stresses, disease or pathological conditions, and environmental and nutritional factors are also likely to play important roles. Although it is now possible to detect and even quantify senescent cells in vivo with some confidence, the physiological consequences of the existence of such cells are only beginning to be unravelled. Broadly

Perspectives

The role of cellular senescence in a variety of age-associated pathologies is becoming increasingly accepted. As discussed above, compelling experimental evidence has now linked increased rates of cellular senescence with accelerated aging. The extent to which, if any, cellular senescence contributes to the natural life span of any one species however remains to be established. As a case in point, although quantitative estimates of in vivo cellular senescence are just beginning to emerge, the

Acknowledgement

J.C.J. and J.M.S. were supported, in part, by grant R01 AG016694 from the NIH and a Senior Scholar Award in Aging from the Ellison Medical Fundation, both to J.M.S.

References (181)

  • S. Courtois-Cox et al.

    A negative feedback signaling network underlies oncogene-induced senescence

    Cancer Cell

    (2006)
  • V.J. Cristofalo

    SA beta Gal staining: biomarker or delusion

    Exp. Gerontol.

    (2005)
  • T.K. Dilley et al.

    Novel mechanisms of sublethal oxidant toxicity: induction of premature senescence in human fibroblasts confers tumor promoter activity

    Exp. Cell Res.

    (2003)
  • S. Drayton et al.

    Immortalisation and transformation revisited

    Curr. Opin. Genet. Dev.

    (2002)
  • D.M. Feldser et al.

    Short telomeres limit tumor progression in vivo by inducing senescence

    Cancer Cell

    (2007)
  • D. Giri et al.

    Interleukin-1alpha is a paracrine inducer of FGF7, a key epithelial growth factor in benign prostatic hyperplasia

    Am. J. Pathol.

    (2000)
  • E.S. Gonos et al.

    Cloning and identification of genes that associate with mammalian replicative senescence

    Exp. Cell Res.

    (1998)
  • L. Hayflick

    Theories of biological aging

    Exp. Gerontol.

    (1985)
  • L. Hayflick et al.

    The serial cultivation of human diploid cell strains

    Exp. Cell Res.

    (1961)
  • M.T. Hemann et al.

    The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability

    Cell

    (2001)
  • U. Herbig et al.

    Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a)

    Mol. Cell

    (2004)
  • U. Herbig et al.

    Regulation of growth arrest in senescence: telomere damage is not the end of the story

    Mech. Ageing Dev.

    (2006)
  • W. Hornebeck

    Down-regulation of tissue inhibitor of matrix metalloprotease-1 (TIMP-1) in aged human skin contributes to matrix degradation and impaired cell growth and survival

    Pathol. Biol.

    (2003)
  • S. Huang et al.

    Accelerated telomere shortening and replicative senescence in human fibroblasts overexpressing mutant and wild-type lamin A

    Exp. Cell Res.

    (2008)
  • G. Jenkins

    Molecular mechanisms of skin ageing

    Mech. Ageing Dev.

    (2002)
  • J.C. Jeyapalan et al.

    Accumulation of senescent cells in mitotic tissue of aging primates

    Mech. Ageing Dev.

    (2007)
  • S.A. Joosten et al.

    Telomere shortening and cellular senescence in a model of chronic renal allograft rejection

    Am. J. Pathol.

    (2003)
  • J. Kajstura et al.

    Telomere shortening is an in vivo marker of myocyte replication and aging

    Am. J. Pathol.

    (2000)
  • W.Y. Kim et al.

    The regulation of INK4/ARF in cancer and aging

    Cell

    (2006)
  • A. Krtolica et al.

    Cancer and aging: a model for the cancer promoting effects of the aging stroma

    Int. J. Biochem. Cell Biol.

    (2002)
  • K. Ksiazek et al.

    Premature senescence of mesothelial cells is associated with non-telomeric DNA damage

    Biochem. Biophys. Res. Commun.

    (2007)
  • J.L. Lewis et al.

    The influence of INK4 proteins on growth and self-renewal kinetics of hematopoietic progenitor cells

    Blood

    (2001)
  • J. Adams et al.

    No association between socio-economic status and white blood cell telomere length

    Aging Cell

    (2007)
  • R.C. Allsopp et al.

    Effect of TERT over-expression on the long-term transplantation capacity of hematopoietic stem cells

    Nat. Med.

    (2003)
  • M.Y. Armanios et al.

    Telomerase mutations in families with idiopathic pulmonary fibrosis

    N. Engl. J. Med.

    (2007)
  • A. Aviv et al.

    Human telomere biology: pitfalls of moving from the laboratory to epidemiology

    Int. J. Epidemiol.

    (2006)
  • G.M. Baerlocher et al.

    Flow cytometry and FISH to measure the average length of telomeres (flow FISH)

    Nat. Protoc.

    (2006)
  • C. Bavik et al.

    The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms

    Cancer Res.

    (2006)
  • K. Bayreuther et al.

    Differentiation of fibroblast stem cells

    J. Cell Sci. Suppl.

    (1988)
  • E.L. Bierman

    The effect of donor age on the in vitro life span of cultured human arterial smooth-muscle cells

    In Vitro

    (1978)
  • M.A. Blasco

    Telomere length, stem cells and aging

    Nat. Chem. Biol.

    (2007)
  • A.G. Bodnar et al.

    Extension of life-span by introduction of telomerase into normal human cells

    Science

    (1998)
  • P. Boukamp

    Skin aging: a role for telomerase and telomere dynamics?

    Curr. Mol. Med.

    (2005)
  • A.P. Bracken et al.

    The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells

    Genes Dev.

    (2007)
  • M. Braig et al.

    Oncogene-induced senescence as an initial barrier in lymphoma development

    Nature

    (2005)
  • W.T. Brown

    Genetic diseases of premature aging as models of senescence

    Annu. Rev. Gerontol. Geriatr.

    (1990)
  • J. Campisi et al.

    Cellular senescence: when bad things happen to good cells

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • A. Canela et al.

    High-throughput telomere length quantification by FISH and its application to human population studies

    Proc. Natl. Acad. Sci. U.S.A.

    (2007)
  • P. Castro et al.

    Cellular senescence in the pathogenesis of benign prostatic hyperplasia

    Prostate

    (2003)
  • P. Castro et al.

    Interleukin-8 expression is increased in senescent prostatic epithelial cells and promotes the development of benign prostatic hyperplasia

    Prostate

    (2004)
  • Cited by (288)

    • Comprehensive review on Schisandra chinesis

      2024, Pharmacological Research - Modern Chinese Medicine
    View all citing articles on Scopus
    View full text