Cellular senescence in vivo: From cells to tissues to pathologies

https://doi.org/10.1016/j.mad.2020.111308Get rights and content

Highlights

  • Recent data in animal models indicate that cellular senescence plays important roles in the pathogenesis of a range of aging-associated disorders.

  • While senescence is often studied in cell culture, less information is available regarding senescence and its associated states in vivo.

  • Here, we review data available to indicate which cell types can undergo senescence within various mammalian tissue contexts in vivo.

  • The data serve as a foundation for future work using tissue-specific designs to further characterize these cells and their role in aging and disease.

Abstract

Senescent cells accumulate during aging in a variety of tissues. Although scarce, they could influence tissue function non-cell-autonomously via secretion of a range of factors in their neighborhood. Recent studies support a role of senescent cells in age-related morbidity, including neurodegenerative diseases, cardiovascular pathologies, cancers, aging-associated nephrological alterations, chronic pulmonary disease and osteoarthritis, indicating that senescent cells could represent an interesting target for therapeutic exploitation across a range of pathophysiological contexts. In this article, we review data available to indicate which cell types can undergo senescence within various mammalian tissue environments and how these processes may contribute to tissue-specific pathologies associated with old age. We also consider markers used to identify senescent cells in vitro and in vivo. The data discussed may serve as an important starting point for an extended definition of molecular and functional characteristics of senescent cells in different organs and may hence promote the development and refinement of targeting strategies aimed at removing senescent cells from aging tissues.

Introduction

Cellular senescence is a form of stable cell cycle arrest that occurs in diploid cells when they are exposed to certain stressors, including genotoxic agents, nutrient deprivation, hypoxia, mitochondrial dysfunction and oncogene activation. Senescence was first described in primary cells by Hayflick and colleagues (Hayflick and Moorhead, 1961), when they observed that human diploid fibroblasts in vitro could reach only a finite number of cell divisions before their proliferation was arrested irreversibly. This biological phenomenon, referred to as the “Hayflick limit”, was later attributed to progressive telomere shortening upon consecutive rounds of cell divisions (Courtois-Cox et al., 2008). The Hayflick limit represents a physiological cellular response to counter genomic instability (Courtois-Cox et al., 2008; Hayflick and Moorhead, 1961). This phenomenon is now also referred to as replicative senescence. However, an accelerated senescence response, which is independent of telomere shortening, can also arrest growth or proliferation of diploid cells, and it is known as premature senescence (Courtois-Cox et al., 2008; d’Adda di Fagagna et al., 2003; Serrano et al., 1997). Subsequently, it was demonstrated that oncogenic stress (e.g., overexpression of oncogenes and loss of tumor suppressor genes) can induce premature senescence in vitro, a process known as ‘oncogene-induced senescence’ (Counter et al., 1992; Serrano et al., 1997).

Many questions regarding the causes, signaling networks and mechanisms underlying the various types of cellular senescence still remain open, and current evidence is largely based on in vitro experiments. At the molecular level, cell cycle arrest and exit is controlled by the activation of the p53/p21CIP1 and the p16INK4a/Rb tumor suppressor pathways (Herranz and Gil, 2018). The senescence growth arrest is often triggered by a persistent DNA damage response (DDR) caused by either intrinsic (e.g. oxidative damage, telomere attrition, hyperproliferation) or extrinsic insults (e.g. ultraviolet- or γ-irradiation, cytostatic drugs) (Herranz and Gil, 2018). In the context of the DDR signaling pathway, ATM or ATR kinases block cell-cycle progression through stabilization of p53 and transcriptional activation of the cyclin-dependent kinase (Cdk) inhibitor p21 (van Deursen, 2014). Moreover, activated oncogenes, such as oncogenic Ras, are also prominent inducers of senescence. Oncogenic Ras acts through overexpression of Cdc6 and suppression of nucleotide metabolism, resulting in aberrant DNA replication, formation of double-strand DNA breaks (DSBs) and activation of the DDR pathway (Aird et al., 2013; Di Micco et al., 2006). However, senescence caused by E2F3 activation or c-Myc inhibition is DDR-independent and involves p19Arf and p16Ink4a (Lazzerini Denchi et al., 2005; Nardella et al., 2011). Senescence is closely linked to profound metabolic changes (Dörr et al., 2013; Kondoh et al., 2005). Furthermore, various tumor suppressors trigger a senescent growth arrest when inactivated, including RB, PTEN, NF1 and VHL (Nardella et al., 2011; Shamma et al., 2009). While RB inactivation involves the DDR (Shamma et al., 2009), the other tumor suppressors mentioned are DDR-independent and induce senescence through p19Arf and p16Ink4a. An interesting species-specific difference is that senescence pathways in murine cells are more dependent on p19Arf than those in human cells (Ben-Porath and Weinberg, 2005). Recently, the cGAS–cGAMP–STING pathway, in the context of which cGAS senses cytoplasmic DNA as a consequence of nuclear DNA damage, has been shown to connect DNA damage to inflammation, senescence and cancer via activation of the transcription factors IRF3 and NF-κB (Li and Chen, 2018; Yang et al., 2017). In summary, many senescence-inducing stressors or stimuli have in common that they trigger a DDR which then orchestrates downstream signaling mediated by ATM/R kinases or the cGAS–STING axis to acquire a full senescence phenotype. On the other hand, some of the stimuli induce cellular senescence through p19Arf, p16Ink4a and profound metabolic changes suggesting that type, intensity and/or exposure time to senescence-inducing stimuli can play a major role in determining which senescence pathway gets triggered. However, it is important to note that current models regarding the molecular mechanisms involved in cellular senescence are largely based on in vitro data. To what extent each of these pathways is involved in driving cellular senescence within different tissues and cell types in vivo is an important issue that remains to be explored in future studies.

A primary feature of senescence cells, which distinguishes them from the state of quiescence, is irreversible proliferative withdrawal. In vitro, senescent cells often exhibit increased size with a flattened morphology, smooth shape, large vacuoles and possibly multiple nuclei. However, these changes in size and shape of senescent cells may not necessarily occur in the same manner in tissues in vivo (Rhinn et al., 2019). An additional feature of senescent cells is that they fail to respond to growth- and apoptosis-inducing stimuli (Baar et al., 2017; Yosef et al., 2016; Zeng et al., 2018). A typical senescence phenotype is characterized by a number of intrinsic and extrinsic markers, although none of them is specific or universal for senescence. At the molecular level, senescent cells are routinely characterized by upregulation of cell-cycle inhibitors such as p21 and/or p16 (Caliò et al., 2015; He et al., 2017), positive staining of senescence-associated β-galactosidase (SA-β-gal) at pH 6 (Dimri et al., 1995; He et al., 2017; Hernandez-Segura et al., 2018), formation of senescence-associated heterochromatin foci (SAHF) (Yamauchi et al., 2017), accumulation of lipofuscin, loss of lamin B1 (Shimi et al., 2011), senescence-associated distension of satellites (Swanson et al., 2013), the induction of senescence-associated DNA damage (Kim et al., 2017) and the secretion of a large number of factors, including growth factors, cytokines, chemokines, and proteases, which is commonly known as the senescence-associated secretory phenotype (SASP) (Coppe et al., 2010; Lopes-Paciencia et al., 2019). Moreover, enhanced expression of p19ARF, p53 and PAI-1 are also observed in senescent cells and are used as senescence markers (Bernardes de Jesus and Blasco, 2012; Hernandez-Segura et al., 2018). A recent study suggested that c-Met could serve as an early marker of cellular senescence; however, the observations are based on in vitro investigation (Boichuck et al., 2019). Recently, Gal et al. demonstrated the use of the ImageStreamX approach as a powerful method for the detection and quantification of senescent cells in distinct tissues and cell populations (Gal et al., 2019). This novel method combines several senescence-related markers, together with the commonly used senescence-associated β-galactosidase assay and thereby offers a new solution to quantify senescent cells in vivo. For a summary of commonly used markers of cellular senescence, see Table 1.

Cellular senescence is induced under physiological or pathological conditions by a variety of extrinsic or intrinsic cellular signals (Avelar et al., 2020b; He and Sharpless, 2017; Hernandez-Segura et al., 2018; Yanai and Fraifeld, 2018; Zeng et al., 2018). The persistence of senescent cells in tissues may have beneficial function as well as may alter tissue remodeling and homeostasis: transient accumulation of senescent cells in tissues is mainly associated with beneficial functions (Calcinotto et al., 2019), whereas long-term accumulation of senescent cells appears to deteriorate tissue homeostasis. Recently, transgenic mouse models have been developed to permit the elimination of senescent cells in vivo in mice (including the INK/ATTAC model, the p16−3MR model and the ARF-DTR model (Baker et al., 2011; Demaria et al., 2014; Hashimoto et al., 2016), and a series of individual studies using these mouse models have now established clear, causal contributions of senescent cells to lifespan, wound healing, tissue development and programming and to age-related functional decline in specific organ systems (Avelar et al., 2020a; Baker et al., 2016, 2011; Childs et al., 2015; Docherty et al., 2019; Farr and Khosla, 2019; Hashimoto et al., 2016; Tacutu et al., 2011). In additional studies, senolytics, drugs that specifically target senescent cells by inducing apoptosis of senescent cells, were also used for the clearance of senescent cells in mice and humans as an alternative approach to the INK/ATTAC model, and interestingly, comparable therapeutic benefits have been reported (Hickson et al., 2019; Short et al., 2019; Zhang et al., 2019). The accumulation of senescent cells with advancing age in various tissues and their role in age-related tissue alterations and pathologies, such as osteoarthritis (Price et al., 2002) and atherosclerosis (Campisi, 2005), has been documented by a number of studies (Calcinotto et al., 2019; He and Sharpless, 2017; Rhinn et al., 2019). It has also been reported that senescent cells accumulate in various tumors in vivo, where they are thought to influence tumor progression (Campisi, 2005; Lee and Schmitt, 2019; Zeng et al., 2018). Therefore, therapeutic approaches which can selectively clear senescent cells may be useful for the treatment or prevention of a broad range of age-related disorders.

Available evidence indicates that only specific cell types within a given tissue are susceptible to acquiring a senescent phenotype, whereas other cell types remain unaffected by senescence. To further study the cells that are being targeted or should be targeted by senolytic approaches, knowledge of their identities is required. Here, we review currently available evidence linking specific cell types in various tissues to the potential for acquiring a state of senescence in vivo (see main text below and Table 2). We have focused this review on tissue types where cellular senescence has been suggested to play a role in functional alterations of a given tissue in vivo. We have not included tissues where no clear evidence for in vivo senescence, based on multiple methods, is available. We also required the availability of data on cell type identity of senescent cells and demonstrated links to tissue function. The data available may serve as an important starting point for the further definition of molecular and functional characteristics of senescent cells in different organs and may hence promote the development and refinement of targeting strategies aimed at removing senescent cells from aging tissues.

Section snippets

Tissue repair and wound healing

Senescent cells have been demonstrated to play a role in wound repair and the fibrotic response in a number of tissues, including the liver (Krizhanovsky et al., 2008), skin (Jun and Lau, 2010), lung (Schafer et al., 2017) and heart (Zhu et al., 2013). In a preclinical mouse model of liver damage and fibrosis, senescent hepatic stellate cells were identified in the fibrotic lesions in liver (Krizhanovsky et al., 2008). In this study, induction of damage in mice deficient for both the p53 and p16

Concluding remarks

There is plenty of evidence suggesting that a variety of cellular stressors and various insults are key contributors to in vivo cellular senescence, which in turn is thought to contribute to age-related tissue dysfunctions and pathologies. Even a relatively small percentage of senescent cells in organs may impair tissue homeostasis and regeneration, decrease organ function, and contribute to aging phenotypes, as demonstrated by those studies where senescent cells were either genetically or

Acknowledgements

This work was supported by a grant from the Helmholtz Future Topic AMPro (Aging and Metabolic Programming).

References (206)

  • S.J. Chinta et al.

    Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to parkinson’s disease

    Cell Rep.

    (2018)
  • A.B. Chkhotua et al.

    Increased expression of p16(INK4a) and p27(Kip1) cyclin-dependent kinase inhibitor genes in aging human kidney and chronic allograft nephropathy

    Am. J. Kidney Dis.

    (2003)
  • M. Demaria et al.

    An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA

    Dev. Cell

    (2014)
  • A. Denic et al.

    Structural and functional changes with the aging kidney

    Adv. Chronic Kidney Dis.

    (2016)
  • G. Ding et al.

    Tubular cell senescence and expression of TGF-beta1 and p21(WAF1/CIP1) in tubulointerstitial fibrosis of aging rats

    Exp. Mol. Pathol.

    (2001)
  • R.B. Effros

    From Hayflick to Walford: the role of T cell replicative senescence in human aging

    Exp. Gerontol.

    (2004)
  • R.B. Effros

    Role of T lymphocyte replicative senescence in vaccine efficacy

    Vaccine

    (2007)
  • J.N. Farr et al.

    Cellular senescence in bone

    Bone

    (2019)
  • K. Ghosh et al.

    The senescence-associated secretory phenotype: critical effector in skin Cancer and aging

    J. Invest. Dermatol.

    (2016)
  • L. Hayflick et al.

    The serial cultivation of human diploid cell strains

    Exp. Cell Res.

    (1961)
  • S. He et al.

    Senescence in health and disease

    Cell

    (2017)
  • L. He et al.

    Cellular senescence regulated by SWI/SNF complex subunits through p53/p21 and p16/pRB pathway

    Int. J. Biochem. Cell Biol.

    (2017)
  • A. Hernandez-Segura et al.

    Hallmarks of cellular senescence

    Trends Cell Biol.

    (2018)
  • L.J. Hickson et al.

    Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease

    EBioMedicine

    (2019)
  • R. Anderson et al.

    Length-independent telomere damage drives post-mitotic cardiomyocyte senescence

    EMBO J.

    (2019)
  • R.A. Avelar et al.

    A multidimensional systems biology analysis of cellular senescence in aging and disease

    Genome Biol.

    (2020)
  • R.A. Avelar et al.

    A multidimensional systems biology analysis of cellular senescence in aging and disease

    Genome Biol.

    (2020)
  • D. Aw et al.

    Phenotypical and morphological changes in the thymic microenvironment from ageing mice

    Biogerontology

    (2009)
  • M.P. Baar et al.

    Targeted apoptosis of senescent cells restores tissue homeostasis in response to Chemotoxicity and aging

    Cell

    (2017)
  • A.D. Bachstetter et al.

    Microglial p38alpha MAPK is a key regulator of proinflammatory cytokine up-regulation induced by toll-like receptor (TLR) ligands or beta-amyloid (Abeta)

    J. Neuroinflammation

    (2011)
  • D.J. Baker et al.

    Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders

    Nature

    (2011)
  • D.J. Baker et al.

    Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan

    Nature

    (2016)
  • A. Banito et al.

    Senescence impairs successful reprogramming to pluripotent stem cells

    Genes Dev.

    (2009)
  • P.J. Barnes

    Senescence in COPD and its comorbidities

    Annu. Rev. Physiol.

    (2017)
  • B. Berkenkamp et al.

    In vivo and in vitro analysis of age-associated changes and somatic cellular senescence in renal epithelial cells

    PLoS One

    (2014)
  • A. Bernadotte et al.

    Markers of cellular senescence. Telomere shortening as a marker of cellular senescence

    Aging (Albany NY)

    (2016)
  • B. Bernardes de Jesus et al.

    Assessing cell and organ senescence biomarkers

    Circ. Res.

    (2012)
  • R. Bhat et al.

    Astrocyte senescence as a component of Alzheimer’s disease

    PLoS One

    (2012)
  • T.G. Bird et al.

    TGFbeta inhibition restores a regenerative response in acute liver injury by suppressing paracrine senescence

    Sci. Transl. Med.

    (2018)
  • B.G. Bitler et al.

    Wnt5a suppresses epithelial ovarian cancer by promoting cellular senescence

    Cancer Res.

    (2011)
  • M. Boichuck et al.

    c-Met as a new marker of cellular senescence

    Aging (Albany NY)

    (2019)
  • T.J. Bussian et al.

    Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline

    Nature

    (2018)
  • J. Cahu et al.

    A sensitive method to quantify senescent cancer cells

    J. Vis. Exp.

    (2013)
  • A. Calcinotto et al.

    Cellular senescence: aging, Cancer, and injury

    Physiol. Rev.

    (2019)
  • A. Caliò et al.

    Cellular senescence markers p16INK4a and p21CIP1/WAF are predictors of hodgkin lymphoma outcome

    Clin. Cancer Res.: An Off. J. Am. Assoc. Cancer Res.

    (2015)
  • A. Carnero

    Markers of cellular senescence

    Methods Mol. Biol.

    (2013)
  • X. Chen et al.

    Epithelial cell senescence induces pulmonary fibrosis through Nanog-mediated fibroblast activation

    Aging (Albany NY)

    (2019)
  • A. Chiche et al.

    Injury-induced senescence enables in vivo reprogramming in skeletal muscle

    Cell Stem Cell

    (2017)
  • B.G. Childs et al.

    Cellular senescence in aging and age-related disease: from mechanisms to therapy

    Nat. Med.

    (2015)
  • B.G. Childs et al.

    Senescent intimal foam cells are deleterious at all stages of atherosclerosis

    Science

    (2016)
  • Cited by (7)

    • Ageing-associated phenotypes in mice

      2023, Mechanisms of Ageing and Development
    • Pathological implication of CaMKII in NF-κB pathway and SASP during cardiomyocytes senescence

      2023, Mechanisms of Ageing and Development
      Citation Excerpt :

      Hence, CaMKII may be involved in heart ageing-associated NF-κB activation and inflammation. Sustained accumulation of senescent cells is a classical hallmark of ageing organs and leads to age-related pathology and function deterioration (Kumar et al., 2020). Accumulated senescent cells exhibit the senescence-associated secretory phenotype (SASP), which is predominantly induced by the NF-κB activation and is characterized by the overproduction of cytokines, chemokines and extracellular metalloproteinase (ECM) (Birch and Gil, 2020; Di Micco et al., 2021).

    View all citing articles on Scopus
    View full text