26
Epigenetic control of cellular senescence in disease: opportunities for therapeutic intervention Stuart P. Atkinson and W. Nicol Keith* Understanding how senescence is established and maintained is an important area of st ud y both for n or mal cel l ph ysi ol og y and in t um ouri ge ne si s. Modi cati ons to N-terminal tails of hi st one proteins, wh ich can lead to chromatin remodelling, appear to be key to the regulation of the senescence phenotype. Epigenetic mechanisms such as modication of histone proteins ha ve been shown to be suf ci ent to re gula te gene expr essi on levels and specic gene promoters can become epigenetically alt er ed at senescence. This sugge sts that epigenet ic mechanisms are impor tant in senescence and further suggests epigenetic deregulation could play an important role in the bypass of senescence and the acquisition of a tumourigenic phenotype. Tumour suppressor proteins and cellular senescence are intimately linked and such proteins are now known to regulate gene expression through chromatin remodelling, again sugges ting a link bet ween chr omatin modi ca tion and cellular senescence. Telomere dynamics and the expression of the telomerase genes are also both implicitly linked to senescence and tumourigenesis, and epigenetic deregulation of the telomerase gene promoters has been identied as a possible mech an ism for the ac ti va ti on of telomere main tenance mecha nisms in canc er. Recent studie s have also sugges ted that epigenetic deregulation in stem cells could play an important role in carcinogenesis, and new models have been suggested for the attainment of tumourigenesis and byp ass of senescence. Over al l, pr ope r re gulatio n of the chroma tin environment is suggested to ha ve an important rol e in the se nescence pathway, such that its deregulation could lead to tumourigenesis. Centre for Oncology and Applied Pharmacology, University of Glasgow, Cancer Research UK Beatson Laboratories, Bearsden, Glasgow, G61 1BD, UK. *Corr esp ond ing aut hor : W. Nic ol Kei th, Ce ntre for Onc olo gy and App lie d Pha rma col ogy, University of Glasgow, Cancer Research UK Beatson Laboratories, Alexander Stone Building, Garsc ube Esta te, Switchbac k Rd, Bearsden , Glasg ow , G61 1BD, UK. T el: +44 (0)141 330 481 1; Fax: +44 (0)141 330 4127; E-mail: n.keith@be atson.gla.ac.uk expert reviews http: //www. expertreviews.org/ i n mol ecul ar medici ne 1  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007 & 2007 Cambridge University Press       E      p       i      g      e      n      e       t       i      c      c      o      n       t      r      o       l      o       f      c      e       l       l      u       l      a      r      s      e      n      e      s      c      e      n      c      e       i      n       d       i      s      e      a      s      e      :      o      p      p      o      r       t      u      n       i       t       i      e      s       f      o      r       t       h      e      r      a      p      e      u       t       i      c       i      n       t      e      r      v      e      n       t       i      o      n

Senescence in Disease

Embed Size (px)

Citation preview

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 1/26

Epigenetic control of cellular

senescence in disease: opportunities

for therapeutic intervention

Stuart P. Atkinson and W. Nicol Keith*

Understanding how senescence is established and maintained is an important

area of study both for normal cell physiology and in tumourigenesis.

Modifications to N-terminal tails of histone proteins, which can lead to

chromatin remodelling, appear to be key to the regulation of the senescence

phenotype. Epigenetic mechanisms such as modification of histone proteinshave been shown to be sufficient to regulate gene expression levels and

specific gene promoters can become epigenetically altered at senescence.

This suggests that epigenetic mechanisms are important in senescence

and further suggests epigenetic deregulation could play an important role in

the bypass of senescence and the acquisition of a tumourigenic phenotype.

Tumour suppressor proteins and cellular senescence are intimately linked and

such proteins are now known to regulate gene expression through chromatin

remodelling, again suggesting a link between chromatin modification and

cellular senescence. Telomere dynamics and the expression of the telomerase

genes are also both implicitly linked to senescence and tumourigenesis, and

epigenetic deregulation of the telomerase gene promoters has been identified

as a possible mechanism for the activation of telomere maintenance

mechanisms in cancer. Recent studies have also suggested that epigenetic

deregulation in stem cells could play an important role in carcinogenesis, and

new models have been suggested for the attainment of tumourigenesis

and bypass of senescence. Overall, proper regulation of the chromatin

environment is suggested to have an important role in the senescence

pathway, such that its deregulation could lead to tumourigenesis.

Centre for Oncology and Applied Pharmacology, University of Glasgow, Cancer Research UKBeatson Laboratories, Bearsden, Glasgow, G61 1BD, UK.

*Corresponding author: W. Nicol Keith, Centre for Oncology and Applied Pharmacology,University of Glasgow, Cancer Research UK Beatson Laboratories, Alexander Stone Building,Garscube Estate, Switchback Rd, Bearsden, Glasgow, G61 1BD, UK. Tel: +44 (0)141 330 4811;Fax: +44 (0)141 330 4127; E-mail: [email protected]

expert reviewshttp://www.expertreviews.org/ in molecular medicine

1  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 2/26

Cellular senescence provides a barrier toexcessive cellular growth and is triggered bythree main mechanisms. Telomere attritionduring extended cell proliferation leadseventually to cellular senescence (Ref. 1) when

critically short telomeres are recognised asDNA damage, thereby triggering thesenescence arrest. Although this proliferative

  barrier provides a limit to the outgrowth of acell, senescence can also be triggered before thislimit in response to cellular damage (such asDNA damage) or stress and oncogenic signalling.As senescent cells do not proliferate, it has beenproposed that cellular senescence is a major

  barrier to cancerous transformation (Refs 2, 3)and premature induction of cellular senescencecan therefore be targeted for anti-cancer therapyin tumourigenic cells (Refs 4, 5) (Fig. 1).

The senescence phenotype is associated withmultiple cellular changes including alteredcellular morphology, size and, importantly,expression of numerous genes involved inmany aspects of cell physiology (Refs 6, 7, 8, 9,10). The altered regulation of genes and thecomplex phenotype suggest that epigeneticmechanisms may be involved. Indeed, it is nowapparent that, in addition to multiple geneticpathways, epigenetics plays a major contributingrole in senescence (Refs 11, 12) as well as in the

 bypass of senescence leading to tumourigenicity(Refs 13, 14, 15). Therefore, to understand

senescence and tumourigenesis, both geneticand epigenetic mechanisms regulating cellularsenescence must be appreciated.

Epigenetic regulation of gene expressionEpigenetic mechanisms are defined as processesin which changes in gene function can occurwithout a change in the DNA sequence. Suchmechanisms include covalent modifications of histones, DNA methylation, ATP-dependentchromatin remodelling, incorporation of varianthistones, looping and changes in the local

conformation of DNA, and alterations of higher-order chromatin structure. It isunderstood that epigenetic processes such asDNA methylation and post-translationalmodification of histones are fundamental to theregulation of gene expression and therefore tothe regulation of multiple cellular processes.For these reasons it is plausible to suggest thatchanges in the chromatin environment can playa role in the changes in gene expression

involved in senescence and also in the bypassof senescence towards tumourigenesis.

The binding of DNA to histone proteins isrequired for efficient packaging of DNA intothe nucleus of the cell, but this tight binding

creates a barrier for factors that need access toDNA to regulate various cellular processes.Histone proteins bind together to form ahistone octamer comprising two copies each of histones H2A, H2B, H3 and H4 to which DNAwraps round. Each member of the octamer can

  be post-translationally modified and onefunction of these modifications is to mediate

On-rampTelomerase

gene expression

     O    n   -    r    a    m    p

   N  o   t  e   l  o  m  e  r  a  s  e  a  c   t   i  v   i   t  y

   A   L   T  p  a   t   h  w  a  y

  r  e  c  o  m   b   i  n  a   t   i  o  n

Telomerase activity

Immortality

Senescence

E   x  i   t   r  o  u  t  e  

E   x  i   t   r  o  u  t  e  

Normal cell

NeoplasiaImmortality

Routes to immortality

Expert Reviews in Molecular Medicine

© 2007 Cambridge University Press

Figure 1. Routes to immortality. Cellular 

senescence (forward and reverse exit routes)

provides a barrier to excessive cellular growth and

so is considered to be a tumour suppressor 

mechanism. A normal cell can bypass senescence

and move towards neoplasia through

immortalisation by two means, or two different ‘on-

ramps’. First, cells can maintain their telomeres

through telomerase activation (horizontal on-ramp).

Second, cells can engage the alternative

lengthening of telomeres (ALT) pathway (vertical on-

ramp), which allows telomere elongation through

intra-telomeric recombination processes. The

induction of premature cellular senescence of 

neoplastic cells (reverse exit route) may therefore be

a strategy for anti-cancer therapeutics.

expert reviewshttp://www.expertreviews.org/ in molecular medicine

2  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 3/26

the binding of the histone proteins to DNAand therefore to regulate DNA access andthe transcriptional status of DNA therein.Modifications occur mainly on the histoneN-terminal tail, but also occur on the

histone globular domain, and includeacetylation, methylation, phosphorylation,SUMOylation (for ‘small ubiquitin-relatedmodifier’), ubiquitination and ADP ribosylation.It has been hypothesised that one of the roles of these modifications is to act as a ‘histone code’(Refs 16, 17) in which combinations of histonemodifications can be read as a code andtranslated into an output signal; thus, distinctpatterns of histones may be a signal for genetranscription to be turned on or off.Furthermore, levels of modifications mightfunction to regulate levels of transcription (and

therefore high or low levels of gene expression).In general, silenced or repressed chromatin

(heterochromatin) is characterised by hypo-acetylated histones (histones H3 and H4),methylation of residues Lys9 and Lys27 of histone H3 (K9 H3 and K27 H3) andmethylation of Lys20 of histone H4 (K20 H4).Active, de-repressed chromatin (euchromatin)is characterised by hyper-acetylated histonesand can have methylation of Lys4, Lys36and Lys79 of histone H3 (K4 H3, K36 H3 andK79 H3) (Refs 18, 19). In addition, DNAhypermethylation tends to be associated with

silent chromatin whereas hypomethylationgenerally associates with active chromatin.

Senescence, chromatin and diseaseTheinduction of premature senescence of tumourcells by chemotherapeutics is utilised in anti-cancer therapy. However, it is important torealise that cellular senescence contributes tothe ageing process, can lead to the dysfunctionof tissues such as the heart, lungs and brain(Ref. 20), and is also linked to the pathogenesisof multiple disease states (Table 1) such

as smoking-related lung disease, transplantrejection and liver cirrhosis. Interestingly, it hasalso been demonstrated that senescent cellsmay promote tumourigenesis by providinga permissive tissue microenvironment fortumourigenic cell growth (Refs 21, 22, 23).Therefore, although senescence is an important

  barrier to cancer progression, this benefit might  be offset by the contribution of cell arrest toother disease states in later life.

In addition, similarly deregulated epigeneticmechanisms have been shown to contributeto ageing, cardiovascular disease, multipleinherited disorders and also tumourigenesis(Table 2). Furthermore, the epigenetic basis of tumourigenicity is an intensely studied area(Refs 13, 14, 24, 25, 26) and it is now wellestablished that the deregulation of epigeneticmodifying enzymes, leading to deregulatedgene expression, plays an important role intumourigenesis (Ref. 14). Such studies have ledto the establishment of an epigenetic model of 

cancer to complement, or even replace, thegenetic model for cancer and hypothesises thatepigenetic modifications can play a role intumour initiation and progression (Ref. 26).Another interesting model suggests thatepigenetics is at the very root of tumourigenicity,

  by disrupting gene expression at the stem celllevel (Ref. 27). Epigenetic disruption is thereforean important mechanism in the key early eventsin tumourigenesis.

How regulation of the chromatinenvironment affects senescence

Heterochromatin formation and senescenceInitial studies previously established thatchromatin becomes reorganised with increasedpopulation doublings, with large-scale effects

 being evident upon senescence (Ref. 28). It hasnow been shown that the formation of heterochromatin at specific foci plays asignificant role in senescence in response tooncogenic insult and telomere attrition, and thatthis senescence arrest as a cellular response to

Table 1. Senescence and disease

Senescence-associateddisease

Refs

Tumourigenesis 21, 22, 23

Wound healing 173Neurodegeneration 174, 175

  Alzheimer’s disease 176

Chronic obstructive

pulmonary disease (COPD)

177

Emphysema 178

Kidney failure 179

Cardiovascular disease 180

Chronic inflammation 181, 182

Transplant rejection 183

Osteoarthritis 184

Liver cirrhosis 185

  Atherosclerosis 172

expert reviewshttp://www.expertreviews.org/ in molecular medicine

3  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 4/26

stress might act as a tumour suppressor

mechanism (Refs 2, 3). In addition, it has also been observed that many genes upregulated atsenescence are actually physically clustered(Ref. 29), again indicating that re-organisation of chromatin domains are important in senescence.Therefore, formation of heterochromatin may beviewed as a tumour suppressor mechanism.

Early studies showed that expression of oncogenic Ras induced the accumulation of p16INK4a (also known as cyclin-dependent kinase

inhibitor 2A) and hypophosphorylated pRB

(retinoblastoma protein) with a senescence-likearrest in normal human fibroblasts (Ref. 30). Itwas also noted that this senescence-like arrestwas associated with the formation of distinctheterochromatic structures, which weredesignated senescence-associated heterochromatinfoci (SAHF) (Ref. 30). Importantly, SAHF wereshown to be formed by newly depositedheterochromatin rather than from redistributedheterochromatin, and were concentrated for

Table 2. Chromatin and disease

Disease Gene/epigeneticmechanism

Mechanism of diseasepathogenesis

Refs

Inherited

  Angelman syndrome Aberrant genomic imprinting 186Prader– Willi syndrome Aberrant genomic imprinting 186

Beckwith–Wiedemann

syndrome

  Aberrant genomic imprinting 186

ICF syndrome DNMT3a Gene mutation 186

Rett syndrome MECP2 Gene mutation 187

 Autoimmune

Systemic lupus

erythematosus

Changes in DNA methylation and

histone modifications in lupus cells

188

Cardiovascular

Stress-induced

remodelling

Histone acetylation Gene regulation alterations lead to

remodelling/weakening of the heart

189

Neural

Huntington’s disease Histone acetylation Altered gene expression 190

 Alzheimer’s disease HDAC genes Inhibition leading to gene expression

changes may alleviate symptoms

191

Histone acetylation Deregulated acetylation levels

may be threat to neural health

192

Epilepsy Histone acetylation May play role in development

of epilepsy

193

Lung

Lung inflammatory

disease

Histone acetylation Altered expression of inflammatory

genes

194

Spinal

Spinal muscular 

atrophy

Histone acetylation Aberrant changes in gene expression 195

OtherDepression HDAC5 Aberrant changes in gene expression 196

Tumourigenesis Histone modification Aberrant changes in gene expression

and possible increased genetic

instability

14, 39,

154, 155

DNA methylation Aberrant changes in gene expression

and increased genetic instability

124, 197

 Abbreviations:DNMT3a, DNA (cytosine-5-)-methyltransferase 3 alpha; HDACs/HDAC5, histone deacetylases/histone deacetylase 5; ICF syndrome,centromereinstabilityand facial anomaliessyndrome;MECP2, methylCpGbinding protein 2.

expert reviewshttp://www.expertreviews.org/ in molecular medicine

4  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 5/26

repressive histone modifications such asmethylation of K9 H3 and heterochromatinprotein 1 (HP1), whereas permissivemodifications, such as methylated K4 H3 andacetylated K9 H3, were excluded (Fig. 2). It was

also established that active transcription was notoccurring at the sites of SAHF formation and thatchromatin features indicative of SAHF formed onE2F-responsive promoters of genes known to bedownregulated at senescence. E2F is atranscription factor important for regulation of genes associated with the cell cycle and was firstidentified as a cellular factor that had the ability

to bind the promoter for the gene encodingadenovirus E2. The genes that gainedheterochromatic features and becamedownregulated included the genes encodingcyclin A and proliferating cell nuclear antigen

(PCNA), and their promoters were bound by pRB,thereby linking pRB binding to the formation of SAHF. It is known that the upregulation of p16INK4a during senescence can inhibit thephosphorylation of pRB, causing it to bind to E2Fto cause a permanent growth arrest by silencinggrowth regulatory genes through altering thechromatin state of the promoter sequences. pRBhas been shown to interact with factors involvedin the formation of heterochromatin, includinghistone deacetylases (HDACs) (Refs 31, 32, 33),the K9 methyltransferase SUV39H1 (Ref. 34) andHP1 proteins (Ref. 35); thus, pRB binding to

regulatory sequences could be sufficient to formSAHF in order to silence gene expression.Importantly, the authors showed in the samestudy that promoters of genes known to beupregulated at senescence gained euchromaticpatterns at their promoters at the same time asSAHF were being formed on other promoters,

Figure 2. The formation of senescence-

associated heterochromatin foci (SAHF) in

response to oncogenic insult. In this model, (a)

an active gene promoter (e.g. cyclin D1) shows

methylation of Lys4 (K4) H3 and acetylation of 

H3 and H4, and its chromatin has an open(euchromatic) structure. Under these conditions,

the transcription factor E2F mediates transcription,

whereas retinoblastoma protein (pRB) remains in a

phosphorylated state as a result of low p16INK4a

levels and therefore cannot bind to E2F to repress

transcription. (b) Upon oncogenic insult

(e.g. aberrant Ras signalling), pRB becomes

hypophosphorylated, mediated by increased levels

of p16INK4a. Hypophosphorylated pRB is

sequestered to the promoter along with several

other proteins, such as heterochromatin protein 1

(HP1), histone deacetylase (HDAC) and the Lys9

(K9) H3 methyltransferase SUV39H1, which can

bind pRB. (c) This leads to the silencing of gene

expression by altering the chromatin architecture

towards a heterochromatic state by promoting

K9 H3 methylation and removing/inhibiting

euchromatic modifications, as is observed in SAHF

formation. SAHF formation as a mode of gene

expression regulation in response to oncogenic

insult might lead to the onset of premature

senescence and might therefore inhibit

tumourigenic growth.

P

P

PP RB

RB

HP1

E2F

Openchromatinstructure

Closedchromatinstructure

Oncogenic insulte.g. aberrantRas signalling

Increased p16expression

a

b

c

Geneexpression

Generepression

E2F

SAHF formation

SUV39H1 HDAC

RB

HP1HP1

E2F

SUV39H1 HDAC

H3 and H4acetylation

K4 H3methylation

K9 H3methylation

The formation of senescence-associated heterochromatin foci(SAHF) in response to oncogenicinsult

Expert Reviews in Molecular Medicine

© 2007 Cambridge University Press

expert reviewshttp://www.expertreviews.org/ in molecular medicine

5  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 6/26

suggesting that the formation of SAHFs might bedirected towards certain genes and not anindiscriminate global effect. This indicates thatmodification of the chromatin environment of gene promoters is important for the stable

senescence-like arrest attained. It also suggests amodel in which SAHF formation accompanies thesenescence process and indicates a role for pRB-mediated heterochromatin formation at growthregulatory genes during senescence, leading tothe stable repression of these genes. From this itwas further proposed that failure of certainprocesses in these cells (e.g. mutations in genesencoding p16INK4a or pRB, or silencing events)could lead to bypass of senescence andprogression to cancer as a result of the inhibitionof SAHF formation (Ref. 30).

Several other papers have provided more

evidence of the link between chromatinremodelling and senescence. Braig et al. havedemonstrated that oncogenic Ras could inducea senescence-like arrest in mouse primarylymphocytes by a SUV39H1-dependent and K9H3 methylation-related mechanism (Ref. 36). Inthe same study, the deletion of SUV39H1 inprimary lymphocytes led to the impropermethylation of K9 H3 and the promotion of Ras-driven lymphomagenesis. This study alsocorrelates with other work demonstrating thatloss of methylation of K9 H3 and also K20 H4resulted in genomic instability in a variety of 

normal cells and was associated with increasedtumour risk (Ref. 37). Decreased expression of SUV39H1 has also been observed in the earlypremalignant stage of tumour development inthe context of hepatocarcinogenesis induced bymethyl deficiency in rats, whereas loss of tri-methylation of K20 H4 was also observed at

  both preneoplastic and tumour stages of livercancer and corresponded to a decrease in theK20 H4 histone methyltransferase, SUV420H2(Ref. 38). Interestingly, loss of tri-methylation of K20 H4, as well as loss of acetylated K16 H4

and DNA hypomethylation, has been shown to be a common epigenetic change in tumourigeniccells (i.e. an ‘epigenetic signature’ of cancer)(Ref. 39). These studies provide evidence thatimproper heterochromatin maintenance mightlead to genomic instability and progression totumourigenicity.

Michaloglou et al. have also demonstratedthat BRAFE600-mediated oncogenic signallingin melanocytes induces a senescence-like

phenotype with the formation of SAHF (Ref. 40).BRAF is a protein kinase and downstreamregulator of Ras, and the V600E mutation in thegene encoding BRAF is an oncogenic mutationobserved frequently in human naevi (moles). A

further study focusing on Ras utilised a mousemodel with a conditional oncogenic K-rasV12allele activated by the Cre recombinase (Ref. 41).Upon activation of oncogenic Ras, the micedeveloped multiple lung adenomas (pre-malignant) and a few lung adenocarcinomas(malignant), and markers for senescence wereevident in the adenomas but not in thecarcinomas. Interestingly, HP1g was stronglypositive in the adenomas, whereas thecarcinomas were negative. As the HP1 proteinscan bind methylated K9 H3 to mediate genesilencing, this study further reinforces the role

of epigenetics in senescence. The formation of SAHF has also been shown in response tooverexpression of E2F3 (Ref. 42) and repressionof the E1A (adenovirus 5 early region 1Aprotein)-associated protein p400 (Ref. 43). In thefirst study, which analysed whether deregulatedE2F activity would be sufficient to mimic loss of pRB, short-term deregulation of E2F3 led tohyperplasia of quiescent melanotrophs but didnot lead to tumourigenicity due to the inductionof senescence with the appearance of SAHF(Ref. 42). p400, a protein belonging to the SWI2/SNF2 family of chromatin-remodelling proteins

and postulated to be a transforming agent, wasstudied in relation to the p53/p21 senescencepathway (Ref. 43). When p400 expression wasrepressed in normal human fibroblasts and inhuman fibroblasts transduced with humantelomerase reverse transcriptase (hTERT),senescence was induced along with theappearance of SAHF, suggesting that p400expression could repress p53 induction of p21expression, possibly allowing the bypass of thesenescence arrest.

Further studies into the formation of SAHF

have also indicated roles for the chromatinregulators histone regulating A (HIRA) andanti-silencing factor 1 (ASF1), which wereshown to be rate limiting for the formation of SAHF and were required for senescence(Ref. 44), whereas the high-mobility group A(HMGA) proteins have been shown to be majorstructural components of SAHFs (Ref. 45).HMGA proteins are usually associated withthe formation of chromatin environments

expert reviewshttp://www.expertreviews.org/ in molecular medicine

6  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 7/26

permissive for gene transcription, but have beenshown to be vital for SAHF formation and generepression in Ras-induced senescence of normalhuman fibroblasts (Ref. 45). Also, similar to therole of pRB, the growth regulatory protein

prohibitin, which has previously been linked tothe regulation of senescence (Ref. 46), has also  been shown to localise to SAHFs induced byDNA-damaging agents (Ref. 47). It was shownthat prohibitin plays an integral role in thestability of the senescent phenotype bymaintaining the repression of E2F-responsivepromoters through the maintenance of arepressive chromatin environment by interactionwith HP1g. Reduction in the cellular levels of prohibitin led to a reduction in the numberof SAHFs and impaired the ability of the cells toundergo senescence, further indicating an

integral role for epigenetics in cellular senescence.The formation of SAHF in senescence arrest has

also been studied in vivo in ageing primates(Ref. 48). Increases in the presence of telomere-dysfunction-induced DNA damage foci werecorrelated to increasing age in dermal fibroblastsand, importantly, this was further correlated toSAHF formation. This study indicates thattelomere dysfunction, occurring as a result of telomere attrition with increased cellularproliferation, also induces SAHF formation.Whether this occurs in human cells with ageingis as yet unknown, but this study does indicate

that SAHF formation at senescence is not only aresponse to oncogenic stress, but might also be acommon mechanism in cellular senescence.

In summary, multiple studies have now shownthe strong correlation between the formationof heterochromatin and the attainment andmaintenance of the senescence phenotype.Repression of proliferation-associated genes bythe formation of heterochromatin serves to

 block cellular division and so acts as a potenttumour suppressor mechanism. Furthermore,improper regulation of heterochromatin is

associated with deregulated cellular proliferationand tumourigenesis.

Epigenetic modifications by thesenescence mediators p53 and INGSimilar to pRB, other tumour suppressor proteinshave also been shown to interact with histone-modifying enzymes to regulate transcription(Ref. 49). p53 is an important mediator of senescence and one of its main transcriptional

targets is p21. It is known that p53 binds to andactivates the p21 promoter and is mediated, inpart, by increased promoter-proximal histoneacetylation (Ref. 50) through the recruitmentof histone acetyltransferases (HATs) (Ref. 51)

and the displacement of HDAC1 (Ref. 52).Interactions between p53 and the HAT p300 andprotein arginine methyltransferases, which canalso mediate transcriptional activation by post-translational modifications of histones, have also

 been reported to be required for p53-dependentgene activation (Ref. 53). However, p53 alsointeracts with histone deacetylases to mediategene repression as it has been shown that p53

  binding to promoters was concomitant withincreases in levels of K9 methylation and loss of acetylation of K9 H3 (Refs 54, 55, 56). Given thatepigenetics plays a key role in transcriptional

control by p53, it is important to note thatderegulation of epigenetic mechanisms couldaffect p53-mediated senescence or apoptosis.

Theinhibitor of growth (ING) familyof proteinsare also important regulators of cellularsenescence through the regulation of geneexpression (Ref. 57). Like p53, HATs and HDACshave both been shown to interact with the INGproteins to mediate chromatin-remodellingactivities and gene expression (Ref. 58). Studieshave shown a direct interaction of an ING familymember with chromatin upon senescence,suggesting that ING proteins might be important

mediators of senescence through epigeneticcontrol of gene expression (Refs 59, 60).Furthermore, it has been shown that ING2 can

  bind di- and tri-methylated K4 H3 to mediategene repression through histone deacetylation

  by associated factors in the mSin3a–HDACrepressor complex (Ref. 61), allowing subsequentrepressive lysine methylation required for theformation of heterochromatin. This reversal of chromatic states (from euchromatic to hetero-chromatic) is an important mechanism to repressproliferation-associated genes at senescence, and

one method to realise such a reversal could  be the sequestering to chromatin of ING2-mediated histone deacetylation by a modificationassociated with active gene expression(methylation of K4 H3).

Reversing the irreversible: a newepigenetic mechanism uncoveredMethylation of K9 H3 plays an important role inthe maintenance of gene repression and in

expert reviewshttp://www.expertreviews.org/ in molecular medicine

7  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 8/26

SAHF-mediated gene repression at senescence.Methylation of K9 H3 was generally understoodto be a stable, long-lasting modification, allowingfor effective gene silencing. However, multiplestudies have now shown that methylation of K9

H3 is readily reversible (Refs 62, 63, 64, 65, 66)and it has been suggested that enzymes thatmediate K9 H3 demethylation could also affecttumourigenesis (Ref. 65). GASC1 (for ‘geneamplified in squamous cell carcinoma 1’; alsoknown as JMJD2C, for ‘jumonji domaincontaining 2 family member C’) is one suchprotein found with the ability to demethylate di-and tri-methylated K9 H3 (Ref. 65). This,coupled with previous findings that GASC1 wasamplified in several carcinoma cell lines and thatinhibition of the protein led to a decrease incellular proliferation, led to the suggestion

that GASC1 could act as an oncogene.Hypothetically, overexpression of GASC1 orother K9 H3 demethylases could lead to thedissolution of SAHF and to the bypass of senescence by the de-repression of proliferation-associated genes. In such a model, the formationof SAHF in response to a senescence-inducingsignal would ‘force’ cells to undergo prematuresenescence and stop possible tumourigenicity.However, if SAHF formation was inhibited bythe overexpression or aberrant localisation of aK9 H3 demethylase, this could allow cells tomove preferentially towards tumourigenesis and

 bypass senescence (Fig. 3).

Polycomb group proteins, stem cellsand senescenceAlthough it has been shown that the gain of heterochromatin by increased levels of repressive histone modifications is linked tosenescence, the gain of repressive histone-modifying activity by Polycomb group (PcG)proteins and gene silencing has also beenobserved to allow extension of lifespan and isinvolved in tumourigenesis. There are two

distinct PcG complexes, Polycomb repressivecomplex 1 and 2 (PRC1 and PRC2); thesemultiprotein complexes can repress target geneexpression by catalysing the addition of repressive modifications of histones, such asK27 H3 methylation, and allowing thecompaction and maintenance of chromatin intoa heterochromatic structure.

Loss of function of the PcG protein BMI1 (for‘B-cell-specific Moloney murine leukaemia

virus integration site 1’) was shown to impairthe progression of mouse embryonic fibroblastsinto S-phase; indeed, these cells underwentpremature senescence, thereby linkingtranscriptional repression by PcG proteins with

cell-cycle control and senescence (Ref. 67).Additionally, BMI1 has been shown to bedownregulated in senescing human fibroblasts(Ref. 68), whereas overexpression has beenobserved in tumours and tumour cell lines(Refs 69, 70, 71, 72) and BMI1 is required for theshort-term survival of cancer cells (Ref. 73).

The role of senescence-associatedheterochromatin foci (SAHF) insenescence and tumourigenesis

Expert Reviews in Molecular Medicine

© 2007 Cambridge University Press

‘Normal’cell

SAHF formation

Senescence signala

‘Normal’cell

SAHF formation

Senescence signalb

Senescence Tumourigenesis

Figure 3. The role of senescence-associated

heterochromatin foci (SAHF) in senescence and

tumourigenesis. (a) Formation of SAHF after 

oncogenic stress in a ‘normal’ cell might promote

the induction of senescence over tumourigenesis

by the repression of specific genes. (b) However,

if SAHF formation is inhibited, potentially by

the reversal of Lys9 (K9) H3 methylation,

tumourigenesis may be promoted owing to

‘blockage’ of the route towards senescence.

expert reviewshttp://www.expertreviews.org/ in molecular medicine

8  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 9/26

Interestingly, additional studies have shown thatoverexpression of BMI1 allows immortalisationand transformation of some human cells(Refs 67, 74, 75). Importantly, the INK4a locusfrom which the cyclin-dependent kinase

inhibitors p16INK4a

and p14ARF

are transcribedare crucial downstream targets for BMI1 withregard to its effects on cell proliferationand senescence (Ref. 68). Therefore, throughinhibiting the expression of these cyclin-dependent kinase inhibitors, BMI1 can lead tothe promotion of cell-cycle progression.

EZH2 (for ‘enhancer of zeste homolog 2’),another PcG protein, has been shown tomethylate K27 H3 (Ref. 76) and is alsooverexpressed in some cancers, again linking theformation of heterochromatin to the extension of lifespan (Refs 77, 78, 79). EZH2 expression is

also known to be repressed by pRB andactivated p53, suggesting a possible mechanismfor EZH2 overexpression in cancers (Ref. 79)and is further observed to be specificallydownregulated in senescent cells (Ref. 80). Lossof K27 H3 histone methyltransferase activity atsenescence might correspond to loss of heterochromatin and activation of genes suchas those encoding p16INK4a and p14ARF, leadingto cellular senescence (Ref. 81). By contrast,overexpression of EZH2 in cancers might lead tothe proper maintenance of heterochromatin atthe INK4a locus, allowing the elongation of 

lifespan in cancer cells by bypassing senescencethrough the repression of expression of genessuch as those encoding p16INK4a and p14ARF.

Complementing these reports have been recentstudies on epigenetics in embryonic stem cells(ESCs) and how epigenetics affects pluripotency;these studies have suggested another route bywhich a normal cell might bypass senescence andgain tumourigenic properties. The chromatinenvironment of ESCs has been shown to behyperdynamic and therefore in a highlypermissive state for transcription. However, as a

cell differentiates along various lineages, specificgenes will become repressed, whereas others arefurther activated, leading to a less dynamic andtherefore less permissive chromatin environment(Refs 82, 83, 84). PRC1 and PRC2 have now

  been shown to be integral in maintainingpluripotency by initiating and maintaining thesilencing of genes involved in differentiation bymarking such genes with K27 H3 methylation(Refs 85, 86, 87, 88). Therefore, if any of the

components of PRC1 and PRC2 becomeoverexpressed in a normal cell, genes involved indifferentiation could become epigeneticallysilenced and perhaps allow the reversion of anormal cell towards a more pluripotent state

(Fig. 4). This could allow pluripotent stem-likecells to arise in normal tissues and nucleatecancer growth. Supporting this, as mentionedpreviously, it has been shown that genes suchas those encoding BMI1 and EZH2 areoverexpressed in some cancers (Refs 69, 70, 71, 72,77, 78, 79).

These studies firmly establish a link betweenPcG group proteins, senescence and themodulation of the chromatin environment.Supporting this hypothesis is a recent study of PcG group proteins in prostate cancer (Ref. 89).This study showed that increased expression of 

BMI1 and EZH2, with increases in K27 H3methylation and gene silencing, were linked toincreased malignant potential and metastasis inprostate cancer and were also linked to failurein therapy of prostate cancer. Prior work bythe same group also identified an 11-genesignature that was common between normalstem cells with normal self-renewal capacityand malignant tumour cells (Ref. 90). Thesepapers suggest that PcG-mediated genesilencing can mediate increased malignancyperhaps by bestowing stem cell properties topreviously non-metastatic cancer cells with low

malignant potential; this could occur throughthe repression of multiple developmental anddifferentiation pathways by epigenetic changesto gene expression. Thus, PcG-mediatedpathways are now known to be involved in bothnormal stem cell function and highly malignanttumour cells. This reinforces the concept thatregulation of the chromatin environment andgene regulation by the PcG pathway is veryimportant to proper stem cell function and toprevent highly malignant metastatic tumoursfrom arising.

Support for the ‘reversion’ theory has beenshown in a study of committed progenitor cellsexpressing the mixed lineage leukaemia (MLL)–AF9 fusion protein known to impart leukaemiastem cell (LSC) properties (Ref. 91). These cellsgained the expression of a subset of geneshighly expressed in haematopoietic stem cells(HSCs) and also gained self-renewal properties.The MLL protein is a common fusion partnerin leukaemias and is a trithorax group

expert reviewshttp://www.expertreviews.org/ in molecular medicine

9  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 10/26

(Trx-G) member involved in the transcriptionalactivation of gene expression; MLL is known

to mediate methylation of K4 H3 (Ref. 92).This study is interesting as it links the action of an epigenetic modifier with the reversion of amore committed cell towards a more primitivestem cell type and tumourigenesis.

These studies also raise questions against therising popularity of the stem cell theory of cancer (Ref. 93), by suggesting that somecancers might arise from the gain of  pluripotency by deregulation of the chromatinenvironment in normal somatic cells. However,lending support to the cancer stem cellhypothesis is the premise that epigeneticdisruption of stem cells might be a commonunifying theme in cancer aetiology (Ref. 27),which is an attractive proposition. This so-called ‘epigenetic progenitor model’ might helpto explain many different aspects about cancer,

including initiation, progression, and geneticand epigenetic instability. To understand

Figure 4. Attainment of pluripotency in normal

cells: a possible path towards the cancer stem

cell. (a) In embryonic stem cells (ESCs), Polycomb

group (PcG) complexes [Polycomb repressive

complex 1 and 2 (PRC1 and PRC2)] function to

repress differentiation-associated genes by

repressive histone modifications [Lys9 (K9) H3 and

Lys27 (K27) H3 methylation], whereas pluripotency

genes are expressed with the presence of 

permissive histone modifications [acetylation of 

histones H3 and H4 and methylation of Lys4 (K4)H3]. This allows for the self-renewal capacity

characteristic of stem cells. (b) Loss of the PcG-

mediated repressive modifications leads to the

expression of lineage-specific genes upon

differentiation of ESCs, leading to the cell

becoming more committed to a certain lineage.

Pluripotency genes also become repressed and

therefore these cells lose some ESC

characteristics. (c) Overexpression of PcG proteins

is frequently observed in cancer cells and it is

possible that overexpression of PcG proteins in

normal cells could allow the silencing of lineage-

specific genes involved in differentiation by

repressive histone modifications. (d) This might

also allow the cell to re-express pluripotency-

associated genes and revert back to a pluripotent

state, undergoing deregulated cell proliferation. This

may be an epigenetic mechanism by which normal

cells can bypass senescence and grow without

limit, allowing sustained cell proliferation and self-

renewal. Therefore, this could represent a new

mechanism by which cancer stem cells can arise

and nucleate cancer growth.

 Attainment of pluripotency in normalcells: a possible path towards thecancer stem cell

Expert Reviews in Molecular Medicine

© 2007 Cambridge University Press

Cancerstemcells?

a

b

c

d

Embryonic stem cells

Lineage-specified cells

Deregulated lineage-specified cells

Pluripotent cancer cell

Differentiation

Tumourigenic event

 Attainment ofpluripotency

K27 H3methylation

K9 H3methylation

Pluripotencygenes

Differentiationgenes

H3 and H4acetylation

K4 H3methylation

PRC1/ PRC2

Normal

cells

expert reviewshttp://www.expertreviews.org/ in molecular medicine

10  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 11/26

whether one of these models is predominant orwhether they work in conjunction to fostertumourigenesis is an important goal.

Histone acetylation and senescence

As previously mentioned, histone acetylationmight also have an important role in mediatingthe senescence phenotype. The HATs p300and CREB-binding protein (CBP) were bothstudied in senescing human melanocytes and,interestingly, levels of both proteins wereobserved to decrease as these cells underwentreplicative senescence (Refs 94, 95). Thedecrease in the levels of these HATs led todramatic decreases in total histone H4acetylation levels. Specifically, the promoter forthe gene encoding cyclin E was shown to loseH4 acetylation and become repressed upon

senescence, whereas cyclin E was highlyexpressed and H4 acetylation was high inproliferating cells. This parallels previous studiesof the cyclin E promoter during senescence,which showed that pRB and HDACs bound tothe promoter to repress transcription, linking thesenescent phenotype to the pRB/p16INK4a

pathway and the formation of SAHF (Ref. 30). Asmentioned previously, p53 also interacts withenzymes that control histone acetylation,indicating a role for two main senescencemediatorsin the modulationof histoneacetylation.

However, promotion of histone acetylation bythe use of HDAC inhibitors can induce asenescence-like phenotype in human fibroblasts(Refs 96, 97, 98). Furthermore, in senescingnormal human fibroblasts, HDAC1 becomesdownregulated (Ref. 98), indicating that overallthe promotion of acetylation leads to activationof the senescence phenotype. Interestingly,histone deacetylation has also been shown tohave a role in the extension of cellular lifespan.The yeast protein Sir2 (for ‘silent mating typeinformation regulation 2’), identified as aNADþ-dependent HDAC involved in

ribosomal DNA silencing, contributes to thereplicative lifespan of yeast (Refs 99, 100).Enhanced activity of the gene encoding Sir2was also found to be associated with increasedlongevity in Caenorhabditis elegans (Ref. 101),Drosophila (Ref. 102) and rodents (Ref. 103).Drosophila Sir2 has also been shown to beinvolved in epigenetic gene silencing by thePcG proteins and is physically associated with acomplex containing the Drosophila homologue

of EZH2. This further implicates Sir2 in thecontrol of senescence (Ref. 104) and suggests arole for Sir proteins in PcG-mediated generepression that might affect cellular proliferationand lifespan as discussed above. A human

homologue of yeast Sir2, SIRT1, has also beenassociated with longevity and is believed to actprimarily by inhibiting cellular senescence(Refs 105, 106, 107) and overexpression has beenobserved in leukaemia cells (Ref. 108). SIRT1 andother Sir2 homologues are able to deacetylatetranscription factors and p53, and also have K16H4 deacetylase activity (Ref. 109). Recently, aSIRT1 inhibitor, Sirtinol, was shown to induce asenescence-like growth arrest in human cancercells (Ref. 110) in agreement with previous dataindicating that the use of HDAC inhibitors couldlead to induction of the senescence phenotype.

Additionally, loss of histone acetylation has  been linked to tumourigenesis throughdecreased K16 H4 acetylation, as well asdecreased tri-methylation of K20 H4 and DNAhypomethylation, as demonstrated in cancercells and tissues (Ref. 39).

These studies suggest that theincreases in globalhistoneacetylation can lead to senescence, perhapsmediated by the re-expression of genes previouslysilenced by heterochromatin that, when activated,promote senescence. By contrast, de-acetylationmight serve to repress genes that contribute tothe senescence phenotype. Again, this shows

that regulation of the chromatin environment iscrucial for proper cellular control.

DNA methylation and senescenceGenerally, histone modifications and DNAmethylation are often discussed separately inrelation to their effects on gene expression,although it is now known that they are in factintimately linked (Refs 111, 112, 113, 114), withmultiple interactions now known betweenproteins associated with histone modificationsand those associated with DNA methylation.

Generally, DNA methylation of CpG islands(regions of adjacent cytosine and guanineresidues linked by phosphodiester bonds) atgene promoters is correlated with generepression, whereas expression is linked to thelack of methylation. Global DNA methylationhas been shown to decrease with age (Ref. 115)and this loss had been hypothesised to functionas a counting mechanism for senescence, suchthat senescence would occur when a cell loses a

expert reviewshttp://www.expertreviews.org/ in molecular medicine

11  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 12/26

certain degree of DNA methylation (Refs 115,116). Treatment with 5-azadeoxycytidine(5-azadC), a DNA demethylating agent, has

 been shown to reduce the replicative lifespan of normal human fibroblasts, supporting the DNA

methylation counting mechanism hypothesis(Refs 117, 118). Also in agreement with thishypothesis are the findings that DNAmethyltransferase activity is elevated in cancercells in vitro, in tumours in vivo (Ref. 119) andin transformed cells (Refs 120, 121), whereasDNA methyltransferase activity decreases inserially passaged normal human fibroblasts(Ref. 122). Therefore, the loss of DNAmethylation with age might eventually havedeleterious consequences, such as deregulationof gene expression. Consistent with this view, ithas been hypothesised that DNA methylation

might repress a set of growth inhibitory genes(Refs 24, 123) and, as the number of cellpopulation doublings increases and DNAmethylation decreases, the repression of suchgrowth inhibitory genes might become lessstringent, leading to their eventual expressionand contributing to senescence. However,it has also been shown that a striking feature of some tumours (both benign and malignant) isthat global DNA methylation is reduced(Refs 124, 125). This loss of DNA methylationis also linked to chromosomal instability,which can contribute to tumourigenesis

(Refs 126, 127). Thus, paradoxically, it has beenshown that the loss of DNA methylation cancontribute to senescence and tumourigenicity.However, it is acknowledged that gene-specificmethylation is variable in cancer and that manytumour suppressor genes show promoter-specific DNA hypermethylation leading togene silencing, including the genes encodingpRB (Ref. 128) and p16INK4a (Ref. 129), whereasthe genome is hypomethylated overall.Interestingly, recent data have shown thatp16INK4a is crucially involved in DNA

methylation, leading to gene silencing and breasttumourigenesis (Ref. 130). Loss of p16INK4a leadsto the increased expression of key PcG proteins,resulting in a specific pattern of DNAhypermethylation and importantly the silencingof the gene encoding HOXA9, a transcriptionfactor that is epigenetically silenced in breasttumourigenesis. Therefore, it is likely thatglobal loss of DNA methylation leading tochromosomal instability could trigger cellular

senescence and that promoter-specific alterationsin key tumour suppressor genes might allow the

 bypass of senescence and lead to tumourigenicity.

Senescence and DNA damage detection

by histone modificationHistone modifications can also play a role in thedetection and repair of DNA damage that, aspreviously mentioned, can lead to prematuresenescence. DNA damage can be incurredthrough the actions of various types of radiation(Ref. 131), DNA-damaging drugs (many of which are used as anti-tumourigenic therapies)(Ref. 132) and oxidative stress (Ref. 133), andhistone phosphorylation is now know to play arole in mediating cellular responses to this DNAdamage. Signalling kinases, notably ATM (for‘ataxia telangiectasia mutated’) and ATR (for

‘ataxia telangiectasia and Rad3 related’), arerecruited to the site of damage and are activated,leading to phosphorylation of histone H2AXmolecules (g-H2AX) (Ref. 134). Histone H2AX isa variant of the H2A histone that makes up partof the histone octamer. The phosphorylation of histone H2AX facilitates the recruitment of checkpoint and DNA repair factors includingp53-binding protein 1 (153BP1), and promotesthe activation of checkpoint kinase 1 (Chk1) andChk2, which can activate p53 and therebymediate cellular senescence. Furthermore, it isknown that the uncapping of telomeres can leadto senescence through a DNA damage responsemediated by a similar mechanism (Refs 135, 136).Phosphorylated histone H2AX also serves torecruit histone modifiers and ATP-dependentchromatin remodellers to these sites of DNAdamage and aid the repair of these sites (Ref. 137).

Two recent studies have uncovered a link  between histone lysine methylation and theDNA damage checkpoint and double-strand

 break (DSB) repair proteins 53BP1 in mammalsand Schizosaccharomyces pombe putative homologCrb2 (for ‘Crumbs-like protein 2’): the

relocalisation of 53BP1 and Crb2 to DNA DSBshas been found to require histone methylation(Refs 138, 139). It was proposed that 53BP1senses DSBs through changes in higher-orderchromatin structure that occur upon exposureof the 53BP1 binding site [methylated Lys79 H3(K79 H3), which lies in the globular domain of histone H3] (Ref. 138). In S. pombe, a similarmechanism occurs with Crb2 binding tomethylated K20 (Ref. 139). However, recent

expert reviewshttp://www.expertreviews.org/ in molecular medicine

12  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 13/26

studies have shown that in fact 53BP1 and Crb2  bind to the same modified lysine residue, di-methylated K20 H4 (Ref. 140).

Epigenetic regulation of telomerase

gene expression and senescenceTelomerase and telomeres in senescenceIn adult humans, most normalcells have lowor nodetectable telomerase activity (Ref. 141). The lackof telomerase activity, or other telomeremaintenance mechanisms, leads to the loss of around 50–100 bp of telomeric sequence at eachcell division (Refs 142, 143). The loss of telomeric tract during cell division presents alimit to the maximum number of divisions acell can undertake before entering senescenceand therefore provides a limit to its lifespan(Ref. 144). Cellular senescence as a response to

telomere attrition is thought to have evolved asa tumour suppressive barrier against excessiveclonal expansion. However, the detection of telomerase activity in an estimated 85% of malignant tumours, together with the stronglinks between telomerase activity and malignantcells (Refs 141, 145), indicates the importance of telomerase activity in tumourigenesis and alsosuggests that telomerase activation is the mostcommon mechanism for telomere maintenancein cancer cells. However, several immortal celllines and tumours lack telomerase activity, yethave the ability to elongate their telomeres.Such non-telomerase-dependent mechanisms of telomere maintenance include the alternativelengthening of telomeres (ALT) pathway(Ref. 146) (Fig. 1).

In cells where ALT is utilised, telomere length ismaintained by a recombination-based DNAreplication mechanism (Ref. 147) and leads toheterogeneity in telomere lengths (Ref. 148).Also associated with use of the ALT mechanismis the presence of ALT-associated promyelocyticleukemia protein (PML) bodies, called APBs(Ref. 149), which contain the protein PML,

telomeric DNA, the telomeric-binding proteinstelomere repeat factor 1 (TRF1) and TRF2, andproteins involved in DNA recombination andreplication. The utilisation of ALT in tumourcells is much rarer than the utilisation of telomerase activity (Ref. 148) and it has beenhypothesised that ALT might normally occurunder selective pressure when expression of telomerase is prevented. ALT is commonlyactivated in tumours of mesenchymal origin,

including osteosarcomas, soft tissue sarcomasand glioblastoma multiforme (Refs 150, 151,152, 153). Furthermore, in normal mesenchymaltissues, telomerase activity is more tightlyrepressed that in epithelial cells, possibly leading

to the selective pressure for ALT activation overtelomerase activity in these cell types.

Epigenetic regulation of telomeremaintenance mechanismsExpression of the human telomerase RNAcomponent (hTR) and hTERT are understood to

  be the minimal requirements for telomeraseactivity. Studies have shown that specificepigenetic modifications at the hTR and hTERTpromoters correlate with expression orrepression of hTR and hTERT (Refs 154, 155).Lack of expression of hTR in ALT cell lines, and

hTERT in ALT and normal cell lines, isassociated with a repressive chromatinenvironment at the appropriate gene promoter.Conversely, expression of hTR in ALT andtelomerase-positive cell lines, and hTERTexpression in telomerase-positive cell lines, isassociated with a more permissive chromatinenvironment at the appropriate gene promoter.Moreover, in the pre-senescent normal fibroblastcell line WI38, which has no detectable hTERTexpression and low hTR expression, thetelomerase gene promoters contain no greatquantities of either positive or negativechromatin modifications, and therefore thechromatin environment of the promoter remainsin a neutral chromatin configuration (Fig. 5a).

Furthermore, upon comparison of the hTERTpromoter chromatin environment in ALT celllines (including a cell transformed from theparental WI38 cell line WI38-SV40) withthe normal WI38 cell line, an increase in theamounts of repressive modifications is evidentand links the heavy repression of hTERTexpression by the chromatin environment to theALT phenotype. In the telomerase-positive

tumour cell lines, permissive modifications aregained at the hTERT promoter relative to theWI38 cell line. Similarly, heavy repression of hTR is evident in some ALT cell lines with nohTR expression, whereas high levels of permissive chromatin modifications are linkedto hTR expression. Additionally, treatment of normal and ALT cells with 5-azadC incombination with the HDAC inhibitortrichostatin A (TSA) caused chromatin

expert reviewshttp://www.expertreviews.org/ in molecular medicine

13  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 14/26

remodelling of both promoters and led to theformation of a more permissive chromatinenvironment, as well as reactivation orincreased expression of hTR and hTERTexpression. This suggests a model by which

gain of permissive modifications at thetelomerase gene promoters leads to telomeraseactivity by the expression of hTR and hTERT,whereas the gain of repressive modificationsleads to activation of the ALT mechanism(Fig. 5b). Therefore, epigenetic deregulation of 

Figure 5. Epigenetic regulation of telomere

maintenance mechanisms. (a) Histograms

showing a simplistic view of what we know about

the relationship between the relative levels of 

repressive and permissive chromatin modifications

at the telomerase gene promoters in telomerase-

expressing cancer cells, pre-senescent normalcells and cells that maintain their telomeres by the

alternative lengthening of telomeres (ALT)

mechanism. The presence of permissive histone

modifications at the telomerase gene promoters is

linked to telomerase activity, whereas the presence

of repressive histone modifications is linked to the

activation of ALT. Both of these scenarios are in

contrast to the chromatin environment of pre-

senescent normal cells, in which permissive and

repressive modifications are at a minimum. This

suggests a model by which epigenetic deregulation

of the telomerase gene promoters could lead to

aberrant telomere maintenance. (b) Model depicting

how chromatin modifications might influencetelomere maintenance mechanisms. In a ‘normal’

cell that does not have telomerase activity and has

no or low levels of expression of the telomerase

genes, few chromatin modifications are present at

the gene promoters. The choice of telomere

maintenance mechanism may be influenced

through epigenetic mechanisms and changes

in histone modifications, possibly during

tumourigenesis. In the left-hand pathway shown,

telomerase gene expression is linked to the

accumulation of permissive histone modifications

[acetylation of histones H3 and H4 and methylation

of Lys4 (K4) H3], which leads to telomerase activity

and the elongation/maintenance of telomerelength. However, in the right-hand pathway shown,

heavy repression of the telomerase gene promoters

by the accumulation of repressive histone

modifications [methylation of Lys20 (K20) H4 and/or methylation of Lys9 (K9) H3] could lead to

utilisation of the ALT mechanism rather than

telomerase activity for telomere elongation/maintenance owing to the repression of telomerase

gene expression.

Epigenetic regulation of telomeremaintenance mechanisms

Expert Reviews in Molecular Medicine

© 2007 Cambridge University Press

Telomeraseactivity

Pre-senescentnormal cell

Normal

Epigeneticmechanisms

Telomeraseactivity

Cancer ALT

Telomerase generepression

 ALT

Permissivemodifications

Repressivemodifications

Telomerase generepression

Telomerase geneexpression

b

a

K20 H4methylation K9 H3methylation

H3 and H4acetylation

K4 H3methylation

expert reviewshttp://www.expertreviews.org/ in molecular medicine

14  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 15/26

the telomerase gene promoters might allow theactivation of a telomere maintenancemechanism, allowing bypass of senescence.

Epigenetic deregulation and

tumourigenicity in adult stem cellsFurther studies into the epigenetic regulation of the telomerase gene promoters utilised humanmesenchymal stem cells (hMSCs) (Ref. 156).These cells do not show telomerase activity buthave been shown to be the target for neoplastictransformation after transduction with hTERT(Ref. 155). However, by supplying hTERT, themolecular events required to upregulate hTERTexpression in cancer stem cell development aremissed. Therefore, hMSCs are ideal for theidentification of molecular mechanismsregulating telomerase gene expression in stem

cells.It has now been shown that repression of 

hTERT expression in hMSCs is a result of promoter-specific histone hypo-acetylationcoupled with low RNA polymerase II (RNAPII)and transcription factor IIB (TFIIB) tracking.This repression can be overcome by treatmentof hMSCs with TSA, concomitant with increasesin promoter-specific histone acetylation andincreases in RNAPII and TFIIB tracking. hTRexpression is also increased in TSA-treatedhMSCs, concomitant with changes in RNAPIIand TFIIB dynamics. These data suggestthat deregulation of chromatin modificationsat the hTERT and hTR promoter mightfacilitate tumourigenesis in hMSCs and supportsthe hypothesis that tumourigenesis can arise inepigenetically deregulated stem cell populations.

Epigenetic regulation of telomere lengthIt has been shown that epigenetic modificationscan also regulate telomere length through themodulation of the chromatin environment of the telomeres themselves. Telomeres are rich inheterochromatin features (Ref. 157) and

disruption of this heterochromatin structure has  been shown to lead to altered telomere lengthin mice, established by knockout of theSUV39H1 and SUV39H2 histone methyltransferases in mice (Ref. 158) and by theknockout of either DNA methyltransferase 1(DNMT1) or both DNMT-3a and -3b in mouseESCs (Ref. 159). In the latter study, the lack of DNMTs led to increased intra-telomericrecombination and increased the levels of other

markers of the ALT telomere maintenancemechanism, further suggesting the role forepigenetics in the choice of telomeremaintenance mechanism used in cancer cells.This also shows that epigenetic deregulation can

lead to immortalisation events and mightfacilitate tumourigenesis.

Therapeutic aspects of chromatin-mediated senescence

The intimate links between senescence andchromatin that have been uncovered mayprovide therapeutic targets for anti-cancertreatment. As induction of prematuresenescence in tumour cells is targeted as ananti-cancer therapy (Refs 4, 5), one therapeuticavenue may be to induce premature cellularsenescence through control of the enzymes that

catalyse epigenetic modifications. Intimateknowledge of gene expression changes insenescence might allow targeted upregulationor downregulation of such genes by epigenetictherapies. Also, epigenetic therapy might allowthe reversal of senescence in certain cell typesassociated with senescence-related disease.However, it must be remembered that cellularsenescence also plays roles in multiple otherdisease states and might potentially nurturetumourigenic growth. Overall, it can beappreciated that this mode of therapy wouldrequire strict control.

Current epigenetic therapies include HDACand DNMT inhibitors (Table 3), which promotere-expression of silenced genes and are widelyused in cancer therapies to facilitate there-expression of tumour suppressor genes.However useful these drugs are, other more-targeted means of enzyme function ormodulating gene expression would beadvantageous. As these drugs are not targetedthey will have pan-genomic effects and couldnurture tumour growth by aberrant activation of growth-promoting genes or cancer-associated

genes, further indicating a requirement for strictcontrol for such epigenetic therapies. The use of therapeutic RNA interference (RNAi) can allowgreater targeting of specific enzymes involved inepigenetic processes, whereas changes in specificgene expression by the use of engineeredtranscription factors is an exciting newmechanism of gene control (Ref. 160). By fusingepigenetic modifiers to engineered transcriptionfactors that can bind to specific gene promoters,

expert reviewshttp://www.expertreviews.org/ in molecular medicine

15  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 16/26

gene expression can be altered through epigeneticmechanisms. However, the ultimate therapeuticaim is to find small molecules that could besuccessfully moved into the clinical arena. Thisapproach has recently been successfully used inone study that screened for compounds thatwere able to modulate cellular senescence(Ref. 161) and could allow further detailedanalysis of the senescence pathway andepigenetic mechanisms important to it.

Epigenetic therapy might also allow the

targeting of cancer stem cells as current cancertherapeutics may not efficiently target such cellsdue to their unique cellular physiology. Recentstudies demonstrating the importance of epigenetic silencers in the maintenance of pluripotency and a proposed role intumourigenesis suggests that it may be possibleto target such cells more efficiently throughtargeting epigenetic silencers. Furthermore,studies have shown that the PcG proteins may

 be regulated by signalling pathways unique topluripotent cells and targeting of thesepathways might allow modulation of epigeneticsilencing and hence pluripotency, and thereforemay have direct relevance to the treatment of cancer stem cells (Refs 162, 163, 164). Molecularsignalling pathways such as PTEN (for‘phosphate and tensin homologue’), Wnt,Hedgehog and Notch have been identified astargets for therapies aimed at cancer stem cellsand therefore targeted gene expression changes

of members of these pathways by epigenetictherapies may allow for selectivity (Ref. 165).However, effects on normal stem cells mightalso become compromised. Studies havesuggested other signalling pathways that couldaffect epigenetic modifications and thereforeprovide suitable therapeutic targets. A recentstudy into the Janus kinases/signal transducersand activators of transcription (JAK/STAT)pathway in Drosophila, a pathway whose

Table 3. Current drugs against epigenetic modifiers

Epigenetic therapy Mode of action

S-adenosylhomocysteine Inhibits S-adenosyl-L-methionine (SAM)-dependent

methyltransferases

5-azacytidine (azacytidine) DNA methyltransferase (DNMT) inhibitor (nucleosideanalogue)

5-aza-20-deoxycytidine (decitabine) DNMT inhibitor (nucleoside analogue)

1-( b-D-ribofuranosyl)-1,2-

dihydropyrimidin-2-one (zebularine)

DNMT inhibitor (nucleoside analogue)

(-)Epigallocatechin-3 gallate (EGCG) DNMT inhibitor (nucleoside analogue)

Procaine DNMT inhibitor (nucleoside analogue)

Procainamide DNMT inhibitor (nucleoside analogue)

Hydralazine DNMT inhibitor (nucleoside analogue)

MG98 DNMT1 inhibitor (second-generation phosphorothioate

antisense oligodeoxynucleotide)

Trichostatin A (TSA) Hydroxamate histone deacetylase (HDAC) inhibitor  

Suberoylanilide hydroxamic acid (SAHA) Hydroxamate HDAC inhibitor 

 m-Carboxycinnamic acid bishydroxamide

(CBHA)

Hydroxamate HDAC inhibitor 

LAQ824 Hydroxamate HDAC inhibitor  

PXD101 Hydroxamate HDAC inhibitor  

Depsipeptide Cyclic peptide HDAC inhibitor  

  Apicidin Cyclic peptide HDAC inhibitor  

Cyclic hydroxamic-acid-containing

peptides (CHAPs)

Cyclic peptide HDAC inhibitor 

  Valproic acid Aliphatic acid HDAC inhibitor  

Phenylbutyrate Aliphatic acid HDAC inhibitor  

MS-275 Benzamide HDAC inhibitor  

CI-994 Benzamide HDAC inhibitor  

Trifluoromethyl ketones Electrophilic ketone HDAC inhibitor  

  Alpha-ketoamides Electrophilic ketone HDAC inhibitor  

expert reviewshttp://www.expertreviews.org/ in molecular medicine

16  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 17/26

deregulation has been implicated in human cancer(Ref. 166), has shown that over-activation of JAKkinase disrupted heterochromatin formation andchanged the expression of genes through STAT-independent routes. Further understanding of 

pathways that control epigenetic modifiers couldrepresent a wealth of opportunity for discovery of therapeutic targets.

Data establishing epigenetic processes asimportant regulators of senescence andtumourigenesis also suggest that such epigeneticmodifications may be useful as prognosticmarkers. Expression of genes involved inepigenetic mechanisms have been shown to beassociated with therapy failure in prostate cancer(Ref. 89) and the presence of certain patterns of global histone modifications have been linked torecurrence of prostate cancer (Ref. 167). Loss of 

lysine acetylation and methylation on histone H4has also been identified as an epigeneticsignature of human cancer (Ref. 39), whereasother studies have identified the loss of repressive lysine methylation and the enzymesthat catalyse the addition of these modificationsas early markers of tumourigenesis (Refs 37, 38).Further levels of methylation status of K27 H3might allow the differentiation between weaklyand strongly malignant tumour cells (Ref. 89).Therefore, analysis and detection of suchmodifications and enzymes could be importantin early cancer detection and might also allow

tailored epigenetic therapies. Furthermore, aschemotherapeutics often promote prematurecellular senescence as an anti-cancer therapy andsenescence-associated chromatin changes arenow understood, such changes might be usedas markers of response to chemotherapeutictherapies (Ref. 168).

Conclusions and summary It is now accepted that epigenetic mechanisms arevital regulators of gene expression and thederegulation of epigenetic mechanisms has been

linked to cancer in multiple studies. Pathwaysand mechanisms important to the senescencephenotype have all been shown to regulate or beregulated by epigenetic mechanisms, underlyingthe importance of this level of control.Epigenetics provides the perfect regulatorymechanism for dynamic changes in globalgene expression required for multiple cellularprocesses such as senescence. Deregulation of epigenetic modifications may therefore promote

tumourigenesis through the disruption of thesenescence phenotype and the deregulation of gene expression.

How does the research now proceed? The roleepigenetics has to play in cellular processes such

as senescence, tumourigenesis and stem cellphenotypes has only recently been uncoveredin any great detail. In addition, epigeneticregulation is key to many other cellularprocesses, whereas deregulation is an importantfactor in numerous disease states. Furtheranalysis of global states of chromatin indifferent cell types and comparisons betweennormal and diseased tissues might allowgreater understanding of the causes of thedisease and also potentially lead to thediscovery of therapeutic targets. Such studieshave already begun, as mentioned, in ESCs

(Refs 85, 86, 87, 88) and also in cancer,including the exciting prospect of the humanepigenome project, an international projectaiming to identify all the chemical changes andrelationships among chromatin constituents thatprovide function to the DNA code (Refs 39, 167,169, 170, 171). Such studies will allow us tounderstand how chromatin modificationsmight control distinct cellular processes andwhat enzymes mediate such changes. Fullunderstanding of the signalling pathways thatmodulate enzymatic control is also animportant step to begin to uncover how the

deregulation of these pathways affects theepigenetic control of gene expression andcellular functions, and also potentially to enablethe discovery of novel therapeutic targets.

 Acknowledgements and fundingResearch in the authors’ laboratory is supported

 by Cancer Research UK, University of Glasgowand European Community grant LSHC-CT-2004-502943. The authors thank the anonymouspeer reviewers for their invaluable comments.

References1 Hayflick, L. (1965) The Limited in Vitro Lifetime

of Human Diploid Cell Strains. Exp Cell Res 37,

614-636

2 Campisi, J. (2001) Cellular senescence as a tumor-

suppressor mechanism. Trends Cell Biol 11, S27-31

3 Mathon, N.F. and Lloyd, A.C. (2001) Cell

senescence and cancer. Nat Rev Cancer 1, 203-213

expert reviewshttp://www.expertreviews.org/ in molecular medicine

17  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 18/26

4 Shay, J.W. and Roninson, I.B. (2004) Hallmarks of 

senescence in carcinogenesis and cancer therapy.

Oncogene 23, 2919-2933

5 Roninson, I.B. (2003) Tumor cell senescence in

cancer treatment. Cancer Res 63, 2705-2715

6 Shelton, D.N. et al. (1999) Microarray analysis of replicative senescence. Curr Biol 9, 939-945

7 Hardy, K. et al. (2005) Transcriptional networks

and cellular senescence in human mammary

fibroblasts. Mol Biol Cell 16, 943-953

8 Yoon, I.K. et al. (2004) Exploration of replicative

senescence-associated genes in human dermal

fibroblasts by cDNA microarray technology. Exp

Gerontol 39, 1369-1378

9 Guo, S., Zhang, Z. and Tong, T. (2004) Cloning and

characterization of cellular senescence-associated

genes in human fibroblasts by suppression

subtractive hybridization. Exp Cell Res 298, 465-472

10 Mason, D.X., Jackson, T.J. and Lin, A.W. (2004)Molecular signature of oncogenic ras-induced

senescence. Oncogene 23, 9238-9246

11 Bandyopadhyay, D. and Medrano, E.E. (2003)

The emerging role of epigenetics in cellular

and organismal aging. Exp Gerontol 38,

1299-1307

12 Young, J. and Smith, J.R. (2000) Epigenetic

aspects of cellular senescence. Exp Gerontol 35,

23-32

13 Esteller, M. (2006) Epigenetics provides a new

generation of oncogenes and tumour-suppressor

genes. Br J Cancer 94, 179-183

14 Santos-Rosa, H. and Caldas, C. (2005) Chromatinmodifier enzymes, the histone code and cancer.

Eur J Cancer 41, 2381-2402

15 Baylin, S.B. and Ohm, J.E. (2006) Epigenetic gene

silencing in cancer - a mechanism for early

oncogenic pathway addiction? Nat Rev Cancer 6,

107-116

16 Jenuwein, T. and Allis, C.D. (2001) Translating the

histone code. Science 293, 1074-1080

17 Strahl, B.D. and Allis, C.D. (2000) The language

of covalent histone modifications. Nature 403,

41-45

18 Martin, C. and Zhang, Y. (2005) The diverse

functions of histone lysine methylation. Nat Rev

Mol Cell Biol 6, 838-849

19 Schubeler, D. et al. (2004) The histone modification

pattern of active genes revealed through genome-

wide chromatin analysis of a higher eukaryote.

Genes Dev 18, 1263-1271

20 Campisi, J. (2005) Senescent cells, tumor

suppression, and organismal aging: good citizens,

 bad neighbors. Cell 120, 513-522

21 Krtolica, A. et al. (2001) Senescent fibroblasts

promote epithelial cell growth and tumorigenesis:

a link between cancer and aging. Proc Natl Acad

Sci U S A 98, 12072-12077

22 Campisi, J. et al. (2001) Cellular senescence, cancer

and aging: the telomere connection. Exp Gerontol36, 1619-1637

23 Campisi, J. (2000) Cancer, aging and cellular

senescence. In Vivo 14, 183-188

24 Jones,P.A.and Baylin,S.B. (2002) Thefundamental

role of epigenetic events in cancer. Nat Rev Genet

3, 415-428

25 Hake, S.B., Xiao, A. and Allis, C.D. (2004) Linking

the epigenetic ‘language’ of covalent histone

modifications to cancer. Br J Cancer 90, 761-769

26 Feinberg, A.P. (2004) The epigenetics of cancer

etiology. Semin Cancer Biol 14, 427-432

27 Feinberg, A.P., Ohlsson, R. and Henikoff, S. (2006)

The epigenetic progenitor origin of human cancer.Nat Rev Genet 7, 21-33

28 Macieira-Coelho, A. (1991) Chromatin

reorganization during senescence of proliferating

cells. Mutat Res 256, 81-104

29 Zhang, H., Pan, K.H. and Cohen, S.N. (2003)

Senescence-specific gene expression fingerprints

reveal cell-type-dependent physical clustering of 

up-regulated chromosomalloci. ProcNatl Acad Sci

U S A 100, 3251-3256

30 Narita, M. et al. (2003) Rb-mediated

heterochromatin formation and silencing of 

E2F target genes during cellular senescence.

Cell 113, 703-71631 Luo, R.X., Postigo, A.A. and Dean, D.C. (1998) Rb

interacts with histone deacetylase to repress

transcription. Cell 92, 463-473

32 Magnaghi-Jaulin, L. et al. (1998) Retinoblastoma

protein represses transcription by recruiting

a histone deacetylase. Nature 391, 601-605

33 Brehm, A. et al. (1998) Retinoblastoma protein

recruits histone deacetylase to repress

transcription. Nature 391, 597-601

34 Vandel, L. et al. (2001) Transcriptional repression

 by the retinoblastoma protein through the

recruitment of a histone methyltransferase. Mol

Cell Biol 21, 6484-6494

35 Nielsen, S.J. et al. (2001) Rb targets histone H3

methylation and HP1 to promoters. Nature 412,

561-565

36 Braig, M. et al. (2005) Oncogene-induced

senescence as an initial barrier in lymphoma

development. Nature 436, 660-665

37 Peters,A.H. et al.(2001)Loss of theSuv39 h histone

methyltransferases impairs mammalian

expert reviewshttp://www.expertreviews.org/ in molecular medicine

18  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 19/26

heterochromatin and genome stability. Cell

107, 323-337

38 Pogribny, I.P. et al. (2006) Histone H3 lysine 9 and

H4 lysine 20 trimethylation and the expression of 

Suv4–20h2 and Suv-39h1 histone

methyltransferases in hepatocarcinogenesisinduced by methyl deficiency in rats.

Carcinogenesis 27, 1180-1186

39 Fraga, M.F. et al. (2005) Loss of acetylation at

Lys16 and trimethylation at Lys20 of histone H4

is a common hallmark of human cancer. Nat

Genet 37, 391-400

40 Michaloglou, C. et al. (2005) BRAFE600-associated

senescence-like cell cycle arrest of human naevi.

Nature 436, 720-724

41 Collado, M. et al. (2005) Tumour biology:

senescence in premalignant tumours. Nature

436, 642

42 Lazzerini Denchi, E. et al. (2005) Deregulated E2Factivity induces hyperplasia and senescence-like

features in themouse pituitary gland.Mol Cell Biol

25, 2660-2672

43 Chan, H.M. et al. (2005) The p400 E1A-associated

protein is a novel component of the p53 ! p21

senescence pathway. Genes Dev 19, 196-201

44 Zhang, R. et al. (2005) Formation of MacroH2A-

containing senescence-associated heterochromatin

foci and senescence driven by ASF1a and HIRA.

Dev Cell 8, 19-30

45 Narita, M. et al. (2006) A novel role for

high-mobility group a proteins in cellular

senescence and heterochromatin formation. Cell126, 503-514

46 Liu, X.T. et al. (1994) Prohibitin expression during

cellular senescence of human diploid fibroblasts.

Biochem Biophys Res Commun 201, 409-414

47 Rastogi, S. et al. (2006) Prohibitin facilitates

cellular senescence by recruiting specific

corepressors to inhibit E2F target genes.

Mol Cell Biol 26, 4161-4171

48 Herbig, U. et al. (2006) Cellular senescence in aging

primates. Science 311, 1257

49 Allison, S.J. and Milner, J. (2004) Remodelling

chromatin on a global scale: a novel protective

function of p53. Carcinogenesis 25, 1551-1557

50 Espinosa, J.M. and Emerson, B.M. (2001)

Transcriptional regulation by p53 through intrinsic

DNA/chromatin binding and site-directed

cofactor recruitment. Mol Cell 8, 57-69

51 Barlev, N.A. et al. (2001) Acetylation of p53

activates transcription through recruitment of 

coactivators/histone acetyltransferases. Mol Cell

8, 1243-1254

52 Lagger, G. et al. (2003) The tumor suppressor p53

and histone deacetylase 1 are antagonistic

regulators of the cyclin-dependent kinaseinhibitor

p21/WAF1/CIP1 gene. Mol Cell Biol 23, 2669-2679

53 An, W., Kim, J. and Roeder, R.G. (2004) Ordered

cooperative functions of PRMT1, p300, andCARM1 in transcriptional activation by p53. Cell

117, 735-748

54 Murphy, M. et al. (1999) Transcriptional repression

 by wild-type p53 utilizes histone deacetylases,

mediated by interaction with mSin3a. Genes Dev

13, 2490-2501

55 Nguyen, T.T. et al. (2005) Transcription factor

interactions and chromatin modifications

associated with p53-mediated, developmental

repression of the alpha-fetoprotein gene. Mol Cell

Biol 25, 2147-2157

56 Cui, R. et al. (2005) Family members p53 and p73

act together in chromatin modification and directrepressionof alpha-fetoprotein transcription. J Biol

Chem 280, 39152-39160

57 Russell, M. et al. (2006) Grow-ING, Age-ING and

Die-ING: ING proteins linkcancer, senescence and

apoptosis. Exp Cell Res 312, 951-961

58 Feng, X.,Hara, Y. and Riabowol, K. (2002) Different

HATS of the ING1 gene family. Trends Cell Biol

12, 532-538

59 Vieyra, D. et al. (2002) Human ING1 proteins

differentially regulate histone acetylation. J Biol

Chem 277, 29832-29839

60 Vieyra, D. et al. (2002) ING1 isoforms differentially

affect apoptosis in a cell age-dependent manner.Cancer Res 62, 4445-4452

61 Shi, X.et al. (2006) ING2 PHDdomain links histone

H3 lysine 4 methylation to active gene repression.

Nature 442, 96-99

62 Yamane, K. et al. (2006) JHDM2A, a JmjC-

containing H3K9 demethylase, facilitates

transcription activation by androgen receptor. Cell

125, 483-495

63 Whetstine, J.R. et al. (2006) Reversal of histone

lysine trimethylation by the JMJD2 family of 

histone demethylases. Cell 125, 467-481

64 Klose,R.J. et al.(2006) Thetranscriptionalrepressor

 JHDM3A demethylates trimethyl histone H3

lysine 9 and lysine 36. Nature 442, 312-316

65 Cloos, P.A. et al. (2006) The putative oncogene

GASC1 demethylates tri- and dimethylated lysine

9 on histone H3. Nature 442, 307-311

66 Metzger, E. et al. (2005) LSD1 demethylates

repressive histone marks to promote androgen-

receptor-dependent transcription. Nature 437,

436-439

expert reviewshttp://www.expertreviews.org/ in molecular medicine

19  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 20/26

67 Jacobs, J.J. et al. (1999) The oncogene and

Polycomb-group gene bmi-1 regulates cell

proliferation and senescence through the ink4a

locus. Nature 397, 164-168

68 Itahana, K. et al. (2003) Control of the replicative

life span of human fibroblasts by p16 and thepolycomb protein Bmi-1. Mol Cell Biol 23, 389-401

69 Bea, S. et al. (2001) BMI-1 gene amplification and

overexpression in hematological malignancies

occur mainly in mantle cell lymphomas. Cancer

Res 61, 2409-2412

70 Vonlanthen, S. et al. (2001) The bmi-1 oncoprotein

is differentially expressed in non-small cell lung

cancer and correlates with INK4A-ARF locus

expression. Br J Cancer 84, 1372-1376

71 Nowak, K. et al. (2006) BMI1 is a target gene of 

E2F-1 and is strongly expressed in primary

neuroblastomas. Nucleic Acids Res 34, 1745-1754

72 Sawa, M. et al. (2005) BMI-1 is highly expressed inM0-subtype acutemyeloid leukemia. Int J Hematol

82, 42-47

73 Liu, L., Andrews, L.G. and Tollefsbol, T.O. (2006)

Loss of the human polycomb group protein BMI1

promotes cancer-specific cell death. Oncogene 25,

4370-4375

74 Leung, C. et al. (2004) Bmi1 is essential for

cerebellar development and is overexpressed in

human medulloblastomas. Nature 428, 337-341

75 Dimri, G.P. et al. (2002) The Bmi-1 oncogene

induces telomerase activity and immortalizes

human mammary epithelial cells. Cancer Res 62,

4736-474576 Cao, R. and Zhang, Y. (2004) The functions of E(Z)/

EZH2-mediated methylationof lysine27 in histone

H3. Curr Opin Genet Dev 14, 155-164

77 Varambally, S. et al. (2002) The polycomb group

protein EZH2is involved in progressionof prostate

cancer. Nature 419, 624-629

78 Kleer, C.G. et al. (2003) EZH2 is a marker of 

aggressive breast cancer and promotes neoplastic

transformation of breast epithelial cells. Proc Natl

Acad Sci U S A 100, 11606-11611

79 Bracken, A.P. et al. (2003) EZH2 is downstream

of the pRB-E2F pathway, essential for proliferation

and amplified in cancer. Embo J 22, 5323-5335

80 Tang, X. et al. (2004) Activated p53 suppresses the

histone methyltransferase EZH2 gene. Oncogene

23, 5759-5769

81 Villeponteau, B. (1997) The heterochromatin loss

model of aging. Exp Gerontol 32, 383-394

82 Meshorer, E. et al. (2006) Hyperdynamic plasticity

of chromatin proteins in pluripotent embryonic

stem cells. Dev Cell 10, 105-116

83 Meshorer, E. and Misteli, T. (2006) Chromatin in

pluripotent embryonic stem cells and

differentiation. Nat Rev Mol Cell Biol 7, 540-546

84 Szutorisz, H. and Dillon, N. (2005) The epigenetic

 basis for embryonic stem cell pluripotency.

Bioessays 27, 1286-129385 Boyer, L.A. et al. (2006) Polycomb complexes

repress developmental regulators in murine

embryonic stem cells. Nature 441, 349-353

86 Bernstein, B.E. et al. (2006) A bivalent chromatin

structure marks key developmental genes in

embryonic stem cells. Cell 125, 315-326

87 Azuara, V. et al. (2006) Chromatin signatures of 

pluripotent cell lines. Nat Cell Biol 8, 532-538

88 Lee, T.I. et al. (2006) Control of developmental

regulators by Polycomb in human embryonic stem

cells. Cell 125, 301-313

89 Berezovska, O.P. et al. (2006) Essential role for

activation of the polycomb group (PcG) proteinchromatinsilencing pathway in metastatic prostate

cancer. Cell Cycle 5, 1886-1901

90 Glinsky, G.V., Berezovska, O. and Glinskii, A.B.

(2005) Microarray analysis identifies a death-from-

cancer signature predicting therapy failure in

patients withmultiple types of cancer. J Clin Invest

115, 1503-1521

91 Krivtsov, A.V. et al. (2006) Transformation from

committed progenitor to leukaemia stem cell

initiated by MLL-AF9. Nature 442, 818-822

92 Daser, A. and Rabbitts, T.H. (2005) The versatile

mixed lineage leukaemia gene MLL and its many

associations in leukaemogenesis. Semin CancerBiol 15, 175-188

93 Anonymous (2006) European Journal of Cancer

Special Issue on Cancer Stem Cells: Opportunities

for Novel Diagnostics and Drug Discovery. 42,

1298-1308

94 Bandyopadhyay, D. et al. (2002) Down-regulation

of p300/CBP histone acetyltransferase activates a

senescence checkpoint in human melanocytes.

Cancer Res 62, 6231-6239

95 Yao, T.P. et al. (1998) Gene dosage-dependent

embryonic development and proliferation defects

in mice lacking the transcriptional integrator

p300. Cell 93, 361-372

96 Ogryzko, V.V. et al. (1996) Human fibroblast

commitment to a senescence-like state in response

to histone deacetylase inhibitors is cell cycle

dependent. Mol Cell Biol 16, 5210-5218

97 Munro, J. et al. (2004) Histone deacetylase inhibitors

induce a senescence-like state in human cells by a

p16-dependent mechanism that is independent of a

mitotic clock. Exp Cell Res 295, 525-538

expert reviewshttp://www.expertreviews.org/ in molecular medicine

20  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 21/26

98 Place, R.F., Noonan, E.J. and Giardina, C. (2005)

HDACs and the senescent phenotype of WI-38

cells. BMC Cell Biol 6, 37

99 Kaeberlein, M., McVey, M. and Guarente, L.

(1999) The SIR2/3/4 complex and SIR2 alone

promote longevity in Saccharomyces cerevisiae bytwo different mechanisms. Genes Dev 13,

2570-2580

100 Imai, S. et al. (2000) Transcriptional silencing and

longevity protein Sir2 is an NAD-dependent

histone deacetylase. Nature 403, 795-800

101 Hekimi, S. and Guarente, L. (2003) Genetics

and the specificity of the aging process. Science

299, 1351-1354

102 Rogina, B. and Helfand, S.L. (2004) Sir2 mediates

longevity in the fly through a pathway related to

calorie restriction. Proc Natl Acad Sci U S A 101,

15998-16003

103 Cohen, H.Y. et al. (2004) Calorie restrictionpromotes mammalian cell survivalby inducing the

SIRT1 deacetylase. Science 305, 390-392

104 Furuyama, T. et al. (2004) SIR2 is required for

polycomb silencing and is associated with an E(Z)

histone methyltransferase complex. Curr Biol 14,

1812-1821

105 Yeung, F. et al. (2004) Modulation of NF-

kappaB-dependent transcription and cell

survival by the SIRT1 deacetylase. Embo J 23,

2369-2380

106 Brunet, A. et al. (2004) Stress-dependent regulation

of FOXO transcription factors by the SIRT1

deacetylase. Science 303, 2011-2015107 Vaziri, H. et al. (2001) hSIR2(SIRT1) functions as

an NAD-dependent p53 deacetylase. Cell 107,

149-159

108 Bradbury, C.A. et al. (2005) Histone deacetylases in

acute myeloid leukaemia show a distinctive

pattern of expression that changes selectively in

response to deacetylase inhibitors. Leukemia 19,

1751-1759

109 Michishita, E. et al. (2005) Evolutionarily

conserved and nonconserved cellular localizations

and functions of human SIRT proteins. Mol Biol

Cell 16, 4623-4635

110 Ota, H. et al. (2005) Sirt1 inhibitor, Sirtinol, induces

senescence-like growth arrestwith attenuated Ras-

MAPK signaling in human cancer cells. Oncogene

25, 176-185

111 Stancheva, I. (2005) Caught in conspiracy:

cooperation between DNA methylation and

histone H3K9 methylation in the establishment

and maintenance of heterochromatin. Biochem

Cell Biol 83, 385-395

112 Lachner, M. and Jenuwein, T. (2002) The many

faces of histone lysine methylation. Curr Opin Cell

Biol 14, 286-298

113 Vire, E. et al. (2006) The Polycomb group protein

EZH2 directly controls DNA methylation. Nature

439, 871-874114 Esteve, P.O. et al. (2006) Direct interaction between

DNMT1 and G9a coordinates DNA and histone

methylation during replication. Genes Dev 20,

3089-3103

115 Wilson, V.L. and Jones, P.A. (1983) DNA

methylation decreasesin aging but notin immortal

cells. Science 220, 1055-1057

116 Hoal-van Helden,E.G. and vanHelden, P.D. (1989)

Age-related methylation changes in DNA may

reflect the proliferative potential of organs. Mutat

Res 219, 263-266

117 Holliday, R. (1986) Strong effects of 5-azacytidine

on the in vitro lifespan of human diploidfibroblasts. Exp Cell Res 166, 543-552

118 Fairweather, D.S., Fox, M. and Margison, G.P.

(1987) The in vitro lifespan of MRC-5 cells is

shortened by 5-azacytidine-induced

demethylation. Exp Cell Res 168, 153-159

119 Belinsky, S.A. et al.(1996) Increasedcytosine DNA-

methyltransferase activity is target-cell-specific

and an early event in lung cancer. Proc Natl Acad

Sci U S A 93, 4045-4050

120 Slack, A. et al. (1999) DNA methyltransferase is a

downstream effector of cellular transformation

triggered by simian virus 40 large Tantigen. J Biol

Chem 274, 10105-10112121 Rouleau, J., MacLeod, A.R. and Szyf, M. (1995)

Regulation of the DNA methyltransferase by the

Ras-AP-1 signaling pathway. J Biol Chem 270,

1595-1601

122 Issa, J.P. et al. (1994) Methylation of the oestrogen

receptor CpG island links ageing and neoplasia in

human colon. Nat Genet 7, 536-540

123 Young, J.I. and Smith, J.R. (2001) DNA

methyltransferase inhibition in normal human

fibroblasts induces a p21-dependent cell cycle

withdrawal. J Biol Chem 276, 19610-19616

124 Feinberg, A.P. and Vogelstein, B. (1983)

Hypomethylation distinguishes genes of some

human cancers from their normal counterparts.

Nature 301, 89-92

125 Goelz, S.E. et al. (1985) Hypomethylation of DNA

from benign and malignant human colon

neoplasms. Science 228, 187-190

126 Eden, A. et al. (2003) Chromosomal instability and

tumors promoted by DNA hypomethylation.

Science 300, 455

expert reviewshttp://www.expertreviews.org/ in molecular medicine

21  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 22/26

127 Gaudet, F. et al. (2003) Induction of tumors in

mice by genomic hypomethylation. Science 300,

489-492

128 Sakai, T. et al. (1991) Allele-specific

hypermethylation of the retinoblastoma tumor-

suppressor gene. Am J Hum Genet 48, 880-888129 Gonzalez-Zulueta, M. et al. (1995) Methylation of 

the 50

CpG island of the p16/CDKN2 tumor

suppressor gene in normal and transformed

human tissues correlates with gene silencing.

Cancer Res 55, 4531-4535

130 Reynolds, P.A. et al. (2006) Tumor suppressor

p16INK4A regulates polycomb-mediated DNA

hypermethylation in human mammary epithelial

cells. J Biol Chem 281, 24790-24802

131 Herskind, C. and Rodemann, H.P. (2000)

Spontaneous and radiation-induced differentiation

of fibroblasts. Exp Gerontol 35, 747-755

132 Robles, S.J. and Adami, G.R. (1998) Agents thatcause DNA double strand breaks lead to

p16INK4a enrichment and the premature

senescence of normal fibroblasts. Oncogene 16,

1113-1123

133 von Zglinicki, T. et al. (1995) Mild hyperoxia

shortens telomeres and inhibits proliferation of 

fibroblasts: a model for senescence? Exp Cell Res

220, 186-193

134 Shroff, R. et al. (2004) Distribution and

dynamics of chromatin modification induced by a

defined DNA double-strand break. Curr Biol 14,

1703-1711

135 Karlseder, J., Smogorzewska, A. and de Lange, T.(2002) Senescence induced by altered telomere

state, not telomere loss. Science 295, 2446-2449

136 d’Adda di Fagagna, F. et al.(2003) A DNA damage

checkpoint response in telomere-initiated

senescence. Nature 426, 194-198

137 van Attikum, H. and Gasser, S.M. (2005) The

histone code at DNA breaks: a guide to repair?

Nat Rev Mol Cell Biol 6, 757-765

138 Huyen, Y. et al. (2004) Methylated lysine 79 of 

histone H3 targets 53BP1 to DNA double-strand

 breaks. Nature 432, 406-411

139 Sanders, S.L. et al.(2004)Methylationof histone H4

lysine 20 controls recruitment of Crb2 to sites of 

DNA damage. Cell 119, 603-614

140 Botuyan, M.V. et al. (2006) Structural basis for the

methylation state-specific recognition of histone

H4-K20by53BP1andCrb2inDNArepair.Cell127,

1361-1373

141 Shay, J.W. and Bacchetti, S. (1997) A survey of 

telomerase activity in human cancer. Eur J Cancer

33, 787-791

142 Harley, C.B.,Futcher, A.B. and Greider, C.W. (1990)

Telomeres shorten during ageing of human

fibroblasts. Nature 345, 458-460

143 Hastie, N.D. et al. (1990) Telomere reduction in

human colorectal carcinoma and with ageing.

Nature 346, 866-868144 Allsopp, R.C. et al. (1992) Telomere length predicts

replicative capacity of humanfibroblasts. ProcNatl

Acad Sci U S A 89, 10114-10118

145 Kim, N.W. et al. (1994) Specific association of 

human telomerase activity withimmortalcells and

cancer. Science 266, 2011-2015

146 Reddel, R.R. (2003) Alternative lengthening of 

telomeres, telomerase, and cancer. Cancer Lett

194, 155-162

147 Dunham, M.A. et al. (2000) Telomere maintenance

 by recombination in human cells. Nat Genet 26,

447-450

148 Bryan, T.M. and Reddel, R.R. (1997) Telomeredynamics and telomerase activity in in vitro

immortalised human cells.Eur J Cancer33, 767-773

149 Yeager, T.R. et al. (1999) Telomerase-negative

immortalized human cells contain a novel type of 

promyelocytic leukemia (PML) body. Cancer Res

59, 4175-4179

150 Henson, J.D. et al. (2005) A robust assay for

alternative lengthening of telomeres in tumors

shows the significance of alternative lengthening

of telomeres in sarcomas and astrocytomas. Clin

Cancer Res 11, 217-225

151 Ulaner, G.A. et al. (2003) Absence of a telomere

maintenance mechanism as a favorable prognosticfactor in patients with osteosarcoma. Cancer Res

63, 1759-1763

152 Hakin-Smith, V. et al. (2003) Alternative

lengthening of telomeres and survival in

patients with glioblastomamultiforme.Lancet 361,

836-838

153 Johnson, J.E. et al. (2005) Multiple mechanisms of 

telomere maintenance exist in liposarcomas. Clin

Cancer Res 11, 5347-5355

154 Atkinson, S.P. et al. (2005) Lack of telomerase gene

expression in alternative lengthening of telomere

cells is associated with chromatin remodeling of 

the hTR and hTERT gene promoters. Cancer Res

65, 7585-7590

155 Serakinci, N. et al. (2006) Telomerase promoter

reprogramming and interaction with general

transcription factors in the human mesenchymal

stem cell. Regenerative Medicine 1, 125-131

156 Serakinci, N. et al. (2004) Adult human

mesenchymal stem cell as a target for neoplastic

transformation. Oncogene 23, 5095-5098

expert reviewshttp://www.expertreviews.org/ in molecular medicine

22  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 23/26

157 Blasco, M.A. (2004) Carcinogenesis Young

Investigator Award. Telomere epigenetics: a

higher-order control of telomere length in

mammalian cells. Carcinogenesis 25, 1083-1087

158 Garcia-Cao, M. et al. (2004) Epigenetic regulation

of telomere length in mammalian cells by theSuv39h1 and Suv39h2 histone methyltransferases.

Nat Genet 36, 94-99

159 Gonzalo, S. et al. (2006) DNA methyltransferases

control telomerelength andtelomererecombinationin

mammalian cells. Nat Cell Biol 8, 416-424

160 Papworth, M., Kolasinska, P. and Minczuk, M.

(2006) Designer zinc-finger proteins and their

applications. Gene 366, 27-38

161 Won, J. et al. (2006) Small molecule-based

reversible reprogramming of cellular lifespan.

Nat Chem Biol 2, 369-374

162 Galmozzi,E., Facchetti,F.and La Porta, C.A. (2006)

Cancer stem cells and therapeutic perspectives.Curr Med Chem 13, 603-607

163 Fuchs, E., Tumbar, T. and Guasch, G. (2004)

Socializing withthe neighbors: stem cells and their

niche. Cell 116, 769-778

164 Zheng, X. et al. (2004) Gamma-catenin contributes

to leukemogenesis induced by AML-associated

translocation products by increasing the

self-renewal of very primitive progenitor cells.

Blood 103, 3535-3543

165 Massard, C., Deutsch, E. and Soria, J.C. (2006)

Tumour stem cell-targeted treatment: elimination

or differentiation. Ann Oncol 17, 1620-1624

166 Shi, S. et al. (2006) JAK signaling globallycounteracts heterochromatic gene silencing. Nat

Genet 38, 1071-1076

167 Seligson, D.B. et al. (2005) Global histone

modification patterns predict risk of prostate

cancer recurrence. Nature 435, 1262-1266

168 Collado, M. and Serrano, M. (2006) The power and

the promise of oncogene-induced senescence

markers. Nat Rev Cancer 6, 472-476

169 Fraga, M.F. and Esteller, M. (2005) Towards the

human cancer epigenome: a first draft of histone

modifications. Cell Cycle 4, 1377-1381

170 Esteller, M. (2006) The necessity of a human

epigenome project. Carcinogenesis 27, 1121-1125

171 Jones, P.A. and Martienssen, R. (2005) A blueprint

for a Human Epigenome Project: the AACR

Human Epigenome Workshop. Cancer Res 65,

11241-11246

172 Matthews, C. et al. (2006) Vascular smooth muscle

cells undergo telomere-based senescence in

human atherosclerosis: effects of telomerase and

oxidative stress. Circ Res 99, 156-164

173 Henderson, E.A. (2006) The potential effect of 

fibroblast senescence on wound healing and the

chronic wound environment. J Wound Care 15,

315-318

174 Streit, W.J. (2006) Microglial senescence: does the

 brain’s immune system have an expiration date?Trends Neurosci 29, 506-510

175 Shimada, A. et al. (2002) Age-related progressive

neuronal DNA damage associated with cerebral

degeneration in a mouse model of accelerated

senescence. J Gerontol A Biol Sci Med Sci 57,

B415–421

176 Flanary, B. (2005) The role of microglial cellular

senescence in the aging and Alzheimer diseased

 brain. Rejuvenation Res 8, 82-85

177 Nyunoya, T. et al. (2006) Cigarette smoke induces

cellular senescence. Am J Respir Cell Mol Biol

35, 681-688

178 Muller, K.C. et al. (2006) Lung fibroblasts frompatients with emphysema show markers of 

senescence in vitro. Respir Res 7, 32

179 Jimenez, R. et al. (2005) Replicative senescence in

patients with chronic kidney failure. Kidney Int

Suppl S11– 15

180 Erusalimsky, J.D. and Kurz, D.J. (2005) Cellular

senescence in vivo: its relevance in ageing and

cardiovascular disease. Exp Gerontol 40, 634-642

181 van Baarle, D. et al. (2005) Significance of senescence

for virus-specific memory Tcell responses: rapid

ageing during chronic stimulation of the immune

system. Immunol Lett 97, 19-29

182 Vallejo, A.N., Weyand, C.M. and Goronzy, J.J.(2004) T-cell senescence: a culprit of immune

abnormalities in chronic inflammation and

persistent infection. Trends Mol Med 10, 119-124

183 Joosten, S.A. et al. (2003) Telomere shortening

and cellular senescence in a model of chronic

renal allograft rejection. Am J Pathol 162,

1305-1312

184 Martin, J.A. and Buckwalter, J.A. (2002) Aging,

articular cartilage chondrocyte senescence and

osteoarthritis. Biogerontology 3, 257-264

185 Wiemann, S.U. et al. (2002) Hepatocyte telomere

shortening and senescence are general markers of 

human liver cirrhosis. Faseb J 16, 935-942

186 Lu, Q. et al. (2006) Epigenetics, disease, and

therapeutic interventions. Ageing Res Rev

187 Moretti, P. and Zoghbi, H.Y. (2006) MeCP2

dysfunction in Rett syndrome and related

disorders. Curr Opin Genet Dev 16, 276-281

188 Ballestar, E., Esteller, M. and Richardson, B.C.

(2006) The epigenetic face of systemic lupus

erythematosus. J Immunol 176, 7143-7147

expert reviewshttp://www.expertreviews.org/ in molecular medicine

23  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 24/26

189 McKinsey, T.A. and Olson, E.N. (2005) Toward

transcriptional therapies for the failing heart:

chemical screens to modulate genes. J Clin Invest

115, 538-546

190 Sadri-Vakili, G. and Cha, J.H. (2006) Mechanisms

of disease: Histone modifications in Huntington’sdisease. Nat Clin Pract Neurol 2, 330-338

191 Beglopoulos, V. and Shen, J. (2006) Regulation

of CRE-dependent transcription by

presenilins: prospects for therapy of Alzheimer’s

disease. Trends Pharmacol Sci 27, 33-40

192 Langley, B. et al. (2005) Remodeling chromatin

and stress resistance in the central nervous

system: histone deacetylase inhibitors as

novel and broadly effective neuroprotective

agents. Curr Drug Targets CNS Neurol Disord 4,

41-50

193 Taniura, H., Sng, J.C. and Yoneda, Y. (2006)

Histone modifications in status epilepticus

induced by kainate. Histol Histopathol 21,

785-791

194 Adcock, I.M. and Lee, K.Y. (2006) Abnormal

histone acetylase and deacetylase expression andfunction in lung inflammation. Inflamm Res 55,

311-321

195 Hirtz, D. et al. (2005) Challenges and opportunities

in clinical trials for spinal muscular atrophy.

Neurology 65, 1352-1357

196 Bredy, T.W. (2007) Behavioural epigenetics and

psychiatric disorders. Med Hypotheses 68, 453

197 Herman, J.G. and Baylin, S.B. (2003) Gene

silencing in cancer in association with

promoter hypermethylation. N Engl J Med 349,

2042-2054

expert reviewshttp://www.expertreviews.org/ in molecular medicine

24  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 25/26

Further reading, resources and contacts

The Cancer Research UK Laboratories in Glasgow, UK, houses nearly 20 research groups working towards

understanding cancer growth and its treatment:

http://www.beatson.gla.ac.uk /

 Antibody and protocol resources

Both Upstate and Abcam are providers of ChIP-grade antibodies and are widely used in the chromatin field

http://www.abcam.com

http://www.upstate.com

Upstate also hosts a new informational resource that details significant developments within the field of 

chromatin research:

http://www.histone.com

Chromatin resources

The Chromatin Structure and Function website has excellent links to researchers, journals, search tools and

other related topics. It also includes a mailing list for recent chromatin papers from all the major journals:

http://www.chromatin.us/chrom.html

The Human Epigenome Project homepage:

http://www.epigenome.org/

Chromatin.com keeps up to date with allthe recent high-impact journal articles in thefield of chromatin andalso

with chromatin-related meetings:

http://www.chromatin.com

Other useful resources

The biopharmaceutical company Geron is involved in drug discovery for regenerative medicine and oncology:

http://www.geron.com/

 Various laboratory homepages with excellent links on senescence, telomerase and telomeres, further resources

and journal articles:

Jerry Shay’s and Woodring Wright’s laboratory homepage:

http://www4.utsouthwestern.edu/cellbio/shay-wright/index.html

Igor Roninson’s laboratory homepage

http://www.ordwayresearch.org/profile_roninson.html

Judith Campisi’s laboratory homepage

http://www.lbl.gov/lifesciences/CMB/Campisi.html

expert reviewshttp://www.expertreviews.org/ in molecular medicine

25  Accession information: DOI: 10.1017/S1462399407000269; Vol. 9; Issue 7; March 2007

&2007 Cambridge University Press

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n

8/3/2019 Senescence in Disease

http://slidepdf.com/reader/full/senescence-in-disease 26/26

Features associated with this article

Figures

Figure 1. Routes to immortality.

Figure 2. The formation of senescence-associated heterochromatin foci (SAHF) in response to oncogenic insult.

Figure 3. The role of senescence-associated heterochromatin foci (SAHF) in senescence and tumourigenesis.Figure 4. Attainment of pluripotency in normal cells: a possible path towards the cancer stem cell.

Figure 5. Epigenetic regulation of telomere maintenance mechanisms.

Tables

Table 1. Senescence and disease.

Table 2. Chromatin and disease.

Table 3. Current drugs against epigenetic modifiers.

Citation details for this article

Stuart P. Atkinson and W. Nicol Keith (2007) Epigenetic control of cellular senescence in disease: opportunities

for therapeutic intervention. Expert Rev. Mol. Med. Vol. 9, Issue 7, March 2007, DOI: 10.1017/ S1462399407000269

expert reviewshttp://www.expertreviews.org/ in molecular medicine

26

      E     p      i     g     e     n     e      t      i     c

     c     o     n      t     r     o      l     o      f

     c     e      l      l     u      l     a     r     s     e     n     e     s     c     e     n     c

     e

      i     n

      d      i     s     e     a     s     e     :     o     p     p     o

     r      t     u     n      i      t      i     e     s

      f     o     r      t      h     e     r     a     p     e     u      t      i     c

      i     n      t     e

     r     v     e     n      t      i     o     n