How stem cells age and why

download How stem cells age and why

of 11

Transcript of How stem cells age and why

  • 8/7/2019 How stem cells age and why

    1/11

    Dirs acqird and gntic factors dri th com-plx clllar and organismal procss of mammalianaging. Th procss appars to hastnd y ni-ronmntal and haioral factors inclding osity,diats, nd-stag rnal disas and xposr tomtagns sch as chmothrapy, ltraiolt light andtoacco smok. Likwis, arios gntic lsions thataltr DNA mtaolism and nclar architctr han associatd with progroid syndroms in hmanssch as ataxia tlangictasia, Wrnr syndromand HtchinsonGilford syndrom1. Most comp-lling ha n gntic stdis in yast, Drosophilamelanogaster, Caenorhabditis elegans and mic, whichha stalishd crcial rols for th inslininslin-lik growth factor-1 (IGF1)AKTforkhad trancriptionfactor cla O (FOXO) pathway (s th Riw yRssll and Kahn in this iss) and oxygn-radicalrglators (s th Opinion articl y Plicci andcollags in this iss) as dtrminants of lifspan

    in ths modl systms. Althogh ths disparatxprimntal osrations sggst common molclarthms, hman aging in physiological trms rmainsan nigma.

    In this Riw, w sggst that som aspcts ofmammalian aging rslt from an ag-associatddclin in th rplicati fnction of crtain tiss-spcific rgnrati clls adlt stm clls (FIG. 1).Ths rar and spcializd clls ar rqird fortiss rplacmnt throghot th hman lifspan,and appar to charactrizd y a fw spcificphysiological and iochmical proprtis (riwdin ReFs 24), particlarly th capacity for lf-rnwal.

    Rcnt idnc spports th modl that stm cllsin sral tisss ar largly rtaind in a qiscnt statt can coaxd ack into th cll cycl in rsponsto xtraclllar cs, n aftr prolongd priodsof dormancy. Onc stimlatd to diid, stm cllsyild ndiffrntiatd progny, which in trn prodcdiffrntiatd ffctor clls throgh ssqnt rondsof prolifration (BOX 1). This hirarchical diffrntia-tion schm maks sns from th prspcti of organ-ismal longity it prmits th prodction of largnmrs of diffrntiatd clls from a singl stm clly comining ssqnt stps in diffrntiation withprolifration5,6. Thrfor, this approach alancs thhigh rats of homostatic prolifration that ar rqirdin tisss lik th on marrow and intstin with thlong-trm nd to protct stm clls from mtagnicinslt and carcinognsis. Indd, ndr homostaticconditions, thr is limitd prolifrati dmand onth slf-rnwing stm clls thmsls and so ths

    clls diid infrqntly, sparing stm clls th prilsof DNA rplication and mitosis. Additionally, as stmclls appar to lss mtaolically acti in thir qi-scnt stat, thy may sjctd to lowr lls ofDNA-damag-indcing mtaolic sid prodcts schas racti oxygn spcis (ROS)7.

    Onc lid to rstrictd only to high-trnororgans lik th on marrow and intstin, rsidnt slf-rnwing clls ar now thoght to ha a significant rolin th homostatic maintnanc of many organs, incld-ing organs with lowr trnor rats sch as th rainand pancratic islts8. Althogh thr is disagrmntas to th significanc of sch clls in ths low-trnor

    *Departments of Medicine

    and Genetics, The Lineberger

    Comprehensive Cancer Center,

    The University of North

    Carolina, Chapel Hill, NorthCarolina 275997295, USA.Center for Applied Cancer

    Science of the Belfer Institute

    for Innovative Cancer Science,

    and Department of Medical

    Oncology, DanaFarber

    Cancer Institute; Departments

    of Medicine and Genetics,

    Harvard Medical School,

    Boston, Massachusetts

    02115, USA.

    emails:

    [email protected].

    edu; [email protected]

    doi:10.1038/nrm2241

    Forkhead transcription

    factor class O

    (FOXO). On of a family of

    volutionarily conrvd

    trancription factor that ar

    linkd to lifpan rgulation in

    lowr ytm and to tm-cllmaintnanc in mic. Th

    FOXO protin ar thought to

    xrt th ffct by

    rgulating th xprion of

    gn involvd in apoptoi,

    prolifration, gluco

    mtabolim, DNA rpair,

    rgulation of ractiv oxygn

    pci and othr divr

    cllular proc.

    How stem cells age and whythis makes us grow oldNorman E. Sharpless* and Ronald A. DePinho

    Abstract | Recent data suggest that we age, in part, because our self-renewing stem cells

    grow old as a result of heritable intrinsic events, such as DNA damage, as well as extrinsic

    forces, such as changes in their supporting niches. Mechanisms that suppress the

    development of cancer, such as senescence and apoptosis, which rely on telomere

    shortening and the activities of p53 and p16INK4a, may also induce an unwanted consequence:a decline in the replicative function of certain stem-cell types with advancing age.

    This decreased regenerative capacity appears to contribute to some aspects of mammalian

    ageing, with new findings pointing to a stem-cell hypothesis for human age-associated

    conditions such as frailty, atherosclerosis and type 2 diabetes.

    s t e m c e l l s

    R E V I E W S

    NATuRe RevIeWS |molecular cell biology vOLuMe 8 | SePTeMbeR 2007 |703

    f oc uS on a g E I n g

    http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=208900http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=277700http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=176670mailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=176670http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=277700http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=208900
  • 8/7/2019 How stem cells age and why

    2/11

    Young

    Old

    Stem cell Progenitors Effectors

    Physiological ageing, mutagenexposure or forced regeneration

    Self-renewal

    Th capacity of rplicating

    tm cll to gnrat

    daughtr cll with th am

    biological and molcular profil

    that ndow continud rnwal

    potntial. Thi can occur ithr

    aymmtrically whn a tm

    cll produc anothr tm cll

    and a mor diffrntiatd

    daughtr cll, or ymmtricallywhn tm-cll diviion giv

    ri to two idntical tm cll.

    Importantly, in matur organ

    ytm, mot cll-diviion

    activity that i rponibl for

    tiu maintnanc and

    xpanion i not lf-rnwing.

    Progeny

    Along with prognitor cll,

    th ar rlativly

    undiffrntiatd cll typ that

    ar drivd from aymmtric

    tm-cll diviion and lack th

    capacity to lf-rnw.

    organs in th adlt hman, thir importanc in rodntshas n dmonstratd xprimntally. It is thrforrasonal to srmis that prhaps som charactristicsof aging onc thoght to dgnrati mightrflct a dclin in th rgnrati capacity of rsidntstm clls across many diffrnt tisss.

    Slf-rnwal coms with som dangr for th organ-ism; in particlar, a risk of malignant transformation4,9,10.unrpaird gntic lsions in stm clls ar passd onto thir slf-rnwing daghtrs and accmlat withaging in this way. Fnctional mtations that proid agrowth or srial adantag in trn prodc positislction for th mtant stm-cll clon, with fll-fldgdcancr rslting from th accmlation of mltiplcancr-promoting nts. To offst this possiility, stmclls appar to ha old mltipl rinforcing mcha-nisms that ar aimd at maintaining gnomic intgrityyond that of othr prolifrating clls (riwd inReFs 24). Whn mtations occr dspit ths rror-prntion capacitis, potnt tmor-spprssor mcha-nisms sch as ncnc and apoptosis xist to sns

    damagd stm-cll gnoms with malignant potntialand limit rplicati xpansion or cll sch clons. Thisrlationship twn slf-rnwing clls and cancrraiss th possiility that whil carrying ot a nfi-cial, anti-cancr fnction ths tmor-spprssormchanisms may inadrtntly contrit to aging ycasing stm-cll arrst or attrition.

    Hr, w discss rcnt rfinmnts of this so-calldcancraging hypothsis, according to which cllswithin a tiss ar compromisd y ths anti-cancrmchanisms (s also th Opinion articl y Srrano andblasco in this iss). Spcifically, w rason that growth-inhiitory molcls sch as th cyclin-dpndnt

    kinas inhiitor p16INK4a and th tmor spprssorp53 xrt thir pro-aging ffcts in part throgh thiractiation in spcific slf-rnwing compartmnts schas tiu-pcific tm cll. Frthr, w dscri findingsfrom a sris of rcntly plishd hman associationanalyss that sggst a link twn th INK4/ARFlocs(also calld CDKN2a and CDKN2b) and th onst ofdistinct hman ag-associatd phnotyps. Ths rcntosrations, togthr with th findings in gntic modlsystms, proid xprimntal spport for th concptthat th actiation of tmor-spprssor mchanisms inslf-rnwing compartmnts contrits to th agingprocsss in hmans.

    Do ag?

    A dclin in rplicati fnction with ag is apprcialin many mammalian tisss. In most mammalian tisss,howr, th inaility to prify th rsidnt stm cll tohomognity, as wll as th lack of adqat modlsto tst th fnction of ths clls, has mad it difficlt todtrmin if a dclin in stm-cll fnction is indd a

    cas of th dgradation of th rgnrati capacitisthat is sn in many organs with aging.

    Ageing of haematopoietic stem cells. In th hamato-poitic systm, it is possil to prify hamatopoiticstm clls (HSCs) to nar-homognity and assay thirfnction sing alidatd assays. Thrfor, th qs-tions of whthr and how stm clls ag ar prsntlyst addrssd in this systm, with th caat that stm-cll aging may diffr mchanistically in othr tisss.Sral ffcts of aging on th lood organ ar dscridin hmans: dcrasd immnity11, incrasd incidnc ofon marrow failr and hamatological noplasia12, andmodrat anamia13,14. Oldr indiidals ar mor liklyto sffr toxicity sch as prolongd mylospprssion inrspons to traditional cytotoxic chmothrapy drgs,sggsting a rdcd marrow rgnrati capacity1517.Particlarly tlling is th clinical osration thatincrasd donor ag in on marrow transplantation is aprdictor of transplant-rlatd mortality1822, sggstingthat th diminishd rconstitting aility of HSCs fromldrly donors is partly cll-atonomos. Ths osra-tions sggst a clinically ort dcay in HSC fnctionwith normal hman aging.

    Ths corrlati findings in hmans ar ttrssdy rlatd stdis in rodnts. A srprising findinghas n that althogh HSC fnction clarly dclins

    with ag, th nmr of HSCs dos not ncssarilyalso dclin. In som strains of mic, th HSC nmractally xpands with adancing ag7,2327 and thisag-dpndnt xpansion of HSCs is a transplantal,cll-atonomos proprty of HSCs7,25. Moror, asdmonstratd y Harrison and collags, HSCs can srially transplantd into sqntial rcipints and showprsistnt fnction for >8 yars, ths xcding th lif-tim of th original donor animal28. Ths xprimntsha stalishd that cll-atonomos, rplicati HSCxhastion dos not ncssarily occr dring priods ofnormal aging in som strains of inrd mic that armaintaind ndr laoratory conditions.

    Figure 1 | Hw s s . Stem-cell number and self-renewal (curved arrow) does

    not necessarily decline with ageing, but function the ability to produce progenitors

    (blue) and differentiated effector cells (depicted in different colours) does decline.

    Stem-cell ageing can result in some systems from the heritable accumulation of DNA

    damage, which can engage tumour-suppressor activation as the stem cell attempts to

    divide asymmetrically. DNA damage can occur stochastically with normal ageing as a

    result of exposure to external mutagens, or from increased proliferation as in forced

    regeneration (see main text).

    R E V I E W S

    704 | SePTeMbeR 2007 | vOLuMe 8 www.n./ws/

    R E V I E W S

    http://www.expasy.org/uniprot/P42771http://www.expasy.org/uniprot/P04637http://www.expasy.org/uniprot/P04637http://www.expasy.org/uniprot/P42771
  • 8/7/2019 How stem cells age and why

    3/11

    Stem cells

    Self-renewal

    Multipotent

    progenitors

    Oligopotentprogenitors

    Lineage-restricted

    progenitors

    Effectorcells

    LT-HSC(LIN KIT+ SCA1+ CD34Lo FLK2Lo)

    ST-HSC(LIN KIT+ SCA1+ CD34Hi FLK2Lo)

    MPP(LIN KIT+ SCA1+ CD34Hi FLK2Hi)

    CMP

    MEP GMP

    CLP

    Erythrocytes Platelets Granulocytes Macrophages T cells B cells

    This is not to say, howr, that th rplicati fnc-tion of HSCs is not limitd y anti-cancr mchanismswith aging. It is ntirly possil that snscnt HSCschang thir srfac immnophnotyp (and thrforar no longr idntifid as HSCs y flow cytomtry) orthat th snscnc and apoptosis mchanisms ar notngagd ntil old HSCs attmpt to diid asymmtri-cally. In th lattr modl, snscnc or apoptosis coldlimit HSC fnction withot dcrasing th HSC nm-r. Moror, it is important to not that HSC nmrs

    appar to dclin with ag in othr inrd strains ofmic26,27,2931, and that sral xtrnal stimli (forxampl, chmothrapy, ionizing radiation and so on)hastn stm-cll xhastion in hmans and mic3238.Ths, gntically otrd mammals in th wild mayor may not xprinc HSC xhastion with aging,and xposr to nironmntal strsss that ar notnormally ncontrd in th laoratory stting mayfrthr indc HSC xhastion. Finally, indpndntlyof rplicati fnction, HSCs xhiit cll-intrinsic,fnctional signs of aging. Nmros stdis hashown that aging altrs HSC fnction with rgard tomoilization39, homing7,23,24,31,39 and linag choic7,24,31.In particlar, thr is a loss of lymphoid linag potn-tial with a skwing toward myloid linags in HSCsfrom old mic, and old HSCs dmonstrat rprodc-il changs in gn xprssion with ag, incldingincrasd xprssion of myloid linag transcripts7.Thrfor, th prpondranc of idnc sggststhat HSCs ndrgo cll-intrinsic aging, althoghthr is also mrging idnc that th aging HSC

    micronironmnt may inflnc HSC fnction in anxtrinsic mannr (s low).

    Ageing in other self-renewing compartments. Srallins of idnc sggst that stm clls from othrtisss sffr prolifrati dclin with adancingag (FIG. 1). For xampl, in rodnts, a dclin in thnmr of nw nrons prodcd y nral stm clls(NSCs) with ag or tlomr dysfnction has n doc-mntd40,41, as has th in vivo prolifration of NSCs4143.This dclin in th capacity for mrin nrognsishas n associatd with a progrssi Parkinsoniandisas41 and with an impairmnt of olfactory discrimin-ation with aging44. Likwis, hair grying has nlinkd to dcrasd mlanocyt stm-cll maintnanc,possily in association with mlanolast snscnc45.

    Th capacity of th inslin-prodcing -cll of thpancratic islt to rplicat in adlt rodnts throghotlif has n known for dcads, althogh th importancof this rgnrati capacity with rgard to th dlop-mnt oftyp 2 diats mllits (also calld adlt-onstdiats mllits) has only rcntly n apprciatd.Th prailing iw had n that typ 2 diats mll-its was a mtaolic disas rslting largly from anag-associatd dclin in th aility of mscl and lir torspond to inslin (inslin rsistanc), t rcnt carflanalyss of islt mass ha challngd this iw. Stdis

    in th immdiat post-mortm priod ha shown sig-nificant rats of-cll prodction and apoptosis, nin agd adlts, with an incrasd -cll mass notd inos indiidals and a rlatily rdcd -cll massamong adlts with diats46,47. Thrfor, th de novosynthsis of-clls throgh slf-rnwal appars to th prdominant sorc of islt mass in adlt hmans.Islt rplication appars to dclin with hman aging47,althogh -cll rplication has n rportd in hmanpatints p to 89 yars of ag48. Consistnt with thsfindings in hmans, a sharp dclin in -cll prolifra-tion with aging in mic has n rcntly dscrid49.Mor than 1% of islt clls from yong mic prolifrat

    Box 1 | th hirarhy of haaopoii

    Multipotnt tissue-specific stem cells produce differentiated effector cells through a

    series of increasingly more committed progenitor intermediates. This tissue stem-cell-

    derived differentiation process has been best characterized in the haematopoietic

    system (see figure). Here, long-term haematopoietic stem cells (LT-HSCs) represent the

    true stem cells that self-renew and produce multipotent progenitors (short-term (ST)-

    HSCs and, subsequently, multipotent progeny (MPP)) with no self-renewal capacity.

    These in turn give rise to oligopotent progenitors including the common lymphoid

    progenitor (CLP) and common myeloid progenitor (CMP), which then yields the

    granulocytemacrophage progenitor (GMP) that differentiates into monocytes,

    macrophages and granulocytes, and the megakaryocyteerythrocyte progenitor (MEP)that differentiates into megakaryocytes, platelets and erythrocytes. Similar hierarchies

    are also thought to exist in other stem-cell-containing tissues (for example, brain, gut,

    liver and lung).

    A principal advantage of studies in the haematopoietic system is that stem and

    progenitor cells from this organ can be purified to near-homogeneity by surface

    markers. For example, LT-HSCs express low levels of lineage markers (LIN), high levels

    of the CD117/c-KIT receptor (KIT+), high levels of a surface marker called SCA1 (SCA1 +),

    and low levels of another surface marker, CD34 (CD34 Lo). With each asymmetric

    division of a LIN KIT+SCA1+CD34Lo LT-HSC, there is the production of another LT-HSC

    and a multipotent daughter cell with limited renewal potential, ST-HSC, which has a

    similar surface immunophenotype to LT-HSC except that it has higher levels of CD34

    (CD34Hi). As ST-HSC in turn proliferate to form more differentiated MPP, they increase

    expression of another surface marker, FLK2. Development from oligopotent

    progenitors to mature blood cells proceeds through several intermediate progenitors

    that are not shown.

    R E V I E W S

    NATuRe RevIeWS |molecular cell biology vOLuMe 8 | SePTeMbeR 2007 |705

    f oc uS on a g E I n g

    http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=125853http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=125853http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=125853
  • 8/7/2019 How stem cells age and why

    4/11

    20 40 60 80

    Mouse age (weeks)

    Proliferatingisletcells

    (%oftotalisletcells)

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    1.4

    p16INK4a knockout

    p16INK4a transgenic

    Wild-type

    Senescence

    A pcializd form of growth

    arrt inducd by variou

    trful timuli including lo

    of tlomr function, ractiv

    oxygn pci, om form of

    DNA damag and activation of

    crtain oncogn or

    ractivation of tumour-

    uppror gn. sncnc

    i charactrizd by vral

    markr uch a ncnc-

    aociatd--galactoida,

    altration in chromatin

    tructur (ncnc-

    aociatd htrochromatic

    foci) and a markd incra in

    th crtion of vral

    cytokin and othr bioactiv

    molcul (ncnc-aociatd crtory

    phnotyp).

    Tissue-specific stem cell

    A pcializd cll found in

    many tiu of adult. Th

    cll can rplac thmlv

    through lf-rnwal and ar

    gnrally multipotnt, in that

    thy can giv ri to progny

    that can diffrntiat into

    multipl diffrnt cll typ of

    th aociatd organ.

    Multipotency

    Th ability to giv ri to

    diffrntiatd progny of

    diffrnt pcializd ubtyp.

    Howvr, om lf-rnwing

    cll (for xampl, pancratic

    -cll) hav a narrow potntial

    for diffrntiation, gnrating

    progny imilar to th parntal

    cll. Thi typ of lf-rnwing

    cll i trmd a unipotnt

    prognitor, which can b

    viwd a a pcial tm-cll

    ubtyp, at lat in trm of

    long-trm prolifrativ

    capacity. For convninc, in

    thi Rviw th trm tm cll

    i applid to both typ of

    adult lf-rnwing cll.

    Telomere

    A nucloprotin complx at

    th nd of chromoom that

    maintain chromoomal

    intgrity. It conit of many

    doubl-trandd 5-TTAGGG-3

    rpat, a 3-ingl-trandd

    ovrhang and aociatd

    tlomr-binding protin,

    which togthr gnrat a

    cappd tructur that i

    imprviou to th action of

    complx that rpair DNA

    damag.

    ndr stady-stat conditions, t this frqncydclins y narly tnfold aftr a yar of aging (FIG. 2).Thrfor, rathr than ing mrly a disas of inslinrsistanc, typ 2 diats mllits partly rslts from arlati failr of islt rplication.

    Cell-extrinsic ageing.Ths osrations, howr, donot mak clar which islt-associatd compartmnt isaging: is it th -clls thmsls, a ptati pancraticstm cll or anothr tiss that inflncs-cll prolifr-ation in a cll non-atonomos mannr? In fact, cllnon-atonomos factors ha n shown to ha amajor rol in th rglation of stm-cll aging in othrsystms. Rando and collags ha shown that thaging of a rgnrati cll of mscl, th satllit cll,is inflncd y th agd micronironmnt50. Satllitclls from agd mic wr rjnatd y xposr toa yong lood spply, sggsting that th agd mili,hormonal or othrwis, inflncs th rplicati fnc-tion of satllit clls. Likwis, whil som aspcts of thfnctional dclin that charactrizs aging of HSCs has

    n shown to cll-atonomos, th HSC nich isalso xpctd to inflnc HSC aging to som dgr.For xampl, in mic with shortnd tlomrs, anag-dpndnt dcras was notd in th aility ofrcipint animals to spport lymphopoisis of trans-plantd HSCs from yong mic with normal tlomrs,and nhancd myloid prolifration occrrd whnHSCs wr transplantd into a micronironmnt thatharord dysfnctional tlomrs51.

    Althogh this distinction twn cll-intrinsic andcll-xtrinsic aging is important, aging is, ltimatly, thsmmati proprty of th organism in toto. For xam-pl, th finding that srm from yong mic nhancs

    satllit-cll prolifration sggsts that cll-intrinsicaging of som othr tiss occrs sch that it lao-rats (or fails to laorat) th rlant srm factor thatmodlats th prolifrati capacity of satllit clls. Thaility to targt gns conditionally in a tiss-spcificand tmporal mannr will prmit in vivo analyss todtrmin which compartmnts ag intrinsically, rssthos that ag y proxy.

    Wha au o ag?

    Althogh a fw mchanisms ha n sggstd toxplain clllar aging, indpndnt lins of idncsggst that forms of DNA damag lad to th actiationof tmor-spprssor mchanisms, sch as snscnc,to limit stm-cll fnction with incrasing ag.

    DNA damage results in stem-cell attrition. Th acc-mlation of damag to clllar macromolcls (forxampl, protins and DNA) has n postlatd to a cas of clllar attrition with aging52. DNA issjctd to spontanos and xtrinsic mtational

    nts on a daily asis, and dspit a formidal cap-acity for rpair, som damagd DNA appars to adrpair and accmlats or tim. eidnc in spportof th notion that DNA damag attnats stm-cllfnction with ag has n proidd y th stdy ofHSCs from mic that haror altrations in th DNA-damag rspons. Significant fnctional dfcts ar snin HSCs from mic that ar dficint in DNA-rpairprotins sch as FANCD1(ReF. 53), MSH2 (ReF. 54) oreRCC1 (ReF. 55). A mos strain with a ial, hypo-morphic alll of th DNA-rpair protin DNA ligas Ivthat was rcntly idntifid throgh a mtagnsisscrn xhiits a markd, ag-indcd dclin in HSCnmr and fnction56. Additionally, Morals et al. hashown that mic with a mtant alll of RAD50, a mm-r of th MRe11 DNA-rpair complx, dmonstratprofond on marrow hypoplasia57. This ffct can rscd in th stting of ataxia-tlangictasia mtatd(ATM) kinas dficincy, sggsting that th Rad50alll is hyprmorphic and that xcss DNA-damagsignalling rdcs HSC nmr and/or fnction.Ths data sggst that sral forms of DNA rpairar ndd to maintain HSC gnomic intgrity, and thatactiation of a rspons to DNA damag compromissHSC fnction.

    A dmonstration of DNA damag in a tiss-spcific stm-cll compartmnt with aging coms from

    a rcnt rport y Rossi and collags58. using micwith grmlin dficincis in DNA rpair or tlomrmtaolism (K80-, XPD- and mTeRC-knockot mic),th athors dmonstratd a markd prmatr dclinin th rgnrati fnction pr HSC in sral assays.This stdy frthr notd th incrasd xprssion ofmarkrs of th DNA-damag rspons (for xampl,histon H2AX foci) in highly prifid HSCs with normalphysiological aging n in wild-typ mic. Th athorsnotd an incras in apoptosis of progny from old HSCs,t not a dclin in th aility of old HSCs to prolifratin in vitro assays. Ths osrations corrspond wllwith a diminishd capacity of th hamatopoitic systm

    Figure 2 | Pfn f-s wh . A sharpdecline in pancreatic-cell proliferation with ageingnormally occurs in wild-type mice (blue). This decline is

    modulated by expression of the p16INK4a tumour

    suppressor: increased expression (in a transgenic line;

    green) correlates with reduced proliferation, whereas

    decreased activity (in the p16INK4a knockout line; orange)

    affords a resistance to -cell ageing.

    R E V I E W S

    706 | SePTeMbeR 2007 | vOLuMe 8 www.n./ws/

    R E V I E W S

    http://www.expasy.org/uniprot/Q62388http://www.expasy.org/uniprot/Q62388
  • 8/7/2019 How stem cells age and why

    5/11

    Cancer

    Regenerative failure,SA-SP

    Tissue dysfunctionand failure

    Tissue dysfunctionand failure(for example, MDS)

    Transformation

    Senescence

    Apoptosis

    Dysfunction

    RAS mutation,p53 loss

    Telomeredysfunction

    Unrepaired DSBs

    Y-chromosomeor 5q loss

    Stem cell

    Telomerase

    A ribonucloprotin complx

    that xtnd th nd of

    tlomr aftr rplication by

    uing tlomra rvr

    trancripta (TeRT) and an

    RNA tmplat (TeRC) that i

    part of th nzym complx.

    in tlomr-dysfnctional mic to rcor followingchmothraptic challng, as wll as with a progrs-si nrodgnrati condition that is associatd withdcrasd NSC rsrs and fnction in th tlomra-knockot modl41,59. Togthr, ths rslts ar consist-nt with th iw that DNA damag accmlats withaging in th HSC compartmnt, as idncd y thaccmlation of H2AX foci, and that this damag can physiologically significant if nrpaird.

    bt what cass DNA damag in stm-cll com-partmnts in adlt mic? Rcnt data from Rzankinaet al., who sd a conditional alll for th DNA-dam-ag rspons gn Atr(ataxia tlangictasia and Rad3-rlatd), ar worth considring in this rgard60. LossofAtris toxic to prolifrating clls61 and whn Atrwassomatically xcisd in adlt mic in a widsprad man-

    nr y conditional inactiation, th ast majority of pro-lifrating clls rapidly disappard, prodcing markdintstinal atrophy and on marrow hypoplasia 2 wksaftr conditional actiation. Howr, th animals sr-id this transint priod of cll loss cas rar stmclls that had not rcomind th Atralll rplacdth lost clls. by 1 month aftr conditional actiation, thmic appard largly normal, with rapidly prolifr-ating tisss that had n flly rconstittd y spor-adic Atr-comptnt clls. Srprisingly thogh, thsrconstittd mic thn dlopd a markd progroidphnotyp a fw months latr, with ostopania, gryingand loss of lymphoid and hamatopoitic prognitors.

    Thrfor, th xcss rgnration that was forcd tooccr to rconstitt prolifrati tisss aftr transintAtrinactiation in som prolifrating clls prodcda dral compromis of stm-cll fnction in Atr-comptnt clls (FIG. 1), althogh it is important tomphasiz that th possil adrs ffcts of ATRdltion on th stm-cll nich may also ha contri-td to th acclratd aging. This rslt is consistntwith th iw that sch forcd rgnration in rsponsto homostatic dmands, n in th asnc of xtrnalDNA-damaging agnts, can toxic to stm clls acrossmany organ systms.

    In th DNA-damag accral modl of aging,nrpaird (or improprly rpaird) gnomic damagaccmlats with aging in stm-cll compartmnts.At som point, accmlatd damag prtrs normalstm-cll iology, driing stm clls to a fw possilfats: transformation, snscnc, apoptosis or dysfnc-tion; for xampl, a loss of th aility to rostly pro-dc progny or an impaird potntial for mltilinagdiffrntiation (FIG. 3). As this procss procds with tim,

    dpltd and/or dysfnctional stm-cll compartmntscannot match th rgnrati nds of a gin organand homostatic failr nss. Likwis, if oncognicDNA-damag-indcd lsions accmlat, slf-rnw-ing clons that contain sch lsions ndrgo positislction, lading to cancr. Thrfor, it is tmpting tospclat that cancr and aging ar rlatd ndpointsof accmlating DNA damag within slf-rnwingcompartmnts.

    How telomere shortening could affect stem-cell ageing.Thr has n significant intrst in th possiility thattlomr dysfnction, a spcializd form of DNA dam-ag, contrits to th aging of hman stm clls. In thasnc of adqat tlomras actiity, tlomr short-ning inxoraly occrs with prolifration, ntallytriggring a chang in tlomr strctr that is snsdy th cll as a DNA dol-strand rak. In hmans,rost tlomras actiity is prdominantly rstrictdto grm-cll compartmnts, som somatic arly stm-cll or prognitor compartmnts and prolifratinglymphocyts.

    Whras tlomr dysfnction in th stting of anintact DNA-damag rspons can sr as a potnttmor-spprssor mchanism, th rol of tlomr-mdiatd chckpoints in stm-cll aging rmains anara of acti instigation. Mic ha long tlomrs

    rlati to hmans and, ths, tlomr dysfnctionappars not to a major cas of stm-cll xhastionin many strains ofMus musculus. Accordingly, in thtlomras-knockot mos62, th lack of tlomrasactiityper se prodcs a modst phnotypic ffct inadlt mic59,63. Mrin tlomr lngth can madmor limiting and rdcd to a hmanizd lngth ysrial intrcrossing of mic that ar dficint in tlo-mras actiity. In this xprimntal stting, tlomrlngth coms shortr with ach sccssi gnrationntil tlomr dysfnction nss with dramatic phno-typic ffcts, ths proiding a modl systm to addrssth stm-cll iss.

    Figure 3 | Fs f s s. A limited number of outcomes appear to be

    possible for stem cells with heritable DNA damage. Although it is likely that many

    mutational events do not lead to any alteration of stem-cell function, significant

    damage is expected to induce apoptosis, senescence, transformation or dysfunction.

    Importantly, these outcomes are not necessarily mutually exclusive; for instance, manygenetic events associated with stem-cell dysfunction (for example, the oncogenic

    translocation that fuses BCRandABL (forming the Tyr kinase BCR/ABL) in

    haematopoietic stem cells (HSCs)) are also associated with subsequent transformation.

    Examples of DNA lesions associated with each outcome are indicated. Mutations in RAS

    and the tumour-suppressor p53 are associated with transformation, whereas loss of the

    long arm of chromosome 5 (5q) or the Y-chromosome are associated with dysfunction

    of HSCs, manifesting as myelodysplasia (MDS). Unrepaired double-strand DNA breaks

    (DSBs) and telomere dysfunction induce apoptosis and senescence, respectively.

    SA-SP, senescence-associated secretory phenotype.

    R E V I E W S

    NATuRe RevIeWS |molecular cell biology vOLuMe 8 | SePTeMbeR 2007 |707

    f oc uS on a g E I n g

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=245000&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=245000&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSum
  • 8/7/2019 How stem cells age and why

    6/11

    Chromosome 9p21

    Exon 1

    p15INK4b p16INK4a ARF

    2 1 1 2 3

    CDK4/6 MDM2

    p53

    CHK1 and/or CHK2

    ATM and/or ATR

    DNA damage (for example,telomere dysfunction, ROS,stalled replication forks)

    p21CIPpRB family

    Senescence Apoptosis

    Region of linkage disequilibrium

    Human 9p21 22,000k 22,100k

    ANRIL

    ARF

    p15INK4b

    p16INK4a

    a

    b

    ** * * *** ** *

    Animals with dysfnctional tlomrs dlop fa-trs of prmatr aging casd y th actiation ofsnscnc and apoptosis mchanisms in crtain slf-rnwing compartmnts sch as HSCs, th intstinal

    crypt and th tsts59,6366. Dficincy of p53 rscs manyof th stm-cll dfcts in mic that haror dfctitlomrs, t dos not xtnd th lifspan of ths miccas of incrasd tmorignsis67,68. by contrast, lossof p21CIP, a p53 transcriptional targt that potntly inhi-its th cll cycl (FIG. 4a), partially xtnds th longityof mic with tlomr dysfnction withot incrasdtmorignsis, and attnats som of th prolifratidfcts that ar sn in arios stm-cll compartmntsof tlomr-dficint mic66. Thrfor, DNA dam-ag indcd y tlomr shortning is partly snsdand managd throgh p53, and p53 xrts importantanti-prolifrati ffcts in stm clls throgh p21CIP.

    Senescence contributes to ageing. DNA damag andtlomr dysfnction appar to actiat th classicaltmor-spprssor mchanisms of snscnc andapoptosis. Snscnc rqirs actiation of th rtino-lastoma (Rb) and/or p53 protins and xprssion ofthir rglators, most prominntly p16INK4a and ARF(ReFs 6971)(FIG. 4a). Th notion that snscnc pr-nts cancr is wll-spportd and is not controrsial(riwd in ReFs72,73). Th xprssion of markrs ofsnscnc sch as snscnc-associatd-galactosidasand p16INK4a markdly incrass with aging in manytisss from disparat mammalian spcis (riwd inReF. 73; s also th Riw y Campisi and DAdda diFagagna in this iss). Caloric rstriction (CR) potntlyslows aging in rodnts, and CR and its rlatd ditarychangs rtard or n aolish th ag-indcd incrasin th xprssion of snscnc markrs, inclding thxprssion of p16INK4a(ReFs 7476). Proocatily, CR,similar to p16INK4a dficincy77, nhancs stm-cll fnc-tion with aging78, which sggsts th possiility that CRmay slow aging in mammals y dcrasing th actiation

    of snscnc in slf-rnwing compartmnts.A rol for ROS in snscnc ars particlar rl-

    anc. ROS indcs snscnc in crtain cll-cltrsystms79,80 t may also ha an important rol in ffct-ing th snscnt phnotyp81. In ATM-dficint mosHSCs, incrasd ROS lls appar to compromis HSCfnction in part throgh a p38MAPK-dpndnt actiationofInk4a/Arfxprssion34,35, which can attnatd yantioxidants. Likwis, HSCs that lack th FOXO trans-cription factors show diminishd slf-rnwal and pr-matr xhastion ndr conditions of incrasd ROSprodction82. Thrfor, FOXO transcription factorsmaintain HSC qiscnc and srial, primarily iath rglation of physiological lls of ROS. WhilFOXO-dficint HSCs show ROS-dpndnt nhancdcll-cycl ntry and incrasd apoptosis, incrasd ROSmay also rslt in th accmlation of DNA damagand nschdld actiation of snscnc mchanismsin this stm-cll compartmnt in th long trm.

    Althogh th xprssion of snscnc markrsis associatd with aging, this osration dos notstalish a casal rlationship twn snscnc andaging. Rcnt stdis of slf-rnwal in HSCs, NSCsand pancratic islt clls from p16INK4a-dficint andp16INK4a-orxprssing mic ha gn to addrss thisiss43,49,77,83. Ths rslts showd that incrasing llsof p16INK4a ar not only associatd with aging, t partly

    contrit to th ag-indcd rplicati failr of thstisss. In all thr compartmnts, p16INK4a dficincyattnatd th ag-indcd dclin in prolifrationand fnction. Likwis, orxprssion of p16 INK4aattnatd HSC fnction and islt prolifration in anag-dpndnt mannr (FIG. 2). Th ffcts of p16INK4aloss wr consistnt across ths disparat slf-rnwingtisss, sggsting that p16INK4a can promot agingin tisss that ar dlopmntally distinct. Howr,th loss of p16INK4a did not compltly arogat thffcts of aging in any of th organs stdid, indicat-ing that p16INK4a-indpndnt aging occrs in ach ofths compartmnts.

    Figure 4 | SNPs n - phnps h INK4a/ARF/INK4b s n

    hn hs 9p21. | Proteins encoded by the INK4/ARFlocus on

    chromosome 9p21 regulate the p53 and retinoblastoma protein (pRB) tumour-

    suppressor pathways to promote senescence (p53 and pRB-family activation) or

    apoptosis (p53 activation). Activation of p53 in response to several types of DNA damage

    also can occur via an ataxia-telangiectasia mutated (ATM)/ataxia telangiectasia andRad3-related (ATR)-mediated pathway independent of ARF function. | Single

    nucleotide polymorphisms that are significantly associated with the indicated

    phenotypes are shown (frailty, green; vascular heart disease, red; type 2 diabetes

    mellitus, orange). The open reading frames for p16INK4a, ARF and p15INK4b are indicated,

    as is the ANRIL transcript. The large (>100 kb) region of linkage disequilibrium is also

    indicated. CDK4/6, cyclin-dependent kinases-4/-6; CHK1/2, checkpoint kinases-1/-2;

    MDM2, murine double minute-2, p53-binding protein; ROS, reactive oxygen species.

    R E V I E W S

    708 | SePTeMbeR 2007 | vOLuMe 8 www.n./ws/

    R E V I E W S

    http://www.expasy.org/uniprot/P06400http://www.expasy.org/uniprot/P06400
  • 8/7/2019 How stem cells age and why

    7/11

    Single nucleotide

    polymorphism

    A common, ingl-ba

    diffrnc in a gn among

    individual within a pci.

    Frailty

    A clinically validatd, functional

    maur ud in clinical

    griatric. It i cord a a

    continuou variabl uing a

    ri of routin, aily

    maurd tt uch a gait

    pd. Frail individual ar l

    abl to liv indpndntly, ar

    mor likly to harbour co-morbid illn, and xhibit

    incrad mortality.

    Linkage disequilibrium

    (LD). A maur of gntic

    aociation btwn alll at

    diffrnt loci, which indicat

    whthr alllic or markr

    aociation on th am

    chromoom ar mor

    common than xpctd. Loci

    ar gnrally conidrd to b

    in trong LD if thir corrlation

    i highr than a pr-dfind

    cut-off (for xampl, 0.8).

    Rlatd osrations ha likwis sggstd a pro-aging rol for p53 and its ffctors in mic66,84,85 andhmans86. Th cas for p53, howr, appars to morcomplicatd cas p53 and its downstram ffctorssch as p21CIP also ha important rols in rglating thDNA-damag rspons. In fact, an intriging rcnt stdyy Srrano and collags, who sd carflly dsigndtransgnic strains, has shown that incrasd ARF andp53 actiity can incras th mrin lifspan y prnt-ing cancr withot an attndant incras in organismalaging87. Additionally, HSCs from p21CIP-dficint micdmonstrat prmatr xhastion88, consistnt withth notion that a p53- and p21CIP-dpndnt cll-cyclpas in rspons to DNA damag may importantfor stm-cll longityin vivo. Ths rslts sggstthat p53 actiation can oth pro-aging and anti-aging dpnding on th natr and dration of th strsshind its actiation.

    Aging and h INK4/ARFou

    Th dscrid links twn stm-cll fnction and

    aging prdict that indiidals who diffrntially rg-lat snscnc-promoting mchanisms might xhiitdiffrnt prdispositions to cancr rss aging.Accordingly, hypomorphic allls that affct p16 INK4a orp53 fnction and that ar associatd with incrasd typsof cancr ar wll dscrid (riwd in ReF. 73). Rcntdata also sggst that non-coding polymorphisms narth opn rading frams (ORFs) of p16 INK4a, ARF andp15INK4 modify th onst of ag-associatd phnotypsin hmans. In a rmarkal sris of rcnt stdis,ingl nuclotid polymorphim (SNPs) that ar ry clos(1 M fromth rlatd ORFs ar wll stalishd99 and, thrfor, thINK4/ARFlocs appars wll within th rang of schrglatory lmnts. Althogh thr has n a rcntdscription of an apparntly non-protin-coding tran-script in this rgion (ANRIL100; FIG. 4b), thr dos notappar to anothr protin-coding transcript narths SNPs. Thrfor, in light of th mrin gnticstdis that link Ink4a/Arfand stm-cll fnction, pro-

    tins ncodd y th locs ar th strongst candidatsto mdiat th ffcts of ths polymorphisms on thincidnc of ths common hman disass that arassociatd with aging.

    Ouanding quion

    W li that th data drid from disparat rodntand hman xprimntal systms spport th iw thata dclin in th rgnrati fnction of stm clls withag contrits to mammalian aging and ag-associatddisas. Gin this modl, a fw crcial qstions facingth fild rmain.

    Does telomere dysfunction affect human ageing?Animportant iss is whthr sfficint tlomr attritionoccrs in hman slf-rnwing compartmnts dringphysiological aging to actiat a DNA-damag rsponsand th ssqnt compromis of rplicati fnc-tion. W fl that th crrnt data do not yt proida clar answr. Hmans who haror short tlomrscas of congnital dficincis of componnts of thtlomras complx (th RNA componnt TeRC, tlom-ras rrs transcriptas (TeRT) or dyskrin) dlopan ag-rlatd failr of on marrow or lng101105.Moror, tlomr shortning in th lir prcds thonst of lir failr (cirrhosis) in patints with chronichpatitis106109, an association that has n alidatd in

    th tlomras-dficint mos110. A fw stdis hadmonstratd a corrlation twn tlomr lngthin priphral lood lymphocyts (PbLs) and th onstof crtain disass that ar associatd with aging. Schstdis in non-noplastic disass ha shown that PbLtlomr lngths can proid a iomarkr that forcaststh dlopmnt of athrosclrosis111,112, cancr risk113,114and mortality115. Ths rslts spport a connctiontwn tlomr dysfnction and hman aging.

    by contrast, argmnts can mad against tlo-mr-asd aging in hmans. Mrin aging appars toha many similaritis to hman aging, yt w lithat it occrs largly indpndntly of tlomr lngth.

    R E V I E W S

    NATuRe RevIeWS |molecular cell biology vOLuMe 8 | SePTeMbeR 2007 |709

    f oc uS on a g E I n g

    http://www.expasy.org/uniprot/P42772http://www.expasy.org/uniprot/P42772
  • 8/7/2019 How stem cells age and why

    8/11

    Additionally, althogh tlomr lngth shortns withhman aging in many tisss, orall tlomr lngthdos not linarly corrlat with ag, with th most rapidshortning occrring y yong adlthood102,105,116. It ispossil, howr, that man tlomr lngth is a poorindicator of tlomr stats cas th shortst tlomrappars to capal of actiating th DNA-damagrspons117.

    Althogh th osrations of on marrow failrand plmonary firosis in patints with dfcti tlo-mras componnts stalishs that tlomr dysfnc-tion can cas hman disas, som indiidals fromsch kindrds who haror th dfcti alll do notdmonstrat any ort phnotyp. In fact, htrozygosTeRT and TeRC grmlin mtations in hmans showstrong anticipation102,104,105, which mans that thphnotypic ffcts of th mtation ar mor srin ssqnt gnrations; this is analogos to find-ings in tlomras-dficint mic. Thrfor, TeRT orTeRC dficincy is most phnotypically prononcd inpatints that also inhrit for-shortnd tlomrs from

    thir parnts, sggsting that tlomras dficincyper semay not sfficint to indc ort ag-associatdpathology. Importantly, howr, stdis of th naf-fctd carrirs of ths kindrds ha so far not nsfficint to xcld stl, ag-associatd phnotyps,and phnotypic consqncs of tlomras dficincymay notd in sch indiidals with mor carflosration. Considrd in aggrgat, w li that thdata ar consistnt with oth tlomr-indpndnt andtlomr-dpndnt aging of hman stm clls.

    How do senescence factors contribute to ageing?Thsimplst xplanation for how p16INK4a limits rplica-ti fnction with ag wold that th indction ofp16INK4a, in rspons to snscnc-promoting cs,intrinsically limits rplication y indcing snscncor at last dcrasing cll-cycl ntry. For xampl,pancratic -cll rplication is known to rqir cyclin-dpndnt kinas-4 (CDK4) actiity118,119, th iochmi-cal targt of p16INK4. bcas p16INK4a accmlats withphysiological aging in hman and rodnt islts49,120,and th loss of p16INK4a agmnts islt prolifration inan ag-dpndnt way49, p16INK4a might dirctly limitth rplication of slf-rnwing clls with aging. Thiscll-atonomos modl is frthr spportd y th factthat th nhancd fnction of p16INK4a-dficint HSCs issn n whn sch clls ar transplantd into p16INK4a-

    comptnt rcipint mic77,83. Importantly, howr,p16INK4a xprssion incrass significantly with agingin linag-ngati on marrow clls (of which 1 M) rglation of anothr protin-codingor microRNA transcript. Stdis in hmans and mic

    to ndrstand whthr this corrlation rflcts an ffctofINK4/ARF-asd tmor-spprssor mchanisms onhman aging ar moing at a rapid pac. Sch ffortsar hamprd, howr, y a larg rgion of linkagdisqilirim that compriss th INK4a/ARFlocs aswll as almost all of th disas-corrlatd SNPs (FIG. 4b),which implis that th rlant gntic rglatory ntscold hiding anywhr within a larg rgion arondp15INK4b. Rgardlss of th mchanistic asis, howr, asth minor alll frqncis of th rlant SNPs in achof ths rports ar larg (>10%), ths polymorphismsappar to ha a major rol in dtrmining th onst ofths highly common, ag-indcd phnotyps.

    R E V I E W S

    710 | SePTeMbeR 2007 | vOLuMe 8 www.n./ws/

    R E V I E W S

  • 8/7/2019 How stem cells age and why

    9/11

    conuion

    In smmary, w li th data sggst that w growold partly cas or stm clls grow old as a rslt ofmchanisms that spprss th dlopmnt of cancror a liftim. In this rgard, or slf-rnwing stmclls appar to grow old cas of hrital intrinsicnts, sch as DNA damag, t also d to cll-xtrinsic nts sch as altrations in thir spportingnichs. Anti-cancr mchanisms sch as snscnc and

    apoptosis, which rly on tlomr shortning and/orp53 and p16INK4a actiation, appar to promot agingjst as thir failr is associatd with cancr. W lithat a frthr, mor prcis mchanistic ndrstandingof this procss will rqird for this knowldgcan translatd into hman anti-aging thrapis. Forth tim ing, th most prdnt, clinically alidatdadic appars still to : dont smok, at rasonalyand tak xrcis.

    1. Kudlow, B. A., Kennedy, B. K. & Monnat, R. J. Jr.

    Werner and HutchinsonGilford progeria syndromes:

    mechanistic basis of human progeroid diseases.

    Nature Rev. Mol. Cell Biol. 8, 394404 (2007).

    2. Bryder, D., Rossi, D. J. & Weissman, I. L.

    Hematopoietic stem cells: the paradigmatic tissue-

    specific stem cell. Am. J. Pathol. 169, 338346

    (2006).

    3. Morrison, S. J. & Kimble, J. Asymmetric and

    symmetric stem-cell divisions in development and

    cancer. Nature441, 10681074 (2006).

    4. Sharpless, N. E. & DePinho, R. A. Telomeres, stem

    cells, senescence, and cancer. J. Clin. Invest.113,

    160168 (2004).

    5. Hodgson, G. S. & Bradley, T. R. In vivo kinetic status of

    hematopoietic stem and progenitor cells as inferred

    from labeling with bromodeoxyuridine. Exp. Hematol.

    12, 683687 (1984).

    6. Passegue, E., Wagers, A. J., Giuriato, S.,

    Anderson, W. C. & Weissman, I. L. Global analysis of

    proliferation and cell cycle gene expression in the

    regulation of hematopoietic stem and progenitor cell

    fates. J. Exp. Med. 202, 15991611 (2005).

    7. Rossi, D. J. et al. Cell intrinsic alterations underlie

    hematopoietic stem cell aging. Proc. Natl Acad.

    Sci. USA102, 91949199 (2005).

    8. Weissman, I. L. Stem cells: units of development,

    units of regeneration, and units in evolution. Cell100,

    157168 (2000).

    9. Reya, T., Morrison, S. J., Clarke, M. F. & Weissman, I. L.

    Stem cells, cancer, and cancer stem cells. Nature414,

    105111 (2001).

    10. Campisi, J. Cancer and ageing: rival demons?

    Nature Rev. Cancer3, 339349 (2003).11. Linton, P. J. & Dorshkind, K. Age-related changes in

    lymphocyte development and function. NatureImmunol. 5, 133139 (2004).

    12. Lichtman, M. A. & Rowe, J. M. The relationship of

    patient age to the pathobiology of the clonal myeloid

    diseases. Semin. Oncol. 31, 185197 (2004).

    13. Beghe, C., Wilson, A. & Ershler, W. B. Prevalence and

    outcomes of anemia in geriatrics: a systematic review

    of the literature. Am. J. Med.116 (Suppl 7A), 3S10S

    (2004).14. Guralnik, J. M., Eisenstaedt, R. S., Ferrucci, L.,

    Klein, H. G. & Woodman, R. C. Prevalence of anemia

    in persons 65 years and older in the United States:

    evidence for a high rate of unexplained anemia.

    Blood104, 22632268 (2004).

    15. Appelbaum, F. R. et al. Age and acute myeloid

    leukemia. Blood107, 34813485 (2006).16. Brunello, A. et al. Adjuvant chemotherapy for elderly

    patients (> or =70 years) with early high-risk breast

    cancer: a retrospective analysis of 260 patients.

    Ann. Oncol.16, 12761282 (2005).

    17. Lenhoff, S. et al. Impact of age on survival afterintensive therapy for multiple myeloma: a population-

    based study by the Nordic Myeloma Study Group.

    Br. J. Haematol.133, 389396 (2006).

    18. Kollman, C. et al. Donor characteristics as risk factors

    in recipients after transplantation of bone marrow

    from unrelated donors: the effect of donor age. Blood

    98, 20432051 (2001).

    19. Ash, R. C. et al. Bone marrow transplantation from

    related donors other than HLA-identical siblings: effect

    of T cell depletion. Bone Marrow Transplant.7,

    443452 (1991).

    20. Castro-Malaspina, H. et al. Unrelated donor marrow

    transplantation for myelodysplastic syndromes:

    outcome analysis in 510 transplants facilitated by the

    National Marrow Donor Program. Blood99,

    19431951 (2002).21. Buckner, C. D. et al. Marrow harvesting from normal

    donors. Blood64, 630634 (1984).

    22. Yakoub-Agha, I. et al. Allogeneic marrow stem-cell

    transplantation from human leukocyte antigen-

    identical siblings versus human leukocyte

    antigen-allelic-matched unrelated donors (10/10) in

    patients with standard-risk hematologic malignancy:

    a prospective study from the French Society of Bone

    Marrow Transplantation and Cell Therapy. J. Clin.

    Oncol.24, 56955702 (2006).

    23. Morrison, S. J., Wandycz, A. M., Akashi, K.,

    Globerson, A. & Weissman, I. L. The aging of

    hematopoietic stem cells. Nature Med.2, 10111016

    (1996).

    24. Sudo, K., Ema, H., Morita, Y. & Nakauchi, H. Age-

    associated characteristics of murine hematopoietic

    stem cells. J. Exp. Med. 192, 12731280 (2000).

    25. Pearce, D. J., Anjos-Afonso, F., Ridler, C. M.,

    Eddaoudi, A. & Bonnet, D. Age-dependent increase in

    side population distribution within hematopoiesis:

    implications for our understanding of the mechanism

    of aging. Stem Cells25, 828835 (2006).26. de Haan, G., Nijhof, W. & Van Zant, G. Mouse strain-

    dependent changes in frequency and proliferation of

    hematopoietic stem cells during aging: correlation

    between lifespan and cycling activity. Blood89,

    15431550 (1997).

    27. de Haan, G. & Van Zant, G. Dynamic changes in mouse

    hematopoietic stem cell numbers during aging. Blood

    93, 32943301 (1999).

    28. Harrison, D. E. Mouse erythropoietic stem cell lines

    function normally 100 months: loss related to number

    of transplantations. Mech. Ageing Dev. 9, 427433

    (1979).

    29. Chen, J., Astle, C. M. & Harrison, D. E. Genetic

    regulation of primitive hematopoietic stem cell

    senescence. Exp. Hematol.28, 442450 (2000).

    30. Kamminga, L. M. et al. Impaired hematopoietic stemcell functioning after serial transplantation and during

    normal aging. Stem Cells23, 8292 (2005).

    31. Liang, Y., Van Zant, G. & Szilvassy, S. J. Effects of aging

    on the homing and engraftment of murine

    hematopoietic stem and progenitor cells. Blood106,

    14791487 (2005).

    32. Wang, Y., Schulte, B. A., Larue, A. C., Ogawa, M. &

    Zhou, D. Total body irradiation selectively induces

    murine hematopoietic stem cell senescence. Blood

    107, 358366 (2006).

    An important study showing persistent proliferative

    defects and p16INK4a expression in HSCs after

    exposure to DNA-damaging agents.

    33. Meng, A., Wang, Y., Van Zant, G. & Zhou, D. Ionizing

    radiation and busulfan induce premature senescence

    in murine bone marrow hematopoietic cells. Cancer

    Res. 63, 54145419 (2003).

    34. Ito, K. et al. Regulation of oxidative stress by ATM is

    required for self-renewal of haematopoietic stem cells.

    Nature431, 9971002 (2004).35. Ito, K. et al. Reactive oxygen species act through p38

    MAPK to limit the lifespan of hematopoietic stem cells.

    Nature Med.12, 446451 (2006).

    References 34 and 35 suggest roles for

    senescence-promoting molecules in HSC lifespan in

    the setting of impaired ATM function and increased

    ROS production.

    36. Boccadoro, M. et al. Oral melphalan at diagnosis

    hampers adequate collection of peripheral blood

    progenitor cells in multiple myeloma. Haematologica

    87, 846850 (2002).

    37. Knudsen, L. M., Rasmussen, T., Jensen, L. &

    Johnsen, H. E. Reduced bone marrow stem cell pool

    and progenitor mobilisation in multiple myeloma after

    melphalan treatment. Med. Oncol. 16, 245254

    (1999).38. Gardner, R. V., Astle, C. M. & Harrison, D. E.

    Hematopoietic precursor cell exhaustion is a cause of

    proliferative defect in primitive hematopoietic stem

    cells (PHSC) after chemotherapy. Exp. Hematol. 25,

    495501 (1997).39. Xing, Z. et al. Increased hematopoietic stem cell

    mobilization in aged mice. Blood108, 21902197

    (2006).

    40. Kuhn, H. G., Dickinson-Anson, H. & Gage, F. H.

    Neurogenesis in the dentate gyrus of the adult rat:

    age-related decrease of neuronal progenitor

    proliferation. J. Neurosci. 16, 20272033 (1996).41. Wong, K. K. et al. Telomere dysfunction and ATM

    deficiency compromises organ homeostasis and

    accelerates ageing. Nature421, 643648 (2003).

    An important study showing that the effects of

    ATM loss seen in humans can be reproduced in

    mice if ATM deficiency is combined with telomere

    dysfunction, which implies that the ataxia

    telangiectasia syndrome partly results from

    telomere dysfunction in ATM-deficient cells.

    42. Maslov, A. Y., Barone, T. A., Plunkett, R. J. &

    Pruitt, S. C. Neural stem cell detection,

    characterization, and age-related changes in the

    subventricular zone of mice. J. Neurosci. 24,

    17261733 (2004).43. Molofsky, A. V. et al. Increasing p16INK4a expression

    decreases forebrain progenitors and neurogenesis

    during ageing. Nature443, 448452 (2006).

    Shows that NSCs demonstrate a decrease in

    replicative function with ageing that is decreased in

    the setting of p16INK4a, implying that p16INK4a

    activation contributes to NSC ageing.44. Enwere, E. et al. Aging results in reduced epidermal

    growth factor receptor signaling, diminished olfactory

    neurogenesis, and deficits in fine olfactory

    discrimination. J. Neurosci. 24, 83548365 (2004).

    A careful, functional description of NSC ageing.45. Nishimura, E. K., Granter, S. R. & Fisher, D. E.

    Mechanisms of hair graying: incomplete melanocyte

    stem cell maintenance in the niche. Science307,

    720724 (2005).

    An excellent study suggesting that greying, a

    paradigmatic ageing phenotype, appears to result

    from a defect in melanocyte stem-cell maintenance

    with ageing.

    46. Yoon, K. H. et al. Selective -cell loss and -cellexpansion in patients with type 2 diabetes mellitus in

    Korea. J. Clin. Endocrinol. Metab.88, 23002308

    (2003).

    47. Butler, A. E. et al.-cell deficit and increased -cellapoptosis in humans with type 2 diabetes. Diabetes

    52, 102110 (2003).

    48. Meier, J. J. et al. Direct evidence of attempted cellregeneration in an 89-year-old patient with recent-

    onset type 1 diabetes. Diabetologia49, 18381844

    (2006).

    49. Krishnamurthy, J. et al. p16INK4a induces an age-dependent decline in islet regenerative potential.

    Nature443, 453457 (2006).

    This study, similar to reference 43, shows that

    p16INK4a loss can ameliorate an ageing phenotype,

    in this case in the pancreatic islet.

    50. Conboy, I. M. et al. Rejuvenation of aged progenitor

    cells by exposure to a young systemic environment.

    Nature433, 760764 (2005).

    Using parabiosis, this study provides striking

    evidence that muscle satellite-cell ageing is cell

    non-autonomous.

    51. Ju, Z. et al. Telomere dysfunction induces

    environmental alterations limiting hematopoietic stem

    cell function and engraftment. Nature Med.13,

    742747 (2007).

    This paper shows that telomere dysfunction limits

    HSC niche function, suggesting a cause of extrinsic

    ageing in HSCs.

    R E V I E W S

    NATuRe RevIeWS |molecular cell biology vOLuMe 8 | SePTeMbeR 2007 |711

    f oc uS on a g E I n g

  • 8/7/2019 How stem cells age and why

    10/11

    52. Kirkwood, T. B. Understanding the odd science of

    aging. Cell120, 437447 (2005).53. Navarro, S. et al. Hematopoietic dysfunction in a

    mouse model for Fanconi anemia group D1.

    Mol. Ther.14, 525535 (2006).

    54. Reese, J. S., Liu, L. & Gerson, S. L. Repopulating

    defect of mismatch repair-deficient hematopoietic

    stem cells. Blood102, 16261633 (2003).

    55. Prasher, J. M. et al. Reduced hematopoietic reserves

    in DNA interstrand crosslink repair-deficient Ercc1/

    mice. EMBO J.24, 861871 (2005).

    56. Nijnik, A. et al. DNA repair is limiting forhaematopoietic stem cells during ageing. Nature447,

    686690 (2007).

    57. Morales, M. et al. The Rad50S allele promotes

    ATM-dependent DNA damage responses and

    suppresses ATM deficiency: implications for the Mre11

    complex as a DNA damage sensor. Genes Dev. 19,

    30433054 (2005).

    A striking finding showing that an engineeredRad50Sallele, which is associated with bone

    marrow hypoplasia, can be rescued by ATM

    deficiency. This result indicates that the Rad50S

    allele is hypermorphic, and suggests that activation

    of the DNA-damage response per secan lead to

    stem-cell dysfunction.

    58. Rossi, D. J. et al. Deficiencies in DNA damage repair

    limit the function of haematopoietic stem cells with

    age. Nature447, 725729 (2007).

    An interesting study showing the accumulation of

    H2AX foci in HSCs with murine ageing, and an

    intrinsic role for several DNA-repair mechanisms in

    HSCs to prevent stem-cell ageing of this

    compartment.59. Rudolph, K. L. et al. Longevity, stress response, and

    cancer in aging telomerase-deficient mice. Cell96,

    701712 (1999).

    Classical study that established the importance of

    telomere maintenance in age-related processes

    and lifespan in mammals.

    60. Ruzankina, Y. et al. Deletion of the developmentally

    essential gene Atrin adult mice leads to age-related

    phenotypes and stem cell loss. Cell Stem Cell1,

    113126 (2007).

    A recent demonstration that homeostatic

    proliferation, in the absence of external

    DNA-damaging agents, can promote stem-cell

    dysfunction and ageing.61. Brown, E. J. & Baltimore, D. ATR disruption leads to

    chromosomal fragmentation and early embryonic

    lethality. Genes Dev. 14, 397402 (2000).62. Blasco, M. A. et al. Telomere shortening and tumor

    formation by mouse cells lacking telomerase RNA.Cell91, 2534 (1997).

    63. Lee, H. W. et al. Essential role of mouse telomerase in

    highly proliferative organs. Nature392, 569574

    (1998).

    64. Herrera, E. et al. Disease states associated with

    telomerase deficiency appear earlier in mice with

    short telomeres. EMBO J.18, 29502960

    (1999).

    65. Allsopp, R. C., Morin, G. B., DePinho, R., Harley, C. B.

    & Weissman, I. L. Telomerase is required to slow

    telomere shortening and extend replicative lifespan of

    HSCs during serial transplantation. Blood102,

    517520 (2003).

    This paper shows that telomere dysfunction can

    limit HSC lifespan in serial transplantation.

    66. Choudhury, A. R. et al. Cdkn1a deletion improves

    stem cell function and lifespan of mice with

    dysfunctional telomeres without accelerating cancer

    formation. Nature Genet. 39, 99105 (2007).

    This paper demonstrates that p21CIP is notrequired for the anti-cancer effects of p53

    mediated in response to telomere dysfunction, but

    is important for the pro-ageing effects of p53 in

    HSCs and other stem cells in response to telomere

    dysfunction.

    67. Chin, L. et al. p53 deficiency rescues the adverse

    effects of telomere loss and cooperates with telomere

    dysfunction to accelerate carcinogenesis. Cell97,

    527538 (1999).

    68. Artandi, S. E. et al. Telomere dysfunction promotes

    non-reciprocal translocations and epithelial cancers in

    mice. Nature406, 641645 (2000).

    References 67 and 68 provide insights into the

    mechanisms that underlie the intimate link

    between cancer and ageing, particularly why aged

    individuals develop epithelial cancers and why such

    cancers emerge with radically altered cytogenetic

    profiles.

    69. Kamijo, T. et al. Tumor suppression at the mouse

    INK4a locus mediated by the alternative reading

    frame product p19ARF.Cell91, 649659 (1997).

    70. Stein, G. H., Drullinger, L. F., Soulard, A. & Dulic, V.

    Differential roles for cyclin-dependent kinase inhibitors

    p21 and p16 in the mechanisms of senescence and

    differentiation in human fibroblasts. Mol. Cell. Biol.

    19, 21092117 (1999).

    71. Alcorta, D. A. et al. Involvement of the cyclin-

    dependent kinase inhibitor p16 (INK4a) in replicative

    senescence of normal human fibroblasts. Proc. Natl

    Acad. Sci. USA 93, 1374213747 (1996).72. Campisi, J. Suppressing cancer: the importance of

    being senescent. Science309, 886887 (2005).

    73. Kim, W. Y. & Sharpless, N. E. The regulation of INK4/

    ARF in cancer and aging. Cell127, 265275 (2006).74. Sone, H. & Kagawa, Y. Pancreatic cell senescence

    contributes to the pathogenesis of type 2 diabetes in

    high-fat diet-induced diabetic mice. Diabetologia48,

    5867 (2005).

    75. Krishnamurthy, J. et al. Ink4a/Arf expression is a

    biomarker of aging. J. Clin. Invest. 114, 12991307

    (2004).

    76. Edwards, M. G. et al. Gene expression profiling of

    aging reveals activation of a p53-mediated

    transcriptional program. BMC Genomics8, e80

    (2007).

    77. Janzen, V. et al. Stem-cell ageing modified by the

    cyclin-dependent kinase inhibitor p16INK4a. Nature

    443, 421426 (2006).

    78. Chen, J., Astle, C. M. & Harrison, D. E. Hematopoietic

    senescence is postponed and hematopoietic stem cell

    function is enhanced by dietary restriction. Exp.

    Hematol. 31, 10971103 (2003).

    A provocative description of the effects of caloric

    restriction on HSC function with ageing.

    79. Chen, Q. & Ames, B. N. Senescence-like growth arrest

    induced by hydrogen peroxide in human diploid

    fibroblast F65 cells. Proc. Natl Acad. Sci. USA91,

    41304134 (1994).80. Chen, J. H. et al. Loss of proliferative capacity and

    induction of senescence in oxidatively stressed human

    fibroblasts. J. Biol. Chem.279, 4943949446

    (2004).

    81. Takahashi, A. et al. Mitogenic signalling and the

    p16(INK4a)-Rb pathway cooperate to enforce

    irreversible cellular senescence. Nature Cell Biol. 8,

    12911297 (2006).

    82. Tothova, Z. et al. FoxOs are critical mediators of

    hematopoietic stem cell resistance to physiologic

    oxidative stress. Cell128, 325339 (2007).

    A detailed genetic study that establishes the

    importance of FOXO-dependent regulation ofintracellular ROS in the maintenance of HSCs.

    83. Stepanova, L. & Sorrentino, B. P. A limited role for

    p16Ink4a and p19Arf in the loss of hematopoietic stem

    cells during proliferative stress. Blood106, 827832

    (2005).

    Together with references 43, 49 and 77, this study

    suggests that p16INK4a loss can ameliorate a murine

    ageing phenotype, in this case in the HSCs.84. Tyner, S. D. et al. p53 mutant mice that display early

    ageing-associated phenotypes. Nature415, 4553

    (2002).85. Maier, B. et al. Modulation of mammalian life span by

    the short isoform of p53. Genes Dev. 18, 306319

    (2004).

    References 84 and 85 demonstrate that excess

    p53 activation can markedly accelerate the

    development of ageing-associated phenotypes in

    mice.86. Orsted, D. D., Bojesen, S. E., Tybjaerg-Hansen, A. &

    Nordestgaard, B. G. Tumor suppressor p53 Arg72Propolymorphism and longevity, cancer survival, and risk

    of cancer in the general population. J. Exp. Med. 204,

    12951301 (2007).

    87. Matheu, A. et al. Delayed ageing through damage

    protection by the Arf/p53 pathway. Nature448,

    375379 (2007).

    This paper challenges the view that increased p53

    activity is necessarily pro-ageing.

    88. Cheng, T. et al. Hematopoietic stem cell quiescence

    maintained by p21cip1/waf1. Science287,

    18041808 (2000).

    89. Melzer, D. et al. A common variant of the p16(INK4a)

    genetic region is associated with physical function in

    older people. Mech. Ageing Dev. 128, 370377

    (2007).

    90. Scott, L. J. et al. A genome-wide association study of

    type 2 diabetes in Finns detects multiple susceptibility

    variants. Science316, 13411345 (2007).

    91. Saxena, R. et al. Genome-wide association analysis

    identifies loci for type 2 diabetes and triglyceride

    levels. Science316, 13311336 (2007).

    92. Zeggini, E. et al. Replication of genome-wide

    association signals in UK samples reveals risk loci for

    type 2 diabetes. Science316, 13361341 (2007).

    93. Helgadottir, A. et al. A common variant on

    chromosome 9p21 affects the risk of myocardial

    infarction. Science316, 14911493 (2007).94. McPherson, R. et al. A common allele on chromosome

    9 associated with coronary heart disease. Science

    316, 14881491 (2007).95. The Wellcome Trust Case Control Consortium.

    Genome-wide association study of 14,000 cases of

    seven common diseases and 3,000 shared controls.

    Nature447, 661678 (2007).

    References 8995 describe the identification of

    9p21 SNPs near the INK4/ARFlocus as being

    associated with type 2 diabetes mellitus,

    atherosclerotic heart disease and frailty.96. Matthews, C. et al. Vascular smooth muscle cells

    undergo telomere-based senescence in human

    atherosclerosis: effects of telomerase and oxidative

    stress. Circ. Res. 99, 156164 (2006).

    97. Chimenti, C. et al. Senescence and death of primitive

    cells and myocytes lead to premature cardiac aging

    and heart failure. Circ. Res. 93, 604613 (2003).

    98. Urbanek, K. et al. Myocardial regeneration by

    activation of multipotent cardiac stem cells in ischemic

    heart failure. Proc. Natl Acad. Sci. USA102,

    86928697 (2005).

    99. Kleinjan, D. A. & van Heyningen, V. Long-range control

    of gene expression: emerging mechanisms and

    disruption in disease. Am. J. Hum. Genet. 76, 832

    (2005).

    100. Pasmant, E. et al. Characterization of a germ-line

    deletion, including the entire INK4/ARF locus, in a

    melanoma-neural system tumor family: identification

    of ANRIL, an antisense noncoding RNA whose

    expression coclusters with ARF. Cancer Res. 67,

    39633969 (2007).

    101. Vulliamy, T. et al. The RNA component of telomerase is

    mutated in autosomal dominant dyskeratosis

    congenita. Nature413, 432435 (2001).

    102. Yamaguchi, H. et al. Mutations in TERT, the gene for

    telomerase reverse transcriptase, in aplastic anemia.

    N. Engl. J. Med. 352, 14131424 (2005).103. Mitchell, J. R., Wood, E. & Collins, K. A telomerase

    component is defective in the human disease

    dyskeratosis congenita. Nature402, 551555 (1999).104. Tsakiri, K. D. et al. Adult-onset pulmonary fibrosis

    caused by mutations in telomerase. Proc. Natl Acad.

    Sci. USA104, 75527557 (2007).105. Armanios, M. Y. et al. Telomerase mutations in

    families with idiopathic pulmonary fibrosis. N. Engl. J.

    Med.356, 13171326 (2007).

    References 101105 are important human studies

    describing the role of telomere dysfunction in

    human syndromes such as aplastic anaemia and

    idiopathic pulmonary fibrosis.

    106. Kitada, T., Seki, S., Kawakita, N., Kuroki, T. & Monna, T.

    Telomere shortening in chronic liver diseases.

    Biochem. Biophys. Res. Commun. 211, 3339 (1995).

    107. Miura, N. et al. Progressive telomere shortening and

    telomerase reactivation during hepatocellular

    carcinogenesis. Cancer Genet. Cytogenet. 93, 5662

    (1997).

    108. Urabe, Y. et al. Telomere length in human liver

    diseases. Liver16, 293297 (1996).

    109. Wiemann, S. U. et al. Hepatocyte telomere shortening

    and senescence are general markers of human liver

    cirrhosis. FASEB J.16, 935942 (2002).

    110. Rudolph, K. L., Chang, S., Millard, M., Schreiber-Agus,N. & DePinho, R. A. Inhibition of experimental liver

    cirrhosis in mice by telomerase gene delivery. Science

    287, 12531258 (2000).

    111. Samani, N. J., Boultby, R., Butler, R., Thompson, J. R.

    & Goodall, A. H. Telomere shortening in

    atherosclerosis. Lancet358, 472473 (2001).

    112. Obana, N. et al. Telomere shortening of peripheral

    blood mononuclear cells in coronary disease patients

    with metabolic disorders. Intern. Med.42, 150153

    (2003).

    113. Tabori, U., Nanda, S., Druker, H., Lees, J. & Malkin, D.

    Younger age of cancer initiation is associated with

    shorter telomere length in LiFraumeni syndrome.

    Cancer Res.67, 14151418 (2007).

    In accordance with results in mice (see reference

    67), this provocative human study suggests that

    telomere dysfunction accelerates tumour formation

    in the setting of p53 insufficiency.

    R E V I E W S

    712 | SePTeMbeR 2007 | vOLuMe 8 www.n./ws/

    R E V I E W S

  • 8/7/2019 How stem cells age and why

    11/11

    114. Wu, X. et al. Telomere dysfunction: a potential cancer

    predisposition factor. J. Natl Cancer Inst. 95,

    12111218 (2003).

    115. Cawthon, R. M., Smith, K. R., OBrien, E.,

    Sivatchenko, A. & Kerber, R. A. Association between

    telomere length in blood and mortality in people aged

    60 years or older. Lancet361, 393395 (2003).

    116. Frenck, R. W. Jr., Blackburn, E. H. & Shannon, K. M.

    The rate of telomere sequence loss in human

    leukocytes varies with age. Proc. Natl Acad. Sci. USA

    95, 56075610 (1998).

    117. Hemann, M. T., Strong, M. A., Hao, L. Y. &Greider, C. W. The shortest telomere, not average

    telomere length, is critical for cell viability and

    chromosome stability. Cell107, 6777 (2001).

    118. Rane, S. G. et al. Loss of Cdk4 expression causes

    insulin-deficient diabetes and Cdk4 activation results

    in -islet cell hyperplasia. Nature Genet. 22,4452 (1999).

    119. Tsutsui, T. et al. Targeted disruption of CDK4 delays

    cell cycle entry with enhanced p27(Kip1) activity.

    Mol. Cell. Biol.19, 70117019 (1999).

    120. Nielsen, G. P. et al. Immunohistochemical survey of

    p16INK4Aexpression in normal human adult and infant

    tissues. Lab. Invest.79, 11371143 (1999).

    121. Molofsky, A. V. et al. Bmi-1 dependence distinguishes

    neural stem cell self-renewal from progenitor

    proliferation. Nature425, 962967 (2003).

    122. Bruggeman, S. W. et al. Ink4a and Arf differentially

    affect cell proliferation and neural stem cell self-

    renewal in Bmi1-deficient mice. Genes Dev.19,

    14381443 (2005).

    123. Molofsky, A. V., He, S., Bydon, M., Morrison, S. J. &

    Pardal, R. Bmi-1 promotes neural stem cell self-

    renewal and neural development but not mouse

    growth and survival by repressing the p16 Ink4a and

    p19Arf senescence pathways. Genes Dev. 19,

    14321437 (2005).

    References 121123 are careful characterizations

    of the effects of BMI1 loss on NSCs, and the role of

    p16INK4a and ARF in NSCs in the setting of BMI1

    deficiency.

    124. Jacobs, J. J., Kieboom, K., Marino, S., DePinho, R. A.

    & van Lohuizen, M. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and

    senescence through the ink4a locus. Nature397,

    164168 (1999).

    125. Park, I. K. et al. Bmi-1 is required for maintenance of

    adult self-renewing haematopoietic stem cells. Nature

    423, 302305 (2003).

    126. Oguro, H. et al. Differential impact of INK4a and ARF

    on hematopoietic stem cells and their bone marrow

    microenvironment in BMI1-deficient mice. J. Exp.

    Med.203, 22472253 (2006).

    References 125 and 126 are careful

    characterizations of the effects of BMI1 loss on

    HSCs, and the role of p16INK4a and ARF in the

    setting of BMI1 deficiency.

    AcknowledgementsWe thank E. Sahin, J.-H. Paik, T. Letai and K. Mohlke for criti-

    cal reading and advice on the manuscript. R.A.D. is a director,

    co-founder and scientific advisor of AVEO Pharmaceuticals,

    Inc. in Cambridge, Massachusetts, USA, and is an American

    Cancer Society Research Professor and an Ellison Medical

    Foundation Senior Scholar. This work was supported by grants

    from the Sidney Kimmel Foundation for Cancer Research

    (N.E.S.), the Ellison Medical Foundation (N.E.S. and R.A.D.),

    the American Federation of Aging Research (N.E.S.), the

    Burroughs Wellcome Fund (N.E.S.) and the US National

    Institutes of Health (R.A.D.). R.A.D. is supported by the LeBow

    Fund to Cure Myeloma and the Robert A. and Renee E. Belfer

    Foundation Institute for Innovative Cancer Science.

    Competing interests statement

    The authors declare no competing financial interests.

    DAtABAsesEtrez gee: http://www.ncbi.nlm.nih.gov/entrez/query.

    fcgi?db=gene

    Atr

    oMIM:http://www.ncbi.nlm.nih.gov/entrez/query.

    fcgi?db=OMIM

    ataxia telangiectasia |HutchinsonGilford syndrome |

    type 2 diabetes mellitus | Werner syndrome

    uiPrtKB:http://ca.expasy.org/sprot

    ATM| p15INK4b | p16INK4a |p53 | RB

    FURtHeR INFORmAtIONRld a. DePihs hmepe:http://www.dana-farber.

    org/abo/danafarber/detail.asp?PersonID=51&RD=True

    nrm E. Shrplesss hmepe:

    http://genetics.unc.edu/faculty/sharpless.htm

    all liNkS are active iN tHe oNliNe PdF.

    R E V I E W S

    NATuRe RevIeWS | molecular cell biology vOLuMe 8 | SePTeMbeR 2007 | 713

    f oc uS on a g E I n g

    http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=genehttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=genehttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=245000&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIMhttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIMhttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIMhttp://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=208900http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=176670http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=176670http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=125853http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=277700http://ca.expasy.org/sprothttp://www.expasy.org/uniprot/Q62388http://www.expasy.org/uniprot/P42772http://www.expasy.org/uniprot/P42771http://www.expasy.org/uniprot/P04637http://www.expasy.org/uniprot/P04637http://www.expasy.org/uniprot/P06400http://www.dana-farber.org/abo/danafarber/detail.asp?PersonID=51&RD=Truehttp://www.dana-farber.org/abo/danafarber/detail.asp?PersonID=51&RD=Truehttp://www.dana-farber.org/abo/danafarber/detail.asp?PersonID=51&RD=Truehttp://genetics.unc.edu/faculty/sharpless.htmhttp://genetics.unc.edu/faculty/sharpless.htmhttp://www.dana-farber.org/abo/danafarber/detail.asp?PersonID=51&RD=Truehttp://www.dana-farber.org/abo/danafarber/detail.asp?PersonID=51&RD=Truehttp://www.expasy.org/uniprot/P06400http://www.expasy.org/uniprot/P04637http://www.expasy.org/uniprot/P42771http://www.expasy.org/uniprot/P42772http://www.expasy.org/uniprot/Q62388http://ca.expasy.org/sprothttp://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=277700http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=125853http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=176670http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=208900http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIMhttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIMhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=245000&ordinalpos=1&itool=EntrezSystem2.PEntrez.Gene.Gene_ResultsPanel.Gene_RVDocSumhttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=genehttp://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene