BTG2 Antagonizes Pin1

download BTG2 Antagonizes Pin1

of 14

Transcript of BTG2 Antagonizes Pin1

  • 8/3/2019 BTG2 Antagonizes Pin1

    1/14

    BTG2 antagonizes Pin1 in response to mitogens andtelomere disruption during replicative senescence

    Keith Wheaton, Jennifer Muir, Weili Ma and Samuel

    BenchimolDepartment of Biology, York University, 4700 Keele Street,

    Toronto, Ontario, Canada M3J 1P3

    Summary

    Cellular senescence limits the replicative capacity of nor-

    mal cells and acts as an intrinsic barrier that protects

    against the development of cancer. Telomere shortening

    induced replicative senescence is dependent on the ATM-

    p53-p21 pathway but additional genes likely contribute

    to senescence. Here, we show that the p53-responsive

    gene BTG2 plays an essential role in replicative senes-cence. Similar to p53 and p21 depletion, BTG2 depletion

    in human fibroblasts leads to an extension of cellular life-

    span, and ectopic BTG2 induces senescence indepen-

    dently of p53. The anti-proliferative function of BTG2

    during senescence involves its stabilization in response to

    telomere dysfunction followed by serum-dependent

    binding and relocalization of the cell cycle regulator prol-

    yl isomerase Pin1. Pin1 inhibition leads to senescence in

    late-passage cells, and ectopic Pin1 expression rescues

    cells from BTG2-induced senescence. The neutralization

    of Pin1 by BTG2 provides a critical mechanism to maintain

    senescent arrest in the presence of mitogenic signals in

    normal primary fibroblasts.

    Key words: BTG2; p53; Pin1; replicative; senescence;

    telomeres.

    Introduction

    In response to many forms of cellular stress, the p53 tumor sup-

    pressor protein becomes active and is able to transactivate a

    variety of target genes that regulate diverse cellular processes

    including: cell cycle progression, senescence, DNA repair,

    metabolism and cell survival (reviewed in (Vousden & Lane,2007). Through these processes, p53 protects cells from uncon-

    trolled growth and genomic instability that lead to tumor devel-

    opment. The divergent biological outcomes of p53 are thought

    to be due to differential transcription of p53 target genes.Promoter selection is regulated by posttranslational modifica-

    tions of p53 including phosphorylation as well as by the interac-

    tion of p53 with various protein cofactors (Vousden, 2006;

    Rozan & El-Deiry, 2007).

    Primary human fibroblasts have a finite replicative lifespan

    that terminates with the acquisition of a phenotype having

    distinct morphological and biochemical characteristics termed

    replicative senescence (Hayflick, 1965). In this state, the cells

    accumulate primarily in the G1 phase of the cell cycle and

    remain viable but refractory to mitogenic signals. Thus, replica-

    tive senescence acts as an intrinsic barrier against unrestricted

    cell growth and provides a mechanism for tumor suppression.Recent studies indicate that senescence is as effective as apopto-

    sis in reducing cancer incidence and that senescence bypass is

    an important step in the development of cancer (Dimri, 2005;

    Collado et al., 2007). At the molecular level, senescent cells

    exhibit elevated expression of p21, p16 and cyclin D1, and

    increased activity of p53 (Atadja et al., 1995; Vaziri et al.,

    1997), Rb (Stein et al., 1999) and PKC d (Wheaton & Riabowol,

    2004). Replicative senescence is triggered by critically short telo-

    meres. Telomeres are specialized nucleoprotein complexes that

    cap and protect the ends of linear chromosomes (Verdun & Karl-

    seder, 2007). Telomeres shorten with each round of DNA repli-

    cation because of the end-replication problem the inability of

    DNA polymerases to completely replicate the 3 end of linear

    DNA molecules (Harley et al., 1990). Shortening of telomeric

    DNA leads to uncapping of the telomeres, and this is believed to

    initiate an ATM-dependent DNA damage response that acti-

    vates p53 (Karlseder et al., 2002; Herbig et al., 2004; Stewart &

    Weinberg, 2006).

    The p53 protein has been implicated as one of the key media-

    tors of cellular senescence. In its absence, the replicative capac-

    ity of primary fibroblasts is extended 10-30 population

    doublings (Hara et al., 1991; Shay et al., 1991; Masutomi et al.,

    2003). Cells that bypass senescence as a result of p53 repression

    undergo further rounds of cell division even though they con-

    tinue to lose telomeric DNA and eventually encounter a secondblock in proliferation known as crisis characterized by massive

    cell death. The identity of the transcriptional targets of p53

    required to initiate and maintain the senescence phenotype is

    uncertain. Although the p53 target gene, p21WAF1, was origi-

    nally considered to be central to senescence arrest (el-Deiry

    et al., 1993; Noda et al., 1994; Brown et al., 1997), subsequent

    studies have questioned whether the protein is essential (Wyllie

    et al., 2003) or if it is sufficient alone (Ma et al., 1999; Dulic

    et al., 2000). Recently, PAI-1 was identified as a p53-responsive

    Correspondence

    Keith Wheaton and Sam Benchimol, Department of Biology, York Univer-

    sity, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada. Tel.:

    +1 416 736 2100 Ext. 20893 (Keith Wheaton); +1 416 736 2100 Ext.

    20726 (Sam Benchimol); fax: +1 416 736 5698; e-mails: kwheaton@

    yorku.ca and [email protected]

    Accepted for publication 6 June 2010

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    747

    Aging Cell(2010) 9, pp747760 Doi: 10.1111/j.1474-9726.2010.00601.x

    Aging

    Cell

  • 8/3/2019 BTG2 Antagonizes Pin1

    2/14

    gene that contributes to replicative senescence through its abil-

    ity to inhibit the urokinase-type plasminogen activator, leading

    to downregulation of PI(3)K-PKB signaling (Kortlever et al.,

    2006). Thus, only two p53-responsive genes have been charac-

    terized in the senescence program, neither of which can fully

    recapitulate p53-dependent senescence. Therefore, we wished

    to address the question of what other effector genes were

    required for p53-dependent replicative senescence. Wescreened p53 effectors previously described as being involved in

    cellular arrest and identified BTG2 and GAdd45a as being highly

    upregulated during senescence. Further functional and mecha-

    nistic analysis proved that BTG2 had a significant role in the

    development of senescence.

    BTG2 is a member of the BTGTob family of anti-proliferative

    genes and has been implicated in various cellular processes

    including cell cycle progression, differentiation and apoptosis.

    BTG2 was previously reported to be an effector of p53-depen-

    dent proliferation arrest (Rouault et al., 1996) and to act through

    repression of cyclin D1 and cyclin E1 mRNA (Guardavaccaro

    et al., 2000; Boiko et al., 2006; Kawakubo et al., 2006). BTG2 isinduced by various cellular stresses through p53-dependent and

    p53-independent processes (Lim, 2006). The peptidyl-prolyl

    isomerase Pin1 regulates diverse cellular processes including cell

    cycle progression and apoptosis through its interaction with dif-

    ferent phosphoproteins altering their conformation and stability

    (Yeh & Means, 2007). Two notable studies reported that Pin1

    was required for efficient reentry into the cell cycle in response

    to mitogenic stimulation after G0 arrest (Fujimori et al., 1999;

    You et al., 2002). Furthermore, Pin1 overexpression has been

    correlatedwith oncogenesis (Yeh & Means, 2007).

    Here, we show that BTG2 plays a critical role in promoting

    p53-dependent replicative senescence in human cells through

    its ability to sequester Pin1. This mechanism is unique to primary

    cells, because BTG2 has been commonly reported to suppress

    cyclin D1 levels in transformed cell lines rather than neutralize

    Pin1-mediated cell cycle progression. BTG2 activity during

    senescence is regulated at three levels: transcriptional regulation

    by p53, protein stabilization in response to telomere disruption

    and by mitogenic signaling pathways. Thus, we have further

    explored the p53 genetic program leading to senescence by

    characterizing the role of BTG2.

    Results

    Expression of p53-responsive genes in senescence

    To investigate the expression of p53 effector genes during cellu-

    lar senescence, we measured the transcript levels of p53-

    responsive genes previously associated with cellular arrest

    including BTG2, TOB1, 14-3-3r , Gadd45a , Reprimo and

    p21WAF1. We measured mRNA expression by semi-quantitative

    RTPCR (Fig. 1A) in young, cycling Hs68 human fibroblasts at a

    mean population doubling (MPD) of 45 and in senescent Hs68

    cells (MPD 85). The mRNA levels for BTG2, GADD45a and p21

    were elevated during replicative senescence. This upregulation

    was not due to cellular quiescence, since these gene transcripts

    were not elevated during contact inhibition or serum starvation,

    two conditions known to activate p53 (Itahana et al., 2002;

    Meerson et al., 2004) (Fig. 1A). We confirmed the upregulation

    of BTG2, Gadd45a and p21expression during the development

    of senescence by Northern blotting (Fig. 1B) and Western immu-

    noblot analysis (Fig. 1C). These results are consistent with previ-

    ous reports, indicating that p21 (Noda et al., 1994), Gadd45a

    (Jackson & Pereira-Smith, 2006) and BTG2 (Rouault et al., 1996)

    are upregulated during replicative senescence. The increased

    levels of cyclin D1 served as an additional molecular marker for

    cellular senescence (Dulic et al., 1993; Lucibello et al., 1993).

    Extension of cellular lifespan by shRNA-mediated

    repression of p53, BTG2 and p21

    To investigate the dependency of BTG2, Gadd45a and p21

    expression on p53 during replicative senescence, we inhibited

    p53 expression in BJ and Hs68 fibroblasts using shRNA

    (Figs 2A,B and S1). p53 depletion suppressed the induction of

    BTG2, p21 and Gadd45a that is normally seen as BJ cells enter

    senescence (Fig. 2A,B). p53 shRNA-expressing cells (shp53 cells)

    also failed to induce p21 and BTG2 in response to doxorubicin

    treatment (Fig. S1A) or c-irradiation (Fig. S1C). As a conse-

    quence of sustained p53 inhibition, both BJ and Hs68 cells

    escaped senescence and grew an additional 10 MPDs compared

    with control cells expressing pSuper vector (Figs 2C and S1B).

    Hs68 and BJ fibroblast cell strains were derived from newborn

    human foreskin; we have used both cell strains throughout this

    study to confirm and validate our findings and have not

    detected any differences in their molecular characterization or

    behavior in culture.

    To assess the contribution of BTG2, p21 and Gadd45a to thesenescence program, we inhibited their expression using shRNA

    and developed stable shRNA-expressing BJ cells (shBTG2,

    shp21, and shGadd45a cells). The shBTG2 and shGadd45a cells

    failed to show an increase in BTG2 and Gadd45a protein,

    respectively, at the initiation of senescence and the shp21 cells

    exhibited a reduction in p21 protein induction at senescence

    (Fig. 2A,B). The replicative potential of these cells was compared

    with BJshp53 and BJpSuper control cells. The shp21 and

    shBTG2 cells exhibited increased proliferative potential com-

    pared with control cells but the extended lifespan was not as

    great as the shp53 cells (Fig. 2C). BJ cells expressing shRNA to

    both BTG2 and p21 exhibited little if any induction of BTG2 andp21 protein and had an extended lifespan similar to the shp21

    cells (Fig. 2C). Thus, inhibition of p21 or BTG2 individually or

    together could not fully mimic the loss of p53 with respect to

    proliferative potential. The incomplete inhibition of p21 and

    BTG2 compared with the efficacy of the shRNA against p53,

    however, could account for the differences in replicative poten-

    tial of these cells. shGadd45a cells did not exhibit an extended

    lifespan and underwent senescence at the same time as

    the BJpSuper control cells even though Gadd45a was effec-

    tively repressed. This suggested that Gadd45a does not play a

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    748

  • 8/3/2019 BTG2 Antagonizes Pin1

    3/14

    significant role in replicative senescence, and as a result,

    Gadd45a was not investigated further in this study.

    In the course of these studies, we noted that the BJshp53

    cells were not immortal (Fig. 2C) and that many cells exhibited

    an apoptotic morphology that was consistent with entry into

    crisis (data not shown). In contrast, the shp21, shBTG2 and

    shp21

    shBTG2 cells exhibited a senescence phenotype uponreaching their replicative limit (data not shown).

    Reinstatement of senescence in shp53 cells by

    ectopic expression of p53RR, BTG2 or p21

    The ability of shp53 cells to bypass replicative senescence sug-

    gests that one or more p53-responsive genes are required to ini-

    tiate the senescence program. To determine whether ectopic

    expression of p21, BTG2 or p53 would reinstate the senescence

    program in BJshp53 cells that had bypassed their normal repli-

    cative endpoint, cDNAs encoding BTG2, p21 and p53RR (resis-

    tant to p53 shRNA) were introduced individually by retroviral

    infection at MPD 72. Western blot analysis revealed that each of

    the cDNAs was expressed in the infected BJshp53 cells and that

    p53RR promoted expression of endogenous p21 and BTG2

    (Fig. 3A). Moreover, ectopic p53 was phosphorylated on Ser15,

    which serves as a marker for p53 activation (Fig. 3A). Impor-tantly, ectopic expression of p21, BTG2 and p53RR led to the

    establishment of senescence as measured by the acidic b-galac-

    tosidase assay and by the accumulation of cells in the G1 phase

    of the cell cycle (Fig. 3B). Similarly, in Hs68shp53 fibroblasts

    that had bypassed senescence as a result of p53 repression,

    ectopic BTG2 resulted in cell cycle arrest in G1 and b-galactosi-

    dase staining indicative of senescence (Fig. 3C). These results

    indicate that ectopic expression of BTG2, p21or p53 can

    promote senescence in cells that have escaped their normal

    replicative endpoint.

    (A)

    (B) (C)

    Fig. 1 Expression of p53 target genes in senescent cells. (A) Expression of various p53 target genes was assessed by semi-quantitative RTPCR. RNA was

    extracted from Hs68 fibroblasts that were cycling (MPD 45), contact inhibited (2 weeks), serum starved (48 h) or senescent (MPD 85). RNA levels were normalized

    using GAPDH as an internal control. Means and SD of three independent experiments are shown in the histogram. (B, C) Northern blot analysis (B) and Western

    blot analysis (C) of p53 target genes; cyclin D1 and p53s15 serve as markers of senescence. RNA and protein samples were obtained from Hs68 fibroblasts that

    were cycling (MPD 47), or 728 days afterthe initiation of senescence at MPD 85. The last lane shown in (B) represents RNA obtained from cycling Hs68 cells 3 h

    after c-irradiation with 6 Gy. MPD, mean population doubling.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    749

  • 8/3/2019 BTG2 Antagonizes Pin1

    4/14

    Sustained expression of p21 was recently shown to induce an

    irreversible senescent-like state in early-passage human fibro-

    blasts (Sang et al., 2008). To determine whether BTG2 and p53

    could also promote premature senescence in early-passage

    fibroblasts, we introduced BTG2 and p53RR cDNAs into young

    cells at passage numbers that were well below their established

    proliferative limit, BJshp53 cells at MPD 63 or Hs68shp53 cells

    at MPD 52. Remarkably, neither BTG2 (Fig. 3D,E) nor p53RR

    (Fig. 3D) promoted senescence or inhibited the growth of early-

    passage fibroblasts.

    To investigate the inability of p53 to promote growth arrest in

    early-passage cells, we measured the amount of ectopic p53

    and its phosphorylation by Western blotting in early-passage

    BJshp53 cells (MPD 65) and in late-passage BJshp53 cells

    (MPD 72). The amount of ectopic p53 protein was similar at

    both passage numbers but p53 phosphorylation on Ser15 was

    only observed at late passage (Fig. 3F), suggesting that p53 is

    functionally inactive in early-passage cells. One important differ-

    ence between shp53 cells at early and late passage is the pres-

    ence of intact protected telomeres in the former and short

    disrupted telomeres in the latter. Although shp53 cells bypass

    senescence because they lack p53, these cells continue to lose

    telomeric DNA until they enter crisis. Thus, one interpretation of

    these results is that p53RR is inactive in unstressed low-passage

    cells when telomeres are intact and capped; at high passage

    number, p53RR is posttranslationally activated by ATM-medi-ated signals emanating from disrupted short telomeres.

    The level of ectopic BTG2 protein was low in early-passage

    Hs68pSuper and Hs68shp53 cells but increased in the pres-

    ence of the proteasome inhibitor MG132 (Fig. 3G), suggesting

    that BTG2 protein is normally unstable at early passages. This is

    consistent with a previous report showing that BTG2 is rapidly

    degraded through the ubiquitin proteasomal pathway (Sasajima

    et al., 2002). We expressed BTG2 cDNA in BJshp53 cells before

    (MPD 65) and after (MPD 72) their normal replicative limit, and

    observed higher levels of BTG2 at MPD 72 compared with MPD

    65 (Fig. 3H). The higher level of BTG2 protein at MPD 72 was not

    the result of differences in the level of ectopic BTG2 mRNA

    (Fig. 3H). As shown earlier in Fig. 2, p53 depletion prevents the

    induction of endogenous BTG2 mRNA in these cells. Together,

    these results suggest that BTG2 expression is regulated in senes-

    cent or late-passage cells through two distinct mechanisms: one

    is dependent on p53 and involves transcriptional regulation; the

    other is independent of p53and involves protein stabilization. As

    BTG2 is stable and functional only in late-passage cells, it is possi-

    ble that telomere disruption is also required for BTG2 function.

    BTG2 is required for T-oligo-induced senescence

    To test the idea that the anti-proliferative effect of BTG2 isdependent on disrupted telomeres, we used an experimental

    model in which exogenous oligonucleotides with a sequence

    identical to the telomere 3-overhang sequence (T-oligo) are

    used to induce senescence artificially in human fibroblasts.

    Fibroblasts exposed to the T-oligo were reported to undergo

    p53-mediated senescence through a process that resembles the

    natural uncapping of telomeres in replicative senescence (Li

    et al., 2003). T-oligo treatment induced senescence in BJpSu-

    per control fibroblasts, but not in shp53, shp21 and shBTG2 BJ

    cells as determined by staining for acidic b-galactosidase

    (Fig. 4A) and by cell cycle analysis for G1 arrest (Fig. 4B). Cells

    treated with a control oligonucleotide (C-oligo) containing asequence complementary to the T-oligo showed no b-galactosi-

    dase staining or G1 arrest (Fig. 4A,B). To support these results,

    BJpSuper fibroblasts were treated with T-oligo or C-oligo and

    were analyzed over a 2 -week period for the expression of p21,

    BTG2 and p53 by Western blotting. The levels of these three

    proteins increased initially in response to T-oligo but not C-oligo

    treatment, and the amount of BTG2 and p21 remained elevated

    2 weeks after T-oligo treatment (Fig. 4C). The accumulation of

    BTG2 protein upon T-oligo treatment was also seen by immuno-

    fluorescence microscopy (Fig. S2). The amount of p53 protein

    (C)(A)

    (B)

    Fig. 2 p53, p21 and BTG2 shRNA-expressing cells have an extended proliferative lifespan. (A, B) Western blot analysis of BJ cells expressing various shRNAs

    directed to p53 and its targets p21, BTG2 and GADD45a at MPD 65 (cycling, presenescent) and at later MPDs as indicated. pSuper represents BJ cells transfected

    with the empty pSuper vector. (C) Measurement of the number of mean population doublings over the lifespan of BJ cells expressing various shRNAs as indicated.

    For each shRNA-expressing cell strain, the mean SD of three independent cultures is shown. See also Fig. S1. MPD, mean population doubling.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    750

  • 8/3/2019 BTG2 Antagonizes Pin1

    5/14

    returned to basal level over a 2 -week period but the activated,

    Ser15 phosphorylated form of p53 remained elevated in T-oli-

    go-treated BJ cells. These results indicate a role of p21 and BTG2

    in the p53-dependent senescence pathway induced naturally bytelomere shortening or artificially by treatment with T-oligo.

    Next, we asked whether ectopic BTG2 could sensitize shp53

    cells to undergo T-oligo-induced senescence. BTG2-expressing

    Hs68shp53 cells at MPD 52 exhibited a senescent morphology

    and stained for b-galactosidase upon addition of T-oligo but not

    C-oligo (Fig. 4D). Notably, the level of ectopic BTG2 protein

    in the shp53 cells increased upon T-oligo treatment (Fig. 4E). In

    addition, cyclin D1 levels increased upon T-oligo treatment in

    BTG2-expressing shp53 cells, providing further support for entry

    into senescence. Residual p53 activity is unlikely to contribute to

    senescence induced by BTG2 in shp53 cells because we do not

    detect the Ser15 phosphorylated form of p53 nor p21 induction

    in these cells after T-oligo treatment (Fig. 4E) or after DNA dam-

    age (Fig. S1A,C). These data support the view that uncappedtelomeres represent the senescent-specific signal that promotes

    BTG2 stabilization and anti-proliferative activity.

    BTG2 binds and colocalizes with Pin1 in senescent

    fibroblasts

    Having found that BTG2 is a key effector of telomere-dependent

    replicative senescence and T-oligo-induced senescence, we next

    wanted to identify downstream effectors. Previous reports

    suggested that BTG2 represses cyclin D1 (Guardavaccaro et al.,

    (A) (B)

    (C)(D) (E)

    (F) (G) (H)

    Fig. 3 BTG2, p21 and p53 can reinstate senescence in p53 shRNA-expressing cells. (A) p53 shRNA-expressing BJ cells (BJshp53) at MPD 72 were infected with

    the indicated retroviral expression constructs or with the empty pBabe vector. Western blot analysis was performed with the indicated antibodies 1 week afterinfection. p53RR represents a p53 retroviral construct that is resistant to p53 shRNA. b-actin serves as a loading control. (B) Representative cell cycle profiles

    obtained by flow cytometry after propidium iodide staining of BJshp53 cells 2 weeks after infection with the indicated retroviral constructs. The G1S ratios were

    determined from the cell cycle profiles of cells from four independent viral infections. Staining for senescence-associatedb-galactosidase was performed on

    BJshp53 cells 2 weeks after retroviral overexpression of BTG2, p21 or p53RR. (C, D) Cell cycle profiles and b-galactosidase staining of p53 shRNA-expressing Hs68

    cells (Hs68shp53) at MPD 90 and BJshp53 cells at MPD 63. (E) Growth curve of Hs68pSuper or Hs68shp53 cells after retroviral expression of BTG2 or the

    pBabe.hygro control vector (MPD 57). Each cell strain was initially seeded at 50 000 cells and counted every day over a 6 -day period. (F) Western blot analysis of

    p53 and phospho-p53 S15 in BJshp53 infected with p53RR at MPD 65 and MPD 72. (G) Cells as in (E) were treated with MG132 for 1 h before harvesting for

    Western blot analysis of BTG2. (H) Ectopic BTG2 protein and RNA expression in BJshp53 cells after retroviral infection with a BTG2 cDNA expression constructs at

    MPD 65 or MPD 72. MPD, mean population doubling.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    751

  • 8/3/2019 BTG2 Antagonizes Pin1

    6/14

    2000; Boiko et al., 2006; Kawakubo et al., 2006) and cyclin E

    expression (Lim et al., 1998). We show, however, that cyclin D1

    is highly expressed and that cyclin E expression is unchanged

    during senescence (Figs 1B,C and 4E). Thus, it is unlikely that

    BTG2 promotes replicative senescence through suppression of

    cyclin D1 and cyclin E. BTG2 was also reported to serve as a reg-

    ulator of Pin1 nuclear export in transformed human cells (Hong

    et al., 2005). To test the possibility that BTG2 might sequester

    Pin1 in the cytoplasm of senescent cells and neutralize its func-tion, we expressed a mutant BTG2 protein (BTG2 S147A) that is

    unable to interact with Pin1 (Hong et al., 2005) in late-passage

    BJshp53 cells and tested whether this mutation prevented

    BTG2 from inducing senescence. Unlike wild-type BTG2, BTG2

    S147A was unable to promote senescence in shp53 cells that

    had bypassed their normal replicative limit (Fig. 3B). Next, we

    examined the interaction of BTG2 with Pin1 in young and senes-

    cent Hs68 cells by coimmunoprecipitationWestern blot analy-

    sis. As the binding of Pin1 to BTG2 requires ERK12-dependent

    phosphorylation of BTG2 on Ser147 (Hong et al., 2005), we

    deprived the cells of serum for 72 h and stimulated with serum

    for 24 h prior to cell harvesting. The levels of both Pin1 and

    BTG2 protein were elevated in senescent cells compared with

    young cells and these levels were unaffected by serum (Fig. 5A).

    BTG2 coimmunoprecipitated with Pin1 in both young and

    senescent cells, and the binding between these two proteins

    was greatly enhanced after serum stimulation (Fig. 5B).

    Together, these data indicate that BTG2 binds to Pin1 and they

    suggest that the interaction of these proteins could be impor-tant for the induction of senescence by BTG2.

    BTG2 promotes nuclear export of Pin1

    Next, we examined the expression and localization of endoge-

    nous Pin1 and BTG2 in young and senescent Hs68 cells by con-

    focal microscopy. We deprived the cells of serum for 72 h and

    stimulated with serum for 24 h prior to immunostaining. As

    expected, young cells had no detectable BTG2 protein

    expression and showed nuclear Pin1 staining regardless of

    (A) (D)

    (E)(B)

    (C)

    Fig. 4 BTG2 is required for T-oligo-induced senescence. (A, B) BJ cells expressing the indicated shRNAs were treated with a single dose of T-oligo (40 lM), C-oligo

    (40 lM), or vehicle (H2O). After 2 weeks, cells were fixed and stained for b-galactosidase (A) or propidium iodide and analyzed by flow cytometry (B). Cells were

    collected at similar densities to ensurethe measured growth arrest was not because of contact inhibition. TheG1S ratio was determined for three independentexperiments. (C) Western blot analysis of p53, p53 Ser-15, p21 and BTG2 in BJpSupercells after treatment with T-oligo, C-oligo or vehicle at the times indicated.

    (D) b-galactosidase staining of Hs68shp53 cells expressing BTG2 or empty pBabe vector, 2 weeks after treatment with T-oligo, C-oligo or vehicle at MPD 52. (E)

    Western blot analysis of p53 and BTG2 in Hs68shp53 expressing BTG2 after oligo treatment at the times indicated. MPD, mean population doubling.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    752

  • 8/3/2019 BTG2 Antagonizes Pin1

    7/14

    serum stimulation. In serum-starved senescent cells, Pin1 was

    expressed primarily in the nucleus and BTG2 was expressed dif-

    fusely in the nucleus and cytoplasm. Upon serum stimulation,

    however, 4555% of cells examined showed cytoplasmic stain-

    ing for Pin1 that colocalized with BTG2 (Fig. 5C). The cellular

    redistribution of Pin1 and its colocalization with BTG2 in the

    cytoplasm were also observed when cells were induced to

    undergo senescence using T-oligo (Fig. S2).

    To determine whether ectopic BTG2 affects the localization of

    endogenous Pin1, we expressed BTG2 and BTG2 S147A in late-

    passage BJshp53 cells and monitored expression by immuno-

    fluorescence microscopy (Fig. 6). Endogenous Pin1 is predomi-

    nantly nuclear in unstimulated cells, while ectopic BTG2 and

    BTG2 S147A were expressed diffusely in both the cytoplasm

    and the nucleus of BJshp53 cells. Upon serum stimulation of

    BTG2-expressing cells but not BTG2 S147A-expressing cells,

    Pin1 redistributed and colocalized with BTG2 in the cytoplasm.

    Nearly all serum-stimulated cells showed colocalization of BTG2

    and Pin1. In contrast, there was minimal colocalization of BTG2

    S147A with Pin1 (Fig. 6). These results suggest that BTG2 pro-

    motes senescence in shp53 cells through binding and nuclearexport of Pin1 and that the neutralization of Pin1 contributes to

    cellular senescence.

    Pin1 inhibition leads to senescence in p53

    knockdown cells that have bypassed their normal

    proliferative limit

    If the primary role of p53-induced BTG2 expression is to neutral-

    ize Pin1, we reasoned that Pin1 inhibition should lead to senes-

    cence in shp53 cells. Because these cells fail to induce BTG2

    expression (Fig. 2A) and have an extended lifespan, they provide

    a good experimental model to investigate Pin1 function in the

    absence of BTG2. Pin1 promotes S phase entry in response to

    mitogenic stimulation (Fujimori et al., 1999; You et al., 2002);

    hence, we cultured BJshp53 cells at MPD 67 or 73 in serum-

    free media for 48 h and stimulated with serum for 24 h in

    the absence or presence of the Pin1 inhibitor, diethyl-1,3,

    6,8-tetrahydro-1,3,6,8-tetraoxobenzo[lmn][3,8]phenanthroline-

    2,7-diacetate (PiB). Cell cycle progression was monitored by

    propidium iodide staining and flow cytometry. Unexpectedly,

    we observed that Pin1 was required for entry into S phase in the

    late-passage cells but not in the early-passage cells (Fig. 7A).

    PiB-treated late-passage cells (but not early-passage cells) accu-

    mulated in G0G1 and stained for b-galactosidase indicative of

    a senescence phenotype (Fig. 7B). Because pharmacological

    inhibition of Pin1 with PiB could have nonspecific or off-target

    effects, we used shRNA specific for Pin1 to knockdown its

    expression. shRNA-mediated knockdown of Pin1 reduced the

    level of endogenous Pin1 in early- (MPD 63) and late-passage

    (MPD 73) BJshp53 cells (Fig. 7C). Pin1 knockdown did not

    decrease the proliferative activity of early- or late-passageBJshp53 cells in culture. Serum starvation of BJshp53 and

    BJshp53shPin1 cells at early and late passage resulted in

    G0G1 arrest without any evidence of b-galactosidase staining

    even after 2 weeks in serum-free media (Fig. 7D, upper panel).

    Upon serum stimulation, however, only the late-passage

    BJshp53shPin1 cells failed to enter S phase and stained for

    b-galactosidase (Fig. 7D, lower panel). Hence, Pin1 inhibition

    using PiB or shRNA reveals a role of Pin1 in promoting S phase

    entry only in late-passage human fibroblasts. Moreover, late-

    passage shp53 cells that are unable to enter S phase from

    (A)

    (B)

    (C)

    Fig. 5 BTG2 binds and colocalizes with Pin1 in senescent fibroblasts. (A and B) CoimmunoprecipitationWestern blot analysis of Pin1 and BTG2 in young (MPD

    52) and senescent Hs68 fibroblasts (MPD 84). Cells were serum starved (SS) for 72 h or serum stimulated for 24 h. Panel A shows total levels of Pin1 and BTG2 byWestern blot analysis and panel B shows the coimmunoprecipitationWestern blot. (C) Immunofluorescence staining of Pin1 and BTG2 in Hs68 cells, young (MPD

    50) and senescent (MPD 82). Cells were serum starved for 72 h or serum stimulated for 24 h prior to fixation. 4555% ofcells examined showed cytoplasmic

    staining for Pin1 that colocalized with BTG2 after serum stimulation. Nuclear counterstaining used Draq5. Images show a single optical layer of a confocal image

    at 400 magnification. See also Fig. S2. MPD, mean population doubling.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    753

  • 8/3/2019 BTG2 Antagonizes Pin1

    8/14

    G0G1 because of Pin1 inhibition are redirected into a senes-

    cence program upon mitogenic stimulation.

    Exogenous expression of Pin1 rescues BTG2-

    mediated senescence in late-passage cellsBecause Pin1 inhibition leads to senescence and BTG2 expres-

    sion promotes senescence in late-passage cells, possibly through

    its interaction with Pin1, we tested whether ectopically

    expressed Pin1 could prevent BTG2-induced senescence in

    shp53 cells. Pin1 was coexpressed with BTG2 in late-passage

    shp53 cells (Fig. 8A). Approximately 80% of the cells expressing

    BTG2 alone exhibited b-galactosidase staining and flattened

    morphology typical of senescent cells when compared with

    10.4% of control shp53 cells infected with empty vector. Coex-

    pression of Pin1 and BTG2 resulted in only 17.5% of the cells

    staining for b-galactosidase (Fig. 8B). These results show that

    Pin1 expression can rescue cells from BTG2-dependent senes-cence and serve to demonstrate a functional relationship

    between BTG2 and Pin1.

    Discussion

    The repression of p53 by shRNA allows normal human fibro-

    blasts to bypass their normal replicative limit. While this demon-

    strates that p53 activation is a critical step in the development of

    senescence, the downstream p53 effectors required to induce

    senescence have not been fully established. In this study, we

    show that the p53-responsive gene BTG2 plays an essential role

    in replicative senescence and in T-oligo-induced senescence (an

    oligonucleotide-based model that mimics the natural uncapping

    of telomeres during replicative senescence). Endogenous BTG2

    protein levels rise during replicative and T-oligo-induced senes-

    cence and shRNA-mediated repression of BTG2 extends prolifer-ative lifespan and prevents T-oligo-induced senescence.

    Moreover, ectopic BTG2 promotes senescence in cells that have

    bypassed their normal replicative limit because of shRNA-medi-

    ated silencing of p53 and restores the sensitivity of shp53 cells

    to undergo T-oligo-induced senescence. These results demon-

    strate that BTG2 can act independently of p53 to induce senes-

    cence in response to dysfunctional telomeres.

    One striking observation is the ability of ectopic BTG2 or p53

    to promote senescence in shp53 cells that have bypassed their

    normal replicative limit and their inability to promote senescence

    in early-passage cells that have not yet reached their normal pro-

    liferative limit. This is unlike p21, which is capable of arrestingcells at any point during their replicative lifespan (Sang et al.,

    2008). We find that ectopic BTG2 protein is unstable in young

    cells and that BTG2 protein accumulates at higher passage

    number. One possible explanation is that uncapped telomeres

    activate a signaling pathway that promotes BTG2 protein

    stabilization. The activation of ectopic p53 in late-passage but

    not early-passage cells is likely because of telomere-dependent

    ATM activation. We find that ectopic p53 is expressed at similar

    levels in early- and late-passage shp53 cells but that p53 Ser15

    phosphorylation and p53 target gene expression (p21, BTG2)

    Fig. 6 The BTG2 A147S mutant does not colocalize with Pin1. BJshp53 cells beyond their normal replicative limit were retrovirally infected with BTG2 or BTG2

    A147S expression constructs and selected with hygromycin. Cells were serum starved for 72 h or serum stimulated for 24 h, fixed and analyzed by immuno-

    fluorescence staining for BTG2 and Pin1. Nuclear counterstaining used Draq5. Images show a single optical layer of a confocal image at 400 magnification.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    754

  • 8/3/2019 BTG2 Antagonizes Pin1

    9/14

    occur only in late-passage cells. Because telomeres continue to

    shorten in late-passage shp53 cells leading to crisis, the ATM

    pathway is expected to remain active but incompetent at pro-

    moting senescence in the absence of p53. A similar telomere

    dysfunctionDNA damage sensing pathway is likely to be play-

    ing a role in the stabilization of BTG2 in late-passage cells.

    In addition to protein stabilization, BTG2 activation is depen-

    dent on mitogenic stimulation resulting in phosphorylation on

    Ser147. The dual regulation of BTG2 function in response to

    telomere disruption and mitogenic stimulation provides tight

    control over BTG2 expression at senescence. Moreover, thetranscriptional regulation of BTG2 expression by p53 provides a

    third level of control to ensure that BTG2 levels remain elevated

    during senescence (see model, Fig. 8C).

    We demonstrate that BTG2 promotes senescence through its

    ability to bind and sequester Pin1. We show that Pin1 inhibition

    leads to senescence in late-passage cells and that ectopic Pin1

    expression rescues cells from BTG2-induced senescence. Taken

    together, our findings indicate that BTG2 and Pin1 interact phys-

    ically and functionally in senescence. The mitogen-dependent

    binding and colocalization of BTG2 with Pin1 during senescence

    suggest a model in which BTG2 promotes senescence by redis-

    tributing Pin1 in the cytoplasm and preventing Pin1 from inter-

    acting with proteins required for cell cycle progression. This is

    consistent with previous reports showing that Pin1 is required

    for mouse primary cells to escape quiescence in response to

    serum (Fujimori et al., 1999; You et al., 2002). Our results with

    human fibroblasts using PiB to inhibit the catalytic activity of

    Pin1 and shRNA to reduce endogenous Pin1 indicate that Pin1 is

    required for S phase entry only in late-passage cells. Inhibition of

    Pin1 leads to senescence in p53 knockdown cells only after

    these cells are prompted to reenter the cell cycle with serumafter being held in G0G1 by serum deprivation. It is notable

    that senescent cells do not proliferate in response to mitogenic

    stimulation even though mitogenic signaling pathways are

    intact (reviewed in (Wheaton et al., 1996). Moreover, persistent

    mitogenic stimulation is required for the development of the

    senescent phenotype (Satyanarayana et al., 2004). As Pin1 pro-

    motes S phase entry in response to mitogenic stimulation, these

    findings suggest that the interaction between BTG2 and Pin1

    provides a mechanism to neutralize Pin1 function to ensure

    that senescent cells remain in G1 in the presence of mitogenic

    (A) (D)

    (B)

    (C)

    Fig. 7 Pin1 inhibition leads to senescence in late-passage quiescent cells stimulated to reenter the cell cycle. (A) The G1S ratios of BJshp53 cells, 24 h after

    serum stimulation in the presence or absence of the Pin1 inhibitor, PiB (20 lM). (B) b-galactosidase staining of BJshp53 cells, 2 weeks after serum stimulation in

    the presence of absence of PiB. The PiB in the culture medium was replenished every second day. (C) Western blot analysis of Pin1 expression in BJshp53 cells

    expressing shRNA directed to Pin1 or empty pSuper vector (pS) at MPD 63 or 73. (D) Upper 2 panels: The cell cycle profiles of BJshp53 cells and BJshp53shPin1

    cells, 72 h after serum withdrawal. The b-galactosidase staining of the cells, 2 weeks after serum withdrawal is shown immediately below. Lower 2 panels: The cell

    cycle profiles of BJshp53 cells and BJshp53shPin1 cells stimulated with serumfor 24 h following serum starvation for 48 h. The b-galactosidase staining of the

    cells, 2 weeks after serum stimulation, is shown at the bottom. MPD, mean population doubling.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    755

  • 8/3/2019 BTG2 Antagonizes Pin1

    10/14

    signals. Three observations support this idea: first, binding to

    Pin1 requires MAPK-dependent phosphorylation of BTG2 on

    Ser147 (Hong et al., 2005); second, the BTG2 S147A mutant

    protein is unable to induce senescence in late-passage shp53

    and is defective in promoting Pin1 nuclear export; and third,

    ectopic Pin1 expression rescues cells from BTG2-induced senes-

    cence. In this model, p53-dependent induction of p21 initiates

    senescence by arresting cells in G1 and p53-dependent induc-

    tion of BTG2 inhibits Pin1, ensuring that cells remain in G1 dur-

    ing mitogenic stimulation. Although our study does not identify

    the Pin1 substrates that regulate S phase entry and senescence

    in late-passage cells, several candidates, among the manyknown Pin1 targets, may be considered including cyclin D1, cy-

    clin E, b-catenin, Rb and Myc (Yeh & Means, 2007).

    Previous studies reported that Pin1 functions as a positive

    regulator of p53 through an interaction that is dependent on

    DNA damageinduced p53 phosphorylation on several sites

    including Ser 33, Thr 81 and Ser 315 (Zacchi et al., 2002;

    Zheng et al., 2002), and yet our studies suggest that Pin1 must

    be inhibited during p53-dependent senescence. This could

    reflect temporal regulation of Pin1 during senescence, because

    Pin1 has been shown to have a dual role in regulating both

    p53 activity and cell cycle progression. Pin1 protein accumu-

    lates at senescence and may be required to activate p53; how-

    ever, upon activation of signaling cascades by serum, Pin1

    needs to be neutralized to maintain G1 arrest. Pin1 in senes-

    cent cells is inactivated through interaction with BTG2 to block

    activation of target phosphoproteins required for cell cycle

    progression. Thus, BTG2 becomes a significant Pin1 substrate

    only when cells are mitogenically stimulated to enter S phase.

    At other times in the cell cycle, Pin1 is free to interact with all

    its substrates including p53. It should also be noted that the

    involvement of Pin1 in regulating p53 is complex, being

    dependent on cell and tissue type; moreover, the requirementfor Pin1 differs on different p53-responsive genes (Zacchi

    et al., 2002; Zheng et al., 2002). The Pin1-p53 interaction has

    been studied primarily in established cell lines treated with

    DNA damaging agents and the role of Pin1 in enhancing the

    transcriptional activation of p53-regulated genes during repli-

    cative senescence is not known.

    Recently, TRF1 was reported to be a substrate for Pin1, and

    the interaction between Pin1 and TRF1 was shown to regulate

    telomere length in telomerase-positive transformed human cells

    (Lee et al., 2009). Upon Pin1 inhibition, TRF1 protein stability

    (A) (B)

    (C)

    Fig. 8 Exogenous expression of Pin1 rescues late-passage shp53 cells from BTG2-mediated senescence. (A) p53 shRNA-expressing BJ cells (BJshp53) at MPD 72

    were infected with the indicated retroviral expression constructs or with the empty pBabe vector. Western blot analysis was performed with the indicated

    antibodies 1 week after infection. b-actin serves as a loading control. (B) Staining for senescence-associatedb-galactosidase was performed on BJshp53 cells

    2 weeks after retroviral expression of BTG2 and Pin1. (C) Model showing the involvement of BTG2 during p53-dependent replicative senescence. p53 induces

    BTG2 mRNA expression. BTG2 protein is stabilized downstream of telomere-dependent signals and undergoes phosphorylation on Ser147 in response to

    mitogenic stimulation. Phosphorylated BTG2 binds Pin1 and promotes the nuclear export of Pin1. In this model, replicative senescence is dependent on telomere

    disruption, p53 activation and mitogenic stimulation. The model does not exclude the possibility that p53 promotes expression of other responsive genes such as

    p21 and PAIthat function collectively to induce senescence. MPD, mean population doubling.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    756

  • 8/3/2019 BTG2 Antagonizes Pin1

    11/14

    increased resulting in enhanced binding of TRF1 to telomeres

    and gradual telomere erosion in transformed cells. Pin1 knock-

    down, however, had no effect on telomere length in telomer-

    ase-negative normal human cells including human fibroblasts.

    Hence, there is no evidence for the TRF1Pin1 interaction play-

    ing a role in telomere shortening during normal replicative

    senescence, and it is unclear whether Pin1 affects TRF1 stability

    in normal human cells.Several studies using transformed cells report that BTG2 pro-

    motes cell cycle arrest through suppression of cyclin D1 (Guarda-

    vaccaro et al., 2000; Boiko et al., 2006; Kawakubo et al., 2006)

    or cyclin E expression (Lim et al., 1998). In one study, BTG2 was

    reported to contribute to Ras-induced senescence in fibroblasts

    through suppression of cyclin D1 and E levels (Boiko et al.,

    2006). Consistent with previous reports, we found that neither

    of these cyclins was suppressed during replicative senescence in

    human fibroblasts. We provide an alternative mechanism in

    which BTG2 temporally antagonizes Pin1 function in response

    to serum. Thus, senescence induced in response to eroded

    telomeres or oncogenic stress requires BTG2, but the mecha-nisms are divergent. This may reflect differences between nor-

    mal and transformed cells or differences between telomere-

    dependent replicative senescence and telomere-independent

    oncogene-induced senescence. Furthermore, oncogenic Ras

    delivers constitutive and deregulated signals for proliferation,

    whereas mitogens and growth factors deliver physiological

    signals to regulate cell cycle progression. Hence, BTG2-mediated

    cyclin D1 repression may be restricted to transformed cells while

    the interaction between BTG2 and Pin1 may be favoured in

    normal cells. The ability of BTG2 to neutralize Pin1-mediated

    entry in S phase is unique to normal senescent cells.

    Our study reveals a critical role for BTG2 in imposing the p53-

    dependent senescence barrier that limits the proliferative capac-

    ity of human cells. The interaction of BTG2 with Pin1 promotes

    its relocation and neutralizes its function. The inability of Pin1 to

    modify nuclear proteins required for S phase entry contributes

    to the senescence program of human fibroblasts.

    Experimental procedures

    Plasmid constructs

    The shRNA constructs were created using hairpin inserts with

    BglII and XhoI and ligated into pSUPER.retro.puro (Brummelk-

    amp et al., 2002). The shRNA target sequences included: p53,GACTCCAGTGGTAATCTAC; p21, GGTGACTTCGCCTGGGAG-

    CGT; BTG2, CTACGTGATGGCAGTCTCC; GADD45a, AG-

    TCGCTACATGGATCAAT; and Pin1, GCCGAGTGTACTACTT-

    CAA. cDNAs for human p21, BTG2 and Pin 1 were generated by

    PCR using the primers: ggaattcatgtcagaaccggctgg and cag-

    cgtcgacttagggcttcctctt for p21, catgagccacgggaag and atggca-

    gtctccagctagg for BTG2, and cgggatcccatggcggacgagg

    agaaagct and ggaattcctactcagtcggaggatga for Pin1. These

    products were cloned into pBabe.hygro or pBabe.zeo using

    BamHI and EcoRI.

    The p53RR construct was generated using 2 sets of primers

    that amplified the human p53 cDNA and overlapped in the

    region targeted by the shRNA (nt 775793 from the translation

    initiation site). The primers were designed to change the

    sequence of the p53 cDNA in the shRNA target region without

    changing the amino acid sequence. The 5-terminal fragment

    was amplified using cgggatccCATGGAGGAGCCGCAGT and

    CAAGTTGCCCGAGCTATCTTCCAGTGTGATGATGGTG, and the3-terminal fragment was amplified using TAGCTCGGGCA-

    ACTTGCTGGGACGGAACAGCTTTG and ggaattcGAGTCAG-

    TCTGAGTCAGG (underlined italics show the overlapping

    sequences that differ from wild-type p53 and lower case letter-

    ing indicates BamHI and EcoRI sites). A third PCR utilized the

    purified products of the first two reactions as both substrate and

    primer to generate full-length p53RR with silent nucleotide

    changes in the 775793 region. The full-length product was

    digested with BamHI and EcoRI and cloned into pBabe.hygro.

    All constructs were verified by automated sequencing.

    Cell culture and retroviral infection

    Hs68 and BJ human fibroblast cell strains were maintained as

    previously described (Wheaton & Riabowol, 2004). The fibro-

    blasts were infected with amphotropic isotyped virus (Phoenix-A

    packaging cells) containing the ecotropic receptor (pm5-Eco)

    and then selected with G418 (1 mg mL)1) for 2 weeks to gener-

    ate Hs68 and BJ Eco strains. Ecotropic retroviral supernatants

    were produced by cotransfection of HEK 293 cells with the vari-

    ous pBabe or pSuper retroviral constructs and pCL Eco using

    Fugene. After 48 or 72 h, the medium was collected, supple-

    mented with 0.8 lg mL)1 polybrene and used to infect Hs68

    Eco or BJ Eco cells. Selection was performed 4872 h after viral

    infection using 1 lg mL)1 puromycin for 3 days (pSuper orpBabe.puro), 150 lg mL)1 hygromycin for 4 days (pBabe.hy-

    gro), or 80 ng mL)1 zeocin for 3 days (pBabe.zeo). The acidic b-

    galactosidase assay was performed as described (Dimri et al.,

    1995).

    Cell counting

    BJ cells were virally infected at MPD 65, and Hs68 cells were

    infected at MPD 53. Every 7 days, cells were counted by coulter

    counter and replated at 1:10 or 1:20 dilution. The mean popula-

    tion doubling (MPD) was calculated by the formula: MPD = Log

    (Nf

    Ni)

    Log2, where Nf = the number of cells counted andNi = the number of cells seeded. Growth rate experiments used

    Hs68pSuper MPD 52 and Hs68shp53 MPD 52 expressing

    either ectopic BTG2 or pBabe.hygro vector. The resulting cell

    strains were seeded at 50 000 cells in triplicate for each time

    point and harvested for counting daily.

    Cell cycle analysis

    Cells were fixed on ice in 70% ethanol, washed with PBS con-

    taining 1% BSA, incubated with 100 lg mL)1 RNase A for

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    757

  • 8/3/2019 BTG2 Antagonizes Pin1

    12/14

    10 min at 37C and resuspended in PBS containing 50 lg mL)1

    propidium iodide. Cell cycle distribution was examined by flow

    cytometry using a FACScalibur flow cytometer (Becton Dickin-

    son, Mississauga, ON, Canada).

    T-oligo treatment

    The T-oligo (GTTAGGGTTAG) or its compliment (C-oligo;CTAACCCTAAC) were added to cells at 40 lM as described pre-

    viously (Li et al., 2003). Medium was changed after 1 week of

    treatment. Efficient T-oligo-induced senescence in fibroblasts

    depended on three variables: low cell density, timing of T-oligo

    treatment and the use of newborn calf serum. Cells were

    harvested 2 weeks after T-oligo treatment, and the C-oligo or

    vehicle (H2O) controls were harvested at a similar cell density of

    growth-arrested T-oligo-treated cells. In order to facilitate BTG2

    detection, the cells were serum starved for 48 h and stimulated

    with serum for 24 h before harvesting.

    Semiquantitative RTPCR

    Total RNA was isolated using the TRIzol reagent (Gibco-BRL,

    Burlington, ON, Canada) according to the manufacturers

    instructions. Reverse transcriptionPCR (RTPCR) was performed

    on 1 lg of total RNA. The optimal annealing temperature for

    each primer set was determined empirically, and varying cycles

    of PCR were performed to determine the linear range of amplifi-

    cation. PCR was performed using the following primer

    sequences, annealing temperature and cycle number.

    p21: ctggagactctcagggtcgaaa and gattagggcttcctcttgagaa,

    55C, 29 cycles;

    BTG2: gcgagcagaggcttaaggtc and aggccacttccaagcagctc,

    55C, 34 cycles;Reprimo: gcaatctgctcatcaagtccgag and ccccgcattccaagtaag-

    tagc, 55C, 43 cycles;

    14-3-3r: gtgtgtccccagagccatgg and accttctcccggtactcacg,

    55C, 47 cycles;

    Tob1: cacaggatcttagtgtttggatcga and ttcttcattttggtagagccga-

    act, 60C, 40 cycles; Gadd45a: gctctctccctgggcgacctg and

    ccatgtagcgactttcccggc, 55C, 34 cycles; and

    GAPDH: cggagtcaacggatttggtcgtat and agccttctccatggtg-

    gtgaagac, 55 or 60C, 22 cycles. Glyceraldehyde-3-phosphate

    dehydrogenase (GAPDH) served as an internal loading control.

    To ensure linear GAPDH amplification, its primers were added

    for the final 22 cycles of each reaction. The amplified productswere resolved by agarose gel (2%) electrophoresis, stained

    with ethidium bromide, and band intensity was determined

    using UV illumination and IMAGEQUANT (GE Healthcare, Baie

    durfe, QC, Canada) software using GAPDH as an internal

    control.

    Northern blot analysis

    Fifteen micrograms of RNA was resolved on a denaturing aga-

    rose gel and transferred to a nylon membrane. Hybridization of

    32P-radiolabelled probes was performed using standard condi-

    tions. Labeled RNA was detected by autoradiography.

    Western blots and immunoprecipitation

    Cells were lysed directly in 2X Laemmli sample buffer (4%

    SDS, 25 mM TrisHCl [pH 6.8], 20% glycerol, 0.1 M DTT) or

    lysed in PBS containing 1% NP40 and complete proteaseinhibitor cocktail. Protein samples were quantified by Bradford

    assay, resolved by PAGE, transferred onto nitrocellulose and

    blocked overnight in 10% milk or 5% BSA TBS with 0.5%

    Tween 20. Primary or secondary antibodies were diluted in

    5% BSA TBS with 0.5% Tween 20. The following antibodies

    were used: p53 (DO-1), phospho(S-15)-p53 (Cell Signaling,

    Danvers, MA, USA, #9284), BTG2 (Santa Cruz, CA, USA,

    sc-33775 and Aviva, ARP33561), cyclin E (Santa Cruz, sc-198),

    cyclin D1 (NeoMakers, Fremont, CA, USA, RB-010), p21

    (Santa Cruz, sc-397), GADD45a (Santa Cruz, sc-797), Pin1

    (Santa Cruz, sc-46660 and sc-15340) and b-actin (Sigma,

    Oakville, ON, Canada). Anti-rabbit and anti-mouse secondary

    antibodies were conjugated with HRP (Jackson IR, West Grove,

    PA, USA).

    Immunoprecipitations were performed using DSP cross-

    linked cell lysates (Thermoscientific, Nepean, ON, Canada) and

    precleared using Protein ASepharose beads. The mouse Pin1

    antibody (sc46660) or control immunoglobulin (mIgG) was

    incubated overnight at 4C followed by the addition of Protein

    ASepharose beads for a further 60 min at 4C. IPs were

    washed three times using RIPA as previously described (Whea-

    ton & Riabowol, 2004). The bead IP complexes were boiled for

    15 min in protein sample buffer containing 50 mM DTT and

    5% b-mercaptoethanol to reverse cross-linking. Analysis of

    bound BTG2 and Pin1 was performed by Western blottingusing rabbit polyclonal antibodies sc-33775 and sc-15340,

    respectively.

    Immunofluorescence microscopy

    Cells on glass coverslips were fixed with 2% paraformaldehyde

    in PBS for 10 min and permeabilized with 0.5% Triton X for

    15 min. Antibodies were diluted in 1% BSA TBS. BTG2 staining

    utilized anti-BTG2 (sc-30342; 1:50) for 1 h, followed by mouse

    anti-goat (Sigma; 1:200) for 1 hr and goat anti-mouse FITC-con-

    jugated antibody (Sigma). Pin1 staining utilized anti-Pin1

    (sc-15340; 1:200) for 1 hr, followed by anti-rabbit Cy-3 (JacksonIR; 1:800) for 30 min. Coverslips were washed 3 times with TBS

    with 0.5% Tween 20 between antibody incubations. The nuclei

    were stained with Draq5 (Enzolife Sciences International, Inc.,

    Plymouth Meeting, PA, USA) according to the manufacturers

    instructions. Images were obtained using an Olympus FluoView

    300 confocal laser-scanning microscope (Carsen Group,

    Markhan, ON, Canada), and a single 0.35 -lm optical section at

    400 magnification of each sample is shown. Images were

    analyzed and superimposed using IMAGEJ (NIH, Bethesda, MD,

    USA) software.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    758

  • 8/3/2019 BTG2 Antagonizes Pin1

    13/14

    Acknowledgments

    Rosalyn Fleites Garcia for technical assistance. This research is

    funded by the Canadian Cancer Society and the Canadian

    Institutes of Health Research. SB is supported by a Canada

    Research Chair.

    Author contributions

    KW & SB designed the experiments and wrote the manuscript.

    KW, WM & JM conducted the experiments.

    References

    Atadja P, Wong H, Garkavtsev I, Veillette C, Riabowol K (1995)

    Increased activity of p53 in senescing fibroblasts. Proc. Natl Acad.

    Sci. USA 92, 83488352.

    Boiko AD, Porteous S, Razorenova OV, Krivokrysenko VI, Williams BR,

    Gudkov AV (2006) A systematic search for downstream mediators

    of tumor suppressor function of p53 reveals a major role of BTG2 in

    suppression of Ras-induced transformation. Genes Dev. 20, 236252.

    Brown JP, Wei W, Sedivy JM (1997) Bypass of senescence after disrup-

    tion of p21CIP1WAF1 gene in normal diploid human fibroblasts.

    Science 277, 831834.

    Brummelkamp TR, Bernards R, Agami R (2002) A system for stable

    expression of short interfering RNAs in mammalian cells. Science

    296, 550553.

    Collado M, Blasco MA, Serrano M (2007) Cellular senescence in can-

    cer and aging. Cell 130, 223233.

    el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM,

    Lin D, Mercer WE, Kinzler KW, Vogelstein B (1993) WAF1, a poten-

    tial mediator of p53 tumor suppression. Cell 75, 817825.

    Dimri GP (2005) What has senescence got to do with cancer? Cancer

    Cell 7, 505512.Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano

    EE, Linskens M, Rubelj I, Pereira-Smith O, Peacocke M, Campisi J

    (1995) A biomarker that identifies senescent human cells in culture

    and in aging skin in vivo. Proc. Natl Acad. Sci. USA 92, 93639367.

    Dulic V, Drullinger LF, Lees E, Reed SI, Stein GH (1993) Altered regula-

    tion of G1 cyclins in senescent human diploid fibroblasts: accumula-

    tion of inactive cyclin E-Cdk2 and cyclin D1-Cdk2 complexes. Proc.

    Natl Acad. Sci. USA 90, 1103411038.

    Dulic V, Beney GE, Frebourg G, Drullinger LF, Stein GH (2000) Uncou-

    pling between phenotypic senescence and cell cycle arrest in aging

    p21-deficient fibroblasts. Mol. Cell. Biol. 20, 67416754.

    Fujimori F, Takahashi K, Uchida C, Uchida T (1999) Mice lacking Pin1

    develop normally, but are defective in entering cell cycle from G(0)

    arrest. Biochem. Biophys. Res. Commun. 265, 658663.

    Guardavaccaro D, Corrente G, Covone F, Micheli L, DAgnano I,

    Starace G, Caruso M, Tirone F (2000) Arrest of G(1)-S progres-

    sion by the p53-inducible gene PC3 is Rb dependent and relies

    on the inhibition of cyclin D1 transcription. Mol. Cell. Biol. 20,

    17971815.

    Hara E, Tsurui H, Shinozaki A, Nakada S, Oda K (1991) Cooperative

    effect of antisense-Rb and antisense-p53 oligomers on the extension

    of life span in human diploid fibroblasts, TIG-1. Biochem. Biophys.

    Res. Commun. 179, 528534.

    Harley CB, Futcher AB, Greider CW (1990) Telomeres shorten during

    ageing of human fibroblasts. Nature 345, 458460.

    Hayflick L (1965) The limited in vitro lifetime of human diploid cell

    strains. Exp. Cell Res. 37, 614636.

    Herbig U, Jobling WA, Chen BP, Chen DJ, Sedivy JM (2004) Telomere

    shortening triggers senescence of human cells through a pathway

    involving ATM, p53, and p21(CIP1), but not p16(INK4a). Mol. Cell

    14, 501513.

    Hong JW, Ryu MS, Lim IK (2005) Phosphorylation of serine 147 of

    tis21BTG2pc3 by p-Erk12 induces Pin-1 binding in cytoplasm

    and cell death. J. Biol. Chem. 280, 2125621263.Itahana K, Dimri GP, Hara E, Itahana Y, Zou Y, Desprez PY, Campisi J

    (2002) A role for p53 in maintaining and establishing the quies-

    cence growth arrest in human cells. J. Biol. Chem. 277, 18206

    18214.

    Jackson JG, Pereira-Smith OM (2006) p53 is preferentially recruited to

    the promoters of growth arrest genes p21 and GADD45 during rep-

    licative senescence of normal human fibroblasts. Cancer Res. 66,

    83568360.

    Karlseder J, Smogorzewska A, de Lange T (2002) Senescence induced

    by altered telomere state, not telomere loss. Science 295, 2446

    2449.

    Kawakubo H, Brachtel E, Hayashida T, Yeo G, Kish J, Muzikansky A,

    Walden PD, Maheswaran S (2006) Loss of B-cell translocation gene-

    2 in estrogen receptor-positive breast carcinoma is associated withtumor grade and overexpression of cyclin d1 protein. Cancer Res.

    66, 70757082.

    Kortlever RM, Higgins PJ, Bernards R (2006) Plasminogen activator

    inhibitor-1 is a critical downstream target of p53 in the induction of

    replicative senescence. Nat. Cell Biol. 8, 877884.

    Lee TH, Tun-Kyi A, Shi R, Lim J, Soohoo C, Finn G, Balastik M, Pastori-

    no L, Wulf G, Zhou XZ, Lu KP (2009) Essential role of Pin1 in the

    regulation of TRF1 stability and telomere maintenance. Nat. Cell

    Biol. 11, 97105.

    Li GZ, Eller MS, Firoozabadi R, Gilchrest BA (2003) Evidence that expo-

    sure of the telomere 3 overhang sequence induces senescence.

    Proc. Natl Acad. Sci. USA 100, 527531.

    Lim IK (2006) TIS21 (BTG2PC3) as a link between ageing and can-

    cer: cell cycle regulator and endogenous cell death molecule. J. Can-

    cer Res. Clin. Oncol. 132, 417426.

    Lim IK, Lee MS, Ryu MS, Park TJ, Fujiki H, Eguchi H, Paik WK (1998)

    Induction of growth inhibition of 293 cells by downregulation of

    the cyclin E and cyclin-dependent kinase 4 proteins due to overex-

    pression of TIS21. Mol. Carcinog. 23, 2535.

    Lucibello FC, Sewing A, Brusselbach S, Burger C, Muller R (1993)

    Deregulation of cyclins D1 and E and suppression of cdk2 and cdk4

    in senescent human fibroblasts. J. Cell Sci. 105(Pt 1), 123133.

    Ma Y, Prigent SA, Born TL, Monell CR, Feramisco JR, Bertolaet BL

    (1999) Microinjection of anti-p21 antibodies induces senescent Hs68

    human fibroblasts to synthesize DNA but not to divide. Cancer Res.

    59, 53415348.

    Masutomi K, Yu EY, Khurts S, Ben-Porath I, Currier JL, Metz GB,

    Brooks MW, Kaneko S, Murakami S, DeCaprio JA, Weinberg RA,

    Stewart SA, Hahn WC (2003) Telomerase maintains telomere struc-

    ture in normal human cells. Cell 114, 241253.

    Meerson A, Milyavsky M, Rotter V (2004) p53 mediates density-

    dependent growth arrest. FEBS Lett. 559, 152158.

    Noda A, Ning Y, Venable SF, Pereira-Smith OM, Smith JR (1994) Clon-

    ing of senescent cell-derived inhibitors of DNA synthesis using an

    expression screen. Exp. Cell Res. 211, 9098.

    Rouault JP, Falette N, Guehenneux F, Guillot C, Rimokh R, Wang Q,

    Berthet C, Moyret-Lalle C, Savatier P, Pain B, Shaw P, Berger R,

    Samarut J, Magaud JP, Ozturk M, Samarut C, Puisieux A (1996)

    Identification of BTG2, an antiproliferative p53-dependent compo-

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    759

  • 8/3/2019 BTG2 Antagonizes Pin1

    14/14

    nent of the DNA damage cellular response pathway. Nat. Genet.

    14, 482486.

    Rozan LM, El-Deiry WS (2007) p53 downstream target genes and

    tumor suppression: a classical view in evolution. Cell Death Differ.

    14, 39.

    Sang L, Coller HA, Roberts JM (2008) Control of the reversibility of

    cellular quiescence by the transcriptional repressor HES1. Science

    321, 10951100.

    Sasajima H, Nakagawa K, Yokosawa H (2002) Antiproliferative pro-teins of the BTGTob family are degraded by the ubiquitin-protea-

    some system. Eur. J. Biochem. 269, 35963604.

    Satyanarayana A, Greenberg RA, Schaetzlein S, Buer J, Masutomi K,

    Hahn WC, Zimmermann S, Martens U, Manns MP, Rudolph KL

    (2004) Mitogen stimulation cooperates with telomere shortening to

    activate DNA damage responses and senescence signaling. Mol.

    Cell. Biol. 24, 54595474.

    Shay JW, Pereira-Smith OM, Wright WE (1991) A role for both RB and

    p53 in the regulation of human cellular senescence. Exp. Cell Res.

    196, 3339.

    Stein GH, Drullinger LF, Soulard A, Dulic V (1999) 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.Stewart SA, Weinberg RA (2006) Telomeres: cancer to human aging.

    Annu. Rev. Cell Dev. Biol. 22, 531557.

    Vaziri H, West MD, Allsopp RC, Davison TS, Wu YS, Arrowsmith CH,

    Poirier GG, Benchimol S (1997) ATM-dependent telomere loss in

    aging human diploid fibroblasts and DNA damage lead to the post-

    translational activation of p53 protein involving poly(ADP-ribose)

    polymerase. EMBO J. 16, 60186033.

    Verdun RE, Karlseder J (2007) Replication and protection of telomeres.

    Nature 447, 924931.

    Vousden KH (2006) Outcomes of p53 activationspoilt for choice.

    J. Cell Sci. 119, 50155020.

    Vousden KH, Lane DP (2007) p53 in health and disease. Nat. Rev.

    Mol. Cell Biol. 8, 275283.

    Wheaton K, Riabowol K (2004) Protein kinase C delta blocks immedi-

    ate-early gene expression in senescent cells by inactivating serum

    response factor. Mol. Cell. Biol. 24, 72987311.

    Wheaton K, Atadja P, Riabowol K (1996) Regulation of transcription

    factor activity during cellular aging. Biochem. Cell Biol. 74, 523

    534.

    Wyllie F, Haughton M, Bartek J, Rowson J, Wynford-Thomas D (2003)

    Mutant p53 can delay growth arrest and loss of CDK2 activity in

    senescing human fibroblasts without reducing p21(WAF1) expres-

    sion. Exp. Cell Res. 285, 236242.

    Yeh ES, Means AR (2007) PIN1, the cell cycle and cancer. Nat. Rev.

    Cancer 7, 381388.You H, Zheng H, Murray SA, Yu Q, Uchida T, Fan D, Xiao ZX (2002)

    IGF-1 induces Pin1 expression in promoting cell cycle S-phase entry.

    J. Cell. Biochem. 84, 211216.

    Zacchi P, Gostissa M, Uchida T, Salvagno C, Avolio F, Volinia S, Ronai

    Z, Blandino G, Schneider C, Del Sal G (2002) The prolyl isomerase

    Pin1 reveals a mechanism to control p53 functions after genotoxic

    insults. Nature 419, 853857.

    Zheng H, You H, Zhou XZ, Murray SA, Uchida T, Wulf G, Gu L, Tang

    X, Lu KP, Xiao ZX (2002) The prolyl isomerase Pin1 is a regulator of

    p53 in genotoxic response. Nature 419, 849853.

    Supporting Information

    Additional supporting information may be found in the online

    version of this article:

    Fig. S1 Characterization of Hs68 cells expressing p53 shRNA,

    related to Fig. 2 (tif file).

    Fig. S2 The cellular redistribution of Pin1 and its colocalization

    with BTG2 in the cytoplasm of BJ cells induced to undergo

    senescence using T-oligo, related to Fig. 5 (tif file).

    As a service to our authors and readers, this journal provides

    supporting information supplied by the authors. Such materials

    are peer-reviewed and may be reorganized for online delivery

    but are not copyedited or typeset. Technical support issues aris-

    ing from supporting information (other than missing files)

    should be addressed to the authors.

    BTG2 neutralizes Pin1 during replicative senescence, K. Wheaton et al.

    2010 The Authors

    Aging Cell 2010 Blackwell Publishing Ltd/Anatomical Society of Great Britain and Ireland

    760