PML IV/ARF interaction enhances p53 SUMO-1 conjugation ... · PML IV on p53 regulators, we first...

6
PML IV/ARF interaction enhances p53 SUMO-1 conjugation, activation, and senescence Lisa Ivanschitz a,b,c , Yuki Takahashi a,b,c , Florence Jollivet a,b,c , Olivier Ayrault d , Morgane Le Bras a,b,c,1 , and Hugues de Thé a,b,c,e,f,1,2 a University Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis 1, Paris cedex 10, France; b INSERM U944, Equipe Labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire dHématologie, Hôpital St. Louis 1, Paris cedex 10, France; c CNRS UMR 7212, Hôpital St. Louis 1, Paris cedex 10, France; d CNRS UMR 3306, INSERM U1005, Équipe Avenir, Laboratoire Signalisation, Développement et Tumeurs Cérébrales, Institut Curie, Campus Universitaire dOrsay, 91400 Orsay, France; e Assistance Publique-Hôpitaux de Paris, Service de Biochimie, Hôpital St. Louis 1, Paris cedex 10, France; and f Collège de France, Chaire dOncologie Cellulaire et Moléculaire, 75005 Paris, France Edited by Louis M. Staudt, National Cancer Institute, NIH, Bethesda, MD, and approved October 6, 2015 (received for review April 18, 2015) Promyelocytic leukemia protein (PML) nuclear bodies (NBs) recruit multiple partners, including p53 and many of its regulators. NBs are believed to facilitate several posttranslational modifications and are key regulators of senescence. PML, the organizer of NBs, is expressed as a number of splice variants that all efficiently recruit p53 partners. However, overexpression of only one of them, PML IV, triggers p53-driven senescence. Here, we show that PML IV specifically binds ARF, a key p53 regulator. Similar to ARF, PML IV enhances global SUMO-1 conjugation, particularly that of p53, resulting in p53 stabilization and activation. ARF interacts with and stabilizes the NB-associated UBC9 SUMO-conjugating enzyme, possibly explaining PML IV-enhanced SUMOylation. These results unexpectedly link two key tumor suppressors, highlighting their convergence for global control of SUMO conjugation, p53 activa- tion, and senescence induction. UBC9 | posttranslational modification | nuclear matrix | splicing | tumor suppression P romyelocytic leukemia protein (PML) is the key organizer of PML nuclear bodies (NBs), organelles that recruit an ever- growing number of unrelated partner proteins (1). Initially dis- covered through its targeting by an oncogenic translocation (2), PML has drawn considerable attention through its complex link with SUMOylation, a posttranslational modification (PTM) by ubiquitin-like peptides (SUMO). Indeed, PML must be SUMOylated to recruit its partners within PML NBs, together with the uni- versal SUMO-conjugating enzyme UBC9 then facilitating the partners modification (1, 35). Alternative splicing of human PML yields at least seven isoforms, and previous work has im- plicated these isoforms in specific interactions or functions (68). In particular, overexpression of a single isoform, PML IV, ini- tiates p53 activation and drives senescence (8). Conversely, the requirement for PML in senescence induction was demonstrated by Rass inability to induce senescence in pml -/- cells (9, 10). In many other experimental systems, p53 activation and/or senes- cence induction can be antagonized by PML extinction (11, 12). p53 is a multifunctional transcription factor, which is a key regulator of survival and proliferation (13). p53 activity is tightly regulated through multiple PTMs that control interactions with partner proteins or DNA. How exactly PML controls p53 is still debated. Numerous studies have pointed out the ability of PML to recruit p53 and many of its specific regulators (HIPK2, HAUSP, DAXX, CBP, HDM2, SIRT1, Chk2, MageA2, MOZ, etc.) into NBs (10, 1420). Partner recruitment/retention within NBs was proposed to enhance various p53 PTMs, particularly acetylation of lysine K382. However, neither partner nor p53 recruitment is specific for PML IV (8), so the basis for the ex- quisite specificity of PML IV to control p53 signaling remains ill-understood. A study proposed that PML IV represses TBX2, a gene amplified in some cancers that controls the CDKN2A promoter (21). The CDKN2A locus encodes two related products, ARF and p16INK4A, both involved in senescence activation through p53 and pRB, respectively. Besides its control of p53 abundance, ARF also exerts p53-independent functions, no- tably on the regulation of cell growth (22). These functions were proposed to rely, at least in part, on ARFs ability to enhance global SUMOylation (2327). Here, we demonstrate that PML IV specifically binds ARF and enhances p53 SUMO-1 conjugation, resulting in p53 stabi- lization and senescence. Our results unexpectedly bridge PML and ARF, two key senescence players, and highlight their roles in SUMOylation and control of p53-induced senescence. Results PML IV C Terminus Is Required to Trigger Senescence. PML IV exon 8a is shared with PML I (Fig. 1A) and is thus unlikely to account on its own for senescence induction. Exon 8b, specific for PML IV, encodes a 13-amino-acid sequence conserved among species (Fig. 1A and Fig. S1). Remarkably, this short amino acid stretch contains seven cyclic amino acids with high steric hindrance. In particular, three bulky aromatic residues (human residues F623, Y627, and F631) are perfectly conserved between man and mouse (Fig. 1A) and in most mammals (Fig. S1). To test whether these amino acids are involved in the specific function(s) of PML IV, we generated two mutants, either deleted for the PML IV-specific sequence (PML IVΔC) or with substi- tutions of aromatic residues for alanine (PML IV3A: F623A, Y627A, and F631A) (Fig. 1A). Overexpression of PML IVΔC, PML IV3A, or PML I in WI38 human primary fibroblasts failed to induce growth arrest, whereas PML IV or oncogenic Ras did (Fig. 1B) (8, 10). Moreover, Ras- and PML IV-transduced cells Significance Promyelocytic leukemia protein (PML) nuclear bodies (NBs) are subnuclear domains proposed to facilitate posttranslational modifications. Among NB partners, proteins are p53 and are most of the regulators. Overexpression of a single PML splice variant, PML IV, triggers p53-driven senescence. We demonstrate an interaction between PML IV C terminus and the ARF tumor suppressor. This interaction is required to promote p53 SUMOylation and subsequent stabilization. These results unexpectedly bridge three key tumor suppressors and stress the key role of PML NBs as SUMOylation factories and regu- lators of senescence. Author contributions: M.L.B. and H.d.T. designed research; L.I., Y.T., F.J., O.A., and M.L.B. performed research; F.J. contributed new reagents/analytic tools; L.I., Y.T., M.L.B., and H.d.T. analyzed data; and L.I., M.L.B., and H.d.T. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. 1 M.L.B. and H.d.T. contributed equally to this work. 2 To whom correspondence should be addressed. Email: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1507540112/-/DCSupplemental. 1427814283 | PNAS | November 17, 2015 | vol. 112 | no. 46 www.pnas.org/cgi/doi/10.1073/pnas.1507540112 Downloaded by guest on March 15, 2020

Transcript of PML IV/ARF interaction enhances p53 SUMO-1 conjugation ... · PML IV on p53 regulators, we first...

Page 1: PML IV/ARF interaction enhances p53 SUMO-1 conjugation ... · PML IV on p53 regulators, we first investigated SUMO modi-fication of HDMX and HDM2. In HeLa cells or H1299 cells stably

PML IV/ARF interaction enhances p53 SUMO-1conjugation, activation, and senescenceLisa Ivanschitza,b,c, Yuki Takahashia,b,c, Florence Jolliveta,b,c, Olivier Ayraultd, Morgane Le Brasa,b,c,1,and Hugues de Théa,b,c,e,f,1,2

aUniversity Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis 1, Paris cedex 10, France; bINSERM U944, Equipe Labellisée par la Ligue Nationale contrele Cancer, Institut Universitaire d’Hématologie, Hôpital St. Louis 1, Paris cedex 10, France; cCNRS UMR 7212, Hôpital St. Louis 1, Paris cedex 10, France; dCNRSUMR 3306, INSERM U1005, Équipe Avenir, Laboratoire Signalisation, Développement et Tumeurs Cérébrales, Institut Curie, Campus Universitaire d’Orsay,91400 Orsay, France; eAssistance Publique-Hôpitaux de Paris, Service de Biochimie, Hôpital St. Louis 1, Paris cedex 10, France; and fCollège de France,Chaire d’Oncologie Cellulaire et Moléculaire, 75005 Paris, France

Edited by Louis M. Staudt, National Cancer Institute, NIH, Bethesda, MD, and approved October 6, 2015 (received for review April 18, 2015)

Promyelocytic leukemia protein (PML) nuclear bodies (NBs) recruitmultiple partners, including p53 and many of its regulators. NBsare believed to facilitate several posttranslational modificationsand are key regulators of senescence. PML, the organizer of NBs, isexpressed as a number of splice variants that all efficiently recruitp53 partners. However, overexpression of only one of them, PMLIV, triggers p53-driven senescence. Here, we show that PML IVspecifically binds ARF, a key p53 regulator. Similar to ARF, PML IVenhances global SUMO-1 conjugation, particularly that of p53,resulting in p53 stabilization and activation. ARF interacts withand stabilizes the NB-associated UBC9 SUMO-conjugating enzyme,possibly explaining PML IV-enhanced SUMOylation. These resultsunexpectedly link two key tumor suppressors, highlighting theirconvergence for global control of SUMO conjugation, p53 activa-tion, and senescence induction.

UBC9 | posttranslational modification | nuclear matrix | splicing |tumor suppression

Promyelocytic leukemia protein (PML) is the key organizer ofPML nuclear bodies (NBs), organelles that recruit an ever-

growing number of unrelated partner proteins (1). Initially dis-covered through its targeting by an oncogenic translocation (2),PML has drawn considerable attention through its complex linkwith SUMOylation, a posttranslational modification (PTM) byubiquitin-like peptides (SUMO). Indeed, PMLmust be SUMOylatedto recruit its partners within PML NBs, together with the uni-versal SUMO-conjugating enzyme UBC9 then facilitating thepartner’s modification (1, 3–5). Alternative splicing of humanPML yields at least seven isoforms, and previous work has im-plicated these isoforms in specific interactions or functions (6–8).In particular, overexpression of a single isoform, PML IV, ini-tiates p53 activation and drives senescence (8). Conversely, therequirement for PML in senescence induction was demonstratedby Ras’s inability to induce senescence in pml−/− cells (9, 10). Inmany other experimental systems, p53 activation and/or senes-cence induction can be antagonized by PML extinction (11, 12).p53 is a multifunctional transcription factor, which is a key

regulator of survival and proliferation (13). p53 activity is tightlyregulated through multiple PTMs that control interactions withpartner proteins or DNA. How exactly PML controls p53 is stilldebated. Numerous studies have pointed out the ability of PMLto recruit p53 and many of its specific regulators (HIPK2,HAUSP, DAXX, CBP, HDM2, SIRT1, Chk2, MageA2, MOZ,etc.) into NBs (10, 14–20). Partner recruitment/retention withinNBs was proposed to enhance various p53 PTMs, particularlyacetylation of lysine K382. However, neither partner nor p53recruitment is specific for PML IV (8), so the basis for the ex-quisite specificity of PML IV to control p53 signaling remainsill-understood. A study proposed that PML IV represses TBX2,a gene amplified in some cancers that controls the CDKN2Apromoter (21). The CDKN2A locus encodes two related products,ARF and p16INK4A, both involved in senescence activation

through p53 and pRB, respectively. Besides its control of p53abundance, ARF also exerts p53-independent functions, no-tably on the regulation of cell growth (22). These functions wereproposed to rely, at least in part, on ARF’s ability to enhanceglobal SUMOylation (23–27).Here, we demonstrate that PML IV specifically binds ARF

and enhances p53 SUMO-1 conjugation, resulting in p53 stabi-lization and senescence. Our results unexpectedly bridge PMLand ARF, two key senescence players, and highlight their roles inSUMOylation and control of p53-induced senescence.

ResultsPML IV C Terminus Is Required to Trigger Senescence. PML IV exon8a is shared with PML I (Fig. 1A) and is thus unlikely to accounton its own for senescence induction. Exon 8b, specific for PMLIV, encodes a 13-amino-acid sequence conserved among species(Fig. 1A and Fig. S1). Remarkably, this short amino acid stretchcontains seven cyclic amino acids with high steric hindrance. Inparticular, three bulky aromatic residues (human residues F623,Y627, and F631) are perfectly conserved between man andmouse (Fig. 1A) and in most mammals (Fig. S1).To test whether these amino acids are involved in the specific

function(s) of PML IV, we generated two mutants, either deletedfor the PML IV-specific sequence (PML IVΔC) or with substi-tutions of aromatic residues for alanine (PML IV3A: F623A,Y627A, and F631A) (Fig. 1A). Overexpression of PML IVΔC,PML IV3A, or PML I in WI38 human primary fibroblasts failedto induce growth arrest, whereas PML IV or oncogenic Ras did(Fig. 1B) (8, 10). Moreover, Ras- and PML IV-transduced cells

Significance

Promyelocytic leukemia protein (PML) nuclear bodies (NBs) aresubnuclear domains proposed to facilitate posttranslationalmodifications. Among NB partners, proteins are p53 and aremost of the regulators. Overexpression of a single PML splicevariant, PML IV, triggers p53-driven senescence. We demonstratean interaction between PML IV C terminus and the ARF tumorsuppressor. This interaction is required to promote p53SUMOylation and subsequent stabilization. These resultsunexpectedly bridge three key tumor suppressors and stressthe key role of PML NBs as SUMOylation factories and regu-lators of senescence.

Author contributions: M.L.B. and H.d.T. designed research; L.I., Y.T., F.J., O.A., and M.L.B.performed research; F.J. contributed new reagents/analytic tools; L.I., Y.T., M.L.B., andH.d.T. analyzed data; and L.I., M.L.B., and H.d.T. wrote the paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.1M.L.B. and H.d.T. contributed equally to this work.2To whom correspondence should be addressed. Email: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1507540112/-/DCSupplemental.

14278–14283 | PNAS | November 17, 2015 | vol. 112 | no. 46 www.pnas.org/cgi/doi/10.1073/pnas.1507540112

Dow

nloa

ded

by g

uest

on

Mar

ch 1

5, 2

020

Page 2: PML IV/ARF interaction enhances p53 SUMO-1 conjugation ... · PML IV on p53 regulators, we first investigated SUMO modi-fication of HDMX and HDM2. In HeLa cells or H1299 cells stably

exhibited senescence-associated β-galactosidase activity (SA-β-Gal), whereas the two PML IV C-terminal mutants did not(Fig. 1C). In WI38 fibroblasts, PML I, IV, or IVΔC induced p53recruitment within NBs, as previously shown (8). However, al-though PML IV stabilized p53 (8–10), the mutants defective forsenescence induction did not (Fig. 1D). p53 silencing by shRNAreversed PML IV-induced growth arrest (8). Thus, PML IVstabilization of p53 and activation of senescence require thethree bulky hydrophobic/cyclic residues within its C terminus.In an attempt to identify p53 target genes specifically activated

by PML IV, we compared the transcriptional changes elicited byPML IV or PML IVΔC. We performed microarray analysis inWI38 cells at early time points (12, 18, and 24 h) after infectionwith PML-expressing lentiviruses. A twofold induction of PMLexpression was demonstrated as early as 12 h postinfection, to-gether with an unambiguous modulation of genes known to beregulated by PML [IFN targets (28, 29) and TBX2 (21)]. Im-portantly, no significant difference between PML IV and PMLIVΔC transcriptional profiles was observed, suggesting that theprimary changes elicited by PML IV are not transcriptional. Yetsubsequent p53 activation should modulate gene expression at latertime points, when the senescence program becomes activated.

PML IV Induces p53–SUMO-1 Conjugation. Upon stress, NBs facili-tate partner SUMOylation through recruitment of UBC9 (1, 5).P53 can be SUMO-modified on a single consensus site K386 inits C-terminal regulatory region (30). To assess whether PML IVcould facilitate p53 SUMO conjugation, we transiently trans-fected p53, or a p53 mutant defective for SUMOylation, inH1299 p53-null cells stably expressing His-tagged SUMO-1 or -2(H1299-HisS1 or H1299-HisS2). His purification demonstrated abasal p53 modification on K386 by either SUMO-1 or SUMO-2(Fig. 2A). Remarkably, PML IV enhanced p53 SUMOylation bySUMO-1, but not SUMO-2, whereas PML I or PML IVΔC didnot (Fig. 2A). As expected, ARF expression also sharply en-hanced p53 SUMO-1 conjugation (Fig. 2A) (23, 24, 26). Similarresults were obtained in stable HeLa SUMO-1 transfectants. InPML IV-transduced WI38 cells, when analyzing p53/SUMO-1physical vicinity using in situ Proximity Ligation Assay (PLA), weobserved a significant increase in the interactions of endogenousSUMO-1 and p53 in PML IV- but not PML IVΔC-transducedcells (Fig. 2B). In contrast, PML IV did not enhance p53/SUMO-2interactions (Fig. S2) (31).We then questioned whether PML IV-enhanced p53

SUMOylation could be responsible for the p53 stabilizationobserved in WI38 cells. Cotransfection of p53 and PML IV inH1299-HisS1 cells sharply enhanced p53 expression (Fig. 2C). In

contrast, point mutants of the p53 SUMOylation site or of theSUMOylation consensus (p53K386R or p53E388A) were notstabilized upon PML IV expression (Fig. 2C), demonstrating thatPML IV-triggered p53 stabilization requires its SUMOylation.PML IV-induced senescence was proposed to rely on enhancedacetylation of p53 on K382 (10, 17, 20). In our experimentalconditions, PML IV-enhanced K382 acetylation paralleled p53stabilization (Fig. 2C). Conversely, mutation of the K382 acety-lation site did not affect p53 SUMOylation (Fig. 2D) or stabili-zation by PML IV.To further assess the SUMO paralog specificity of PML IV-

enhanced p53 activation, we cotransduced PML IV in combi-nation with SUMO paralogs in WI38 cells. PML IV-dependentp53 stabilization was modestly enhanced upon SUMO-1 coex-pression, leading to massive p21 induction (Fig. 2E). Strikingly,overexpression of SUMO-2 antagonized PML IV-enhanced p53stabilization and p21 activation, unraveling a strict SUMO paralogselectivity of this process. Collectively, PML IV enhances p53SUMO-1 conjugation, resulting in its stabilization and activation.

PML IV Enhances Global SUMOylation. Most p53 signaling partnersshuttle onto PML NBs (16, 32, 33). To examine the impact ofPML IV on p53 regulators, we first investigated SUMO modi-fication of HDMX and HDM2. In HeLa cells or H1299 cellsstably expressing His–SUMO-1 or -2, ARF sharply stabilizedeither HDMX or HDM2 and enhanced their SUMO-1 orSUMO-2 conjugation, as expected (23, 34) (Fig. 2F and Fig.S3A). Importantly, PML IV (but not IVΔC or IV3A) selectivelytriggered SUMO-1 modification of HDMX but had no effect onHDM2 (Fig. 2F and Fig. S3A).Moreover, in several human cell lines (WI38 primary fibro-

blasts, H1299 and H358 cell lines), PML IV, but not PML IVΔC,enhanced global SUMOylation (Fig. 2G and Fig. S3 B and C).This was observed for both endogenous and stably overex-pressed SUMO-1 and much less or not at all for SUMO-2. Thus,PML IV enhances global SUMO-1 conjugation but likely retainssome target selectivity, most likely for NB partners.

PML IV Specifically Interacts with ARF. We then investigated NB’sassociation of partners that could facilitate SUMO-1 conjugationby PML IV. Human ARF was proposed to promote SUMOy-lation and to localize into NBs in virus-infected cells (35, 36). Wethus investigated a possible interaction between PML IV andARF. By immunoprecipitation, overexpressed PML IV and ARFwere shown to interact (Fig. 3A). This PML–ARF coimmuno-precipitation was observed with all mutants expressing exon 8b(Fig. S4 A–D). This interaction led to a partial relocalization of

A

C D

B

Fig. 1. PML IV C terminus is required for senes-cence induction. (A, Top) Comparison of humanPML isoform I and IV. (Middle) Sequence alignmentof human and murine PML IV C termini. The dottedline indicates the junction between exons 8a and8b. The three conserved aromatic residues areboxed. (Bottom) PML IVΔC and PML IV3A mutants.(B) Proliferation of primary human WI38 fibroblastsinfected by lentiviruses expressing the indicatedconstructs. (C) Quantification of SA-β-Gal staining ofinfected WI38 cells after 10 d (mean ± SD) from fourindependent experiments. (D) Western blot analysisof p53 and PML in WI38 cell extracts 3 d aftertransduction with indicated constructs.

Ivanschitz et al. PNAS | November 17, 2015 | vol. 112 | no. 46 | 14279

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Mar

ch 1

5, 2

020

Page 3: PML IV/ARF interaction enhances p53 SUMO-1 conjugation ... · PML IV on p53 regulators, we first investigated SUMO modi-fication of HDMX and HDM2. In HeLa cells or H1299 cells stably

ARF to PML NBs (Fig. 3B). Interestingly, PML IV K160R, aSUMOylation mutant that fails to recruit most NBs partners (4),still recruits ARF (Fig. 3 A and B). In stable PML transfectants

derived from primary pml−/− mouse embryo fibroblasts (MEFs),only PML IV, but not other isoforms (PML I or V), was able torecruit ARF from the nucleolus into NBs (Fig. 3C). Localization

* *

PML IV C

PML IV

Empty rotcev

A

His

p53 normalized inputs

PMLARF

p53

IV-

C -

--

-+

I-

p53 K386R

IV-

C -

--

-+

I-

H1299-HisS1 cells

55

70kDa

PML100

130

p53-S1

p53

p53-S2

p53

H1299-HisS2 cells

55

70kDa

PML100

130

B

PML IV C

PML p53-S1 Merge

Ctrl

PML IV

PLA endogenous p53-S1

WI38 cells

C

Ac p53K382

PML-SUMO

p53

*

actin

55

55

45

H1299-HisS1 cells

kDa

PML100

130

250

C - IVIV - C

p53 p53 K386R

- IV C

p53 E388A

PML

E Empty vector

WI38 cells

SUMO-1 SUMO-2

PML IV C-

p53

p21

GFP25

25

55kDa

100

130

C- IV C- IV

D

PML-SUMO

His

p53 normalized inputs

55

H1299-HisS1 cells

70 p53-S1

p53

kDa

PML CIV-p53 K382R

PML100

130

His

p53 normalized inputs

FPMLARF

HDMX

IV-

C -

--

-+

I-

IV-

C -

--

-+

I-

His

Inputs

HDMX-SUMO

HDMX

HeLa-HisS1 cells HeLa-HisS2 cells

80

100

130kDa

PML

Inputs130

250

100

kDa

PML -SUMO

GPML

100

130

250

70

SUMO-1

- CIV - CIV

SUMO-2

kDa

kDa

GFPPML

30

100PML-SUMO

WI38 cells

- CIV

crude extracts

PML

PMLARF

p53

IV-

C -

--

-+

I-

p53 K386R

IV-

C -

--

-+

I-

Fig. 2. PML IV increases p53 SUMOylation by SUMO-1. (A) H1299 cells stably expressing His–SUMO-1 or His–SUMO-2 were transfected with the indicatedexpression vectors 2 d before analysis. Purification of His–SUMO-1 (Top) and His–SUMO-2 (Right) conjugates followed by p53 Western blot. Samples werenormalized for p53 expression to compare SUMOylation efficiency. (B) Conjugation of endogenous p53 by SUMO-1 was assessed by PLA in WI38 cells. (Left)Quantification of p53-S1 amplified dots (mean ± SD from three independent experiments). *P < 0.05. (Right) p53–SUMO-1 red dots; PML was detected byimmunofluorescence (green). (Scale bar, 5 μm.) (C) H1299-HisS1 cells were transfected with p53, p53K386R, or p53E388A, in combination with empty vector,PML IV, or PML IVΔC. Protein extracts were immunoblotted with anti-PML, p53, acetyl K382 p53, and actin antibodies. Asterisk, nonspecific band. (D) H1299-HisS1 cells were transfected with p53K382R acetylation mutant, in combination with empty vector, PML IV, or PML IVΔC. Purification of His–SUMO-1 con-jugates was followed by p53 Western blot. (E) Cotransduction of PML with SUMO paralogs in WI38 cells. Protein extracts were normalized for GFP level andimmunoblotted with PML, p53, p21, and GFP antibodies. (F) His–SUMO-1 or His–SUMO-2 purification was performed in stably SUMO-expressing HeLa cells,transiently transfected by HDMX and PML or ARF constructs. Shown is the immunoblotting with HDMX or PML antibodies. (G) WI38 primary cells weretransduced with PML expression vectors. After 3 d, global conjugation of endogenous SUMO-1 (Left) or SUMO-2 (Right) was analyzed by Western blot. Levelsof GFP and PML are shown as controls.

14280 | www.pnas.org/cgi/doi/10.1073/pnas.1507540112 Ivanschitz et al.

Dow

nloa

ded

by g

uest

on

Mar

ch 1

5, 2

020

Page 4: PML IV/ARF interaction enhances p53 SUMO-1 conjugation ... · PML IV on p53 regulators, we first investigated SUMO modi-fication of HDMX and HDM2. In HeLa cells or H1299 cells stably

of other nucleolar proteins like B23/NPM was unaffected byPML IV (Fig. S5). Using isoform-specific PML antibodies (6), wedemonstrated endogenous PML IV/ARF interaction by coimmu-noprecipitation in 293T cells (Fig. 3D). Existence of this PML IV/ARF interaction was also substantiated by a significantly increasedPLA in PML IV-transduced WI38 cells (Fig. 3E), PML IV/ARF-cotransduced WI38 cells (Fig. S6B), or H1299-transfected cells(Fig. S6C). Collectively, these results demonstrate that PML IVspecifically interacts with ARF through its C terminus.

ARF Binds to and Stabilizes UBC9. PML IV-induced p53 SUMOylationwas dampened upon ARF extinction, suggesting that ARFcould be implicated in PML IV-enhanced SUMOylation of p53(Fig. 4A). Global ARF-enhanced SUMOylation was suggestedto rely on its ability to interact with UBC9 (26). Indeed, UBC9protein and ARF coimmunoprecipitated (Fig. 4B). The associationbetween endogenous ARF and UBC9 was further demonstrated byPLA in WI38 cells (Fig. 4C) and was significantly enhanced by PMLIV, but not PML IVΔC, expression (Fig. 4C and Fig. S6). Moreover,we observed a sharp stabilization of transfected UBC9 upon PMLIV or ARF expression, with a synergistic impact for PML IV–ARFcombination, which is suggestive for complex formation (Fig. 4D).PML binds UBC9 through its RING domain and ARF through its Cterminus, which could favor formation of a tripartite complex thatcould explain their mutual stabilization in an environment with ac-tive proteolysis (37, 38).

NB Recruitment of Partners Is Essential for PML IV-Driven Senescence.Our studies suggest that ARF recruitment within NBs enhancesactivity of UBC9, notably on p53 and its modulators, and drives PMLIV-initiated senescence. However, a previous study argued that PMLIV does not need to form NBs and/or recruit a partner to initiatesenescence. A single SUMOylation site in PML, K160, is requiredfor recruitment of partner proteins through their own SUMO-interacting motif (SIM) (4, 5). We therefore assessed whetherPML IV K160R would induce senescence. Remarkably, PML IVK160R did not affect the growth of WI38 cells (Fig. 5A) or p53activation, as shown by p21 stabilization 48 h postinfection (Fig.5B). Absence of p53 stabilization upon expression of PML IVK160R was also observed upon transfection of HeLa-S1 (Fig. S7).We also cotransduced PML IV with CMV IE1, a protein dis-

rupting PML NBs or its variant IE1core, which spares NB structure

and only impedes partner recruitment (39). Critically, coexpressionof either IE1 constructs with PML IV completely reversedgrowth arrest, whereas these viral proteins alone did not signif-icantly alter cell proliferation (Fig. 5C). Therefore, PML IV-drivensenescence relies on its ability to recruit partners, such as ARF andp53, into NBs (Fig. 5D).

DiscussionWe unravel a specific interaction between two key senescencedrivers, PML IV and ARF. This interaction likely accounts forPML IV control over p53 stability, activation, and subsequent

A B

H1299 cells

Ctrl I IV C K160R

PML

ARF

Merge

3A

pml-/- MEF cells

MergePML I

PML IV

PML V

PML

C D

WI38 cells

PLA PML-endogenous ARF

PML PML-ARF MergeCtrl

PML I

PML IV

EIn

put

Ctrl ARFIP

ARF

Total PML

PML III

PML IV

Endogenous proteins

293T cells

70

15

100

100kDa

IP PML

PML

ARF

IP ARF

PML

ARF

IV C 3A

ARF + PML

InputsPML

ARFSaOs-2 cells

15

100

15

100

15

100kDa

K16

0R PML

Fig. 3. PML IV specifically interacts with ARFthrough its C terminus. (A) ARF or PML immuno-precipitations followed by PML or ARF Western blotin SaOs-2 cells. Inputs are shown. (B) PML (green)and ARF (red) immunostaining in H1299 cellstransfected as indicated. Arrows point to NBs wherethe two proteins colocalize. (Scale bar, 5 μm.)(C) Primary pml−/− MEFs were stably transduced byexpression vectors encoding PML I, IV, or V isoformsand ARF. Cells were labeled with anti-PML antibody(red) and anti-ARF antibody (green). (Scale bar,5 μm.) Arrows denote PML/ARF colocalization.(D) Interaction of endogenous PML IV and ARF wastested by coimmunoprecipitation in 293T cells. Con-trol serum (Ctrl) or anti-ARF antibody was used forimmunoprecipitation. PML was revealed either withpan-PML antibody or isoform-specific antibodies (6).(E) Empty vector (Ctrl), PML I, or PML IV was trans-duced in WI38 cells. PLA was assessed between PMLand endogenous ARF (red dots), and PML wasimmunostained (green). (Scale bar, 5 μm.)

A B

C D

Fig. 4. PML IV/ARF interaction promotes UBC9 stabilization and p53SUMOylation. (A) H1299-HisS1 cells were transfected with p53 and PML IV-expressing constructs in combination with control or ARF shRNA. His puri-fication was followed by p53 Western blot. Samples were normalized for p53expression to compare SUMOylation efficiency. (B) H1299-HisS1 cells weretransfected as indicated. ARF or HA immunoprecipitation is analyzed byWestern blot. IP Ctrl, immunoprecipitation with nonrelevant serum. (C) Emptyvector (Ctrl), PML IV, or PML IVΔC expression vectors were transduced in WI38cells. PLA was assessed between both endogenous UBC9 and ARF (Left), anddots per cell were quantified (means from 50 cells) (Right). *P < 0.05; **P <0.01. (D) UBC9 protein is stabilized by ARF or PML IV in H1299-HisS1 cells. Theremaining UBC9 signal is indicated (*).

Ivanschitz et al. PNAS | November 17, 2015 | vol. 112 | no. 46 | 14281

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Mar

ch 1

5, 2

020

Page 5: PML IV/ARF interaction enhances p53 SUMO-1 conjugation ... · PML IV on p53 regulators, we first investigated SUMO modi-fication of HDMX and HDM2. In HeLa cells or H1299 cells stably

senescence induction. These unexpected results shed a new lighton the relationship between PML, ARF, or p53 and further linkPML NBs to regulation of global SUMOylation.Most studies assessing the role of PML on p53 function used

PML IV, the only isoform that stabilizes p53 and induces se-nescence. However, to date, the molecular basis for this strikingselectivity remained poorly understood. Here, we demonstratethat the C terminus of PML IV is required for ARF binding, p53SUMOylation, stabilization, and activation. Although ARF no-toriously engages in nonspecific binding when overexpressed, thespecific interaction between endogenous ARF and PML IV (Fig.3 C–E) is supportive for a physiologic relevance. When expressedin pml−/− MEFs, PML IV may be observed surrounding thenucleolus, where ARF is normally located and fusion to GFP ofan exon 8b-containing peptide enforced its nucleolar localization(40). Recruitment of ARF in the p53-enriched PML NB envi-ronment can activate p53 signaling by multiple mechanisms. First,PML IV/ARF-dependent p53 SUMO-1 conjugation is critical forp53 stabilization (Fig. 2C). Note that P53 SUMO-1 conjugationwas implicated in apoptotic control or senescence (11, 41) andthat the unexpected SUMO-1 paralog specificity for p53 stabili-zation and p21 activation deserves further investigation. Second,ARF directly interacts with several NB-associated p53 regulators,such as HDM2 (Fig. 5D). Third, NB-associated p53 regulators(HIPK2, HAUSP, DAXX, CBP, HDM2, SIRT1, Chk2, MageA2,or MOZ) may all be SUMOylated. PML and/or ARF-enhancedSUMOylation could alter their functions, as proposed for HDMX(34). Note that NB-enhanced p53 signaling is not always asso-ciated with p53 stabilization (Fig. 5B) (12), suggesting that

PML-enhanced PTMs may actually suffice for p53 activationand senescence induction.As proposed for DNA damage domains, recruitment of the

SUMOylation machinery into PML NBs targets a functionallydefined group, rather than an individual protein (42). NBs ap-pear as scaffolds that facilitate interactions of ARF, UBC9, andmultiple p53 regulators to favor their reciprocal SUMOylationand interactions, ultimately activating p53 signaling. However,that PML IV, physiologically a minor isoform (6), fails to triggersenescence in pml−/− cells (8) stresses the importance of theother, more abundant, isoforms. These recruit p53 and HDM2(16, 32, 33) and could enforce efficient NB recruitment of otherp53 modifiers by SUMO/SIM interactions (5). A previous studyhad argued that PML IV-initiated senescence does not requireNBs (8). Technical differences in the lentiviral expression vectorused and culture protocols that result in higher PML expressionlevels may explain these discrepancies. NBs facilitate a numberof PTMs by a scaffolding effect, concentrating enzymes andprotein substrates within a restricted domain (1). With highlevels of PML expression, NB formation may no longer be re-quired for p53 activation.ARF tumor-suppressive activities are only partially p53-

dependent and were proposed to also rely on enhanced globalSUMO-1 conjugation. ARF interacts with recombinant UBC9in vitro and favors global SUMO-1 conjugation in cell lines(26). Our results suggest that PML IV favors endogenousARF/UBC9 interaction, at least in part by recruiting themwithin NBs (Fig. 4C). The ARF/UBC9 interaction stabilizesthe proteins (Fig. 4 B and D) and could also enhance the in-trinsic activity of UBC9. In that respect, PML IV sharplyenhanced SUMO-1 conjugation of a p53–UBC9 fusion (Fig.S8). These enhanced ARF/UBC9 interactions could explainglobal PML IV-mediated hyper–SUMO-1 conjugation (Fig.2G), which might contribute to PML IV-driven senescenceindependently of p53 activation (Fig. 5D).Our findings have important implications for cancer biology,

as they directly link two major senescence drivers and upstreamactivators of p53. These results highlight the importance of PML orARF loss in tumors (1, 43) and that of SUMOylation in cancerprogression (44, 45).

Experimental ProceduresPlasmids, Transfection, and Transduction. ARF, PML isoforms I and IV, and PMLIV mutants were expressed from the pSG5 vector. Oncogenic Ras was clonedin the pLPC vector, p53 in pCMV, HDM2 and HDMX in pcDNA3.1(+), and ARFshRNA in pSUPER (kindly provided by S. Lain, Karolinska Institutet, Stock-holm). PML IV mutants (ΔC, 3A, K160, ΔCC, and Δ8a) or p53 K386R andE388A mutants were obtained by site-directed mutagenesis (Quick ChangeKit, Stratagene). HA-tagged UBC9 expression vector was obtained fromAddgene (p3258, hCMV huUBC9-HA). PML RING-finger, B-boxes, coiled-coildomain (RBCC) fused to exon 8b (RBCC-8b) was obtained by assembly PCR. Forlentiviral transduction, the pWPI expression vector containing GFP cDNA andeither PML isoforms ARF or His-SUMO cDNAs was used, and the pTRIP vectorwas used for p53 extinction. For retroviral transduction, all of the constructswere expressed from MSCV-based retroviral vectors. Stable H1299 and HeLacells expressing His–SUMO-1 or -2 were obtained by lentiviral transductionand GFP sorting. Transient transfections were performed with CaCl2 orEffectene Transfection Reagent (Qiagen) and WI38 nucleofection withAmaxa technology (Lonza).

Cell Culture, β-Galactosidase Staining, and BrdU Labeling. H1299, SaOs-2,HEK293T, HeLa, and MEF cell lines were grown in 10% FCS-supplementedDMEM (GIBCO). WI38 primary cells were cultured in Eagle’s minimalessential medium (EMEM) (ATCC) supplemented with 10% FCS andessential amino acids (GIBCO). WI38 population doublings were analyzedas previously described (8). SA-β-Gal activity was detected according tothe manufacturer’s instructions (Senescence β-Galactosidase Staining Kit,Cell Signaling).

CA

D

0 3 6 9 12 15 (days) 0 2 5 8 12 15 (days)

Pop

ulat

ion

doub

lings

Pop

ulat

ion

doub

lings

BPML IV K

160R

wt-

p53

actin

PML

p21

WI38 cells

kDa 130

100

55

45

25

0

5

10

15empty vectorPML IVempty vector + IE1corePML IV + IE1core

0

2

4

6

8

empty vectorPML IVPMLIV K160R

Fig. 5. NB recruitment of partners is essential for PML IV-driven senescence.(A) Proliferation of primary human WI38 fibroblasts infected by lentivirusesexpressing the indicated constructs. (B) Western blot analysis of PML, p53,p21, and actin in WI38 cell extracts 2 d after transduction. (C) Proliferation ofprimary human WI38 fibroblasts transduced by PML IV-expressing lentivi-ruses in combination with the IE1 core (39). (D) PML IV/ARF-controlled se-nescence model. Although UBC9 is recruited through the PML RBCC domain(5), PML IV specifically interacts with ARF, enhancing formation of an ARF/UBC9 complex within PML NBs. This leads to an increase of SUMO-1 conju-gation of p53 and some of its partners. ARF enrichment into PML NBs alsoenhances ARF-mediated HDM2 inhibition, leading to p53 stabilization. Fi-nally, enhanced global SUMOylation could also promote senescence. RING,RING-finger domain; S1, SUMO-1.

14282 | www.pnas.org/cgi/doi/10.1073/pnas.1507540112 Ivanschitz et al.

Dow

nloa

ded

by g

uest

on

Mar

ch 1

5, 2

020

Page 6: PML IV/ARF interaction enhances p53 SUMO-1 conjugation ... · PML IV on p53 regulators, we first investigated SUMO modi-fication of HDMX and HDM2. In HeLa cells or H1299 cells stably

Protein Purification, Immunoprecipitation, and Western Blot. His-tagged pro-tein purification was performed after lysis into buffer (6 M guanidium–HCl,0.1 M NaH2PO4/Na2HPO4, 10 mM imidazole pH 8). NiNTA resin from Invi-trogen was incubated for 2 h, washes were performed with decreasingamounts of guanidium–HCl, and elutions were performed in Laemmli bufferwith 200 mM imidazole. For PML or ARF immunoprecipitation, cells werelysed (lysis buffer, 10 mM Tris·HCl pH 7.5, 120 mM NaCl, 1 mM EDTA, 0.5%Nonidet P-40, 1 mM PMSF, 0.1% SDS, 1 mM DTT, protease inhibitor CompleteMini Roche). Immunoprecipitation was performed using our own designedPML rabbit polyclonal antibody or ARF monoclonal antibody (Calbiochem,clone 14PO2) and protein A agarose beads (Roche). Western blots were per-formed using rabbit polyclonal PML isoform antibodies (6), chicken polyclonalPML antibody, p53 monoclonal DO-1 antibody (Santa Cruz, sc-126), Acetyl-p53K382 polyclonal antibody (Cell Signaling, 2525S), ARF polyclonal antibody(Biosource, clone AHZ0472), actin monoclonal antibody (Sigma, 20–33), HDM2monoclonal antibody (Santa Cruz, SMP14), HDMX monoclonal antibody(Bethyl, A300-287A), SUMO-1 monoclonal antibody kindly provided by M. J.Matunis, Johns Hopkins Bloomberg School of Public Health, Baltimore, SUMO-2 polyclonal antibody (Invitrogen, 51–9100), GFP polyclonal antibody (SantaCruz, sc-8334), UBC9 polyclonal antibody (Santa Cruz, clone H-81), and anti-HisTag monoclonal antibody (Novagen, 71941). Secondary antibodies werecoupled to peroxidase (Jackson Immunoresearch).

Immunostaining, PLA, and Microscopy Image Acquisition. Prepermeabilized(0.1% Triton-X100) cells were fixed in 4% paraformaldehyde and wereimmunolabeled with the indicated antibodies in 0.1% Triton-X100 and 1%

BSA. Primary antibodies using p53 (DO-1, Santa-Cruz, sc-126), chicken PML,ARF (Biosource, AHZ0472), and SUMO-1 polyclonal antibody (Cell Signaling,4930) were detected by secondary antibodies (from Jackson Immuno-Research)coupled to fluorochromes [Alexa Fluor 488 or 594 or aminomethylcoumarinacetate (AMCA)] withMowiol mounting medium. In situ PLAs were performedusing the Duolink technology (Olink Bioscience). Confocal analyses were per-formed with a LSM510 meta-laser microscope (Carl Zeiss Micro-Imaging, Inc.)with a Plan-Apochromat 63× N.A. 1.4 oil immersion objective at 20 °C. Imageswere acquired with LSM510 software (Carl Zeiss).

Statistical Analyses. Bilateral Student’s t tests were used to assess the sta-tistical significance of observed differences. All error bars are SD.

Note Added in Proof. ARF was similarly shown to cooperate with TRIM28, aPML family member, to promote NPM1 SUMOylation (46).

ACKNOWLEDGMENTS. We thank T. Stamminger for the kind gift of IE1expression vectors. We thank N. Setterblad for imaging; C. Larsen for advice;and V. Lallemand-Breitenbach, D. Ribet, C. Esnault, and M. Ogrunc for crit-ical reading of the manuscript. The H.d.T. laboratory is supported by theLigue Nationale contre le Cancer, INSERM, CNRS, University Paris Diderot, Insti-tut National du Cancer, the Association pour la Recherche contre le Cancer (PrixGriffuel), European Research Council Senior Grant 268729–STEMAPL (to H.d.T.),and French National Research Agency “Investissements d’Avenir” ProgramsANR-11-PHUC-002 and ANR-10-IHUB-0002. L.I. is supported by a fellowship fromthe Ligue Nationale contre le Cancer.

1. Lallemand-Breitenbach V, de Thé H (2010) PML nuclear bodies. Cold Spring HarbPerspect Biol 2(5):a000661.

2. de Thé H, et al. (1991) The PML-RAR alpha fusion mRNA generated by the t(15;17)translocation in acute promyelocytic leukemia encodes a functionally altered RAR.Cell 66(4):675–684.

3. Ishov AM, et al. (1999) PML is critical for ND10 formation and recruits the PML-interacting protein daxx to this nuclear structure when modified by SUMO-1. J CellBiol 147(2):221–234.

4. Lallemand-Breitenbach V, et al. (2001) Role of promyelocytic leukemia (PML) sumo-lation in nuclear body formation, 11S proteasome recruitment, and As2O3-inducedPML or PML/retinoic acid receptor alpha degradation. J Exp Med 193(12):1361–1371.

5. Sahin U, et al. (2014) Oxidative stress-induced assembly of PML nuclear bodies con-trols sumoylation of partner proteins. J Cell Biol 204(6):931–945.

6. Condemine W, et al. (2006) Characterization of endogenous human promyelocyticleukemia isoforms. Cancer Res 66(12):6192–6198.

7. Cuchet D, et al. (2011) PML isoforms I and II participate in PML-dependent restrictionof HSV-1 replication. J Cell Sci 124(Pt 2):280–291.

8. Bischof O, et al. (2002) Deconstructing PML-induced premature senescence. EMBO J21(13):3358–3369.

9. Ferbeyre G, et al. (2000) PML is induced by oncogenic ras and promotes prematuresenescence. Genes Dev 14(16):2015–2027.

10. Pearson M, et al. (2000) PML regulates p53 acetylation and premature senescenceinduced by oncogenic Ras. Nature 406(6792):207–210.

11. Marcos-Villar L, et al. (2013) SUMOylation of p53 mediates interferon activities. CellCycle 12(17):2809–2816.

12. Ablain J, et al. (2014) Activation of a promyelocytic leukemia-tumor protein 53 axisunderlies acute promyelocytic leukemia cure. Nat Med 20(2):167–174.

13. Bieging KT, Mello SS, Attardi LD (2014) Unravelling mechanisms of p53-mediatedtumour suppression. Nat Rev Cancer 14(5):359–370.

14. Möller A, et al. (2003) PML is required for homeodomain-interacting protein kinase 2(HIPK2)-mediated p53 phosphorylation and cell cycle arrest but is dispensable for theformation of HIPK domains. Cancer Res 63(15):4310–4314.

15. Kim EJ, Park JS, Um SJ (2003) Identification of Daxx interacting with p73, one of thep53 family, and its regulation of p53 activity by competitive interaction with PML.Nucleic Acids Res 31(18):5356–5367.

16. Wei X, et al. (2003) Physical and functional interactions between PML and MDM2.J Biol Chem 278(31):29288–29297.

17. Langley E, et al. (2002) Human SIR2 deacetylates p53 and antagonizes PML/p53-induced cellular senescence. EMBO J 21(10):2383–2396.

18. Louria-Hayon I, et al. (2003) The promyelocytic leukemia protein protects p53 fromMdm2-mediated inhibition and degradation. J Biol Chem 278(35):33134–33141.

19. Peche LY, Scolz M, Ladelfa MF, Monte M, Schneider C (2012) MageA2 restrains cel-lular senescence by targeting the function of PMLIV/p53 axis at the PML-NBs. CellDeath Differ 19(6):926–936.

20. Rokudai S, et al. (2013) MOZ increases p53 acetylation and premature senescencethrough its complex formation with PML. Proc Natl Acad Sci USA 110(10):3895–3900.

21. Martin N, et al. (2012) Physical and functional interaction between PML and TBX2 inthe establishment of cellular senescence. EMBO J 31(1):95–109.

22. Weber JD, et al. (2000) p53-independent functions of the p19(ARF) tumor suppressor.Genes Dev 14(18):2358–2365.

23. Xirodimas DP, Chisholm J, Desterro JM, Lane DP, Hay RT (2002) P14ARF promotesaccumulation of SUMO-1 conjugated (H)Mdm2. FEBS Lett 528(1-3):207–211.

24. Chen L, Chen J (2003) MDM2-ARF complex regulates p53 sumoylation. Oncogene22(34):5348–5357.

25. Tago K, Chiocca S, Sherr CJ (2005) Sumoylation induced by the Arf tumor suppressor:A p53-independent function. Proc Natl Acad Sci USA 102(21):7689–7694.

26. Rizos H, Woodruff S, Kefford RF (2005) p14ARF interacts with the SUMO-conjugatingenzyme Ubc9 and promotes the sumoylation of its binding partners. Cell Cycle 4(4):597–603.

27. Vivo M, et al. (2009) Downregulation of DeltaNp63alpha in keratinocytes by p14ARF-mediated SUMO-conjugation and degradation. Cell Cycle 8(21):3545–3551.

28. Choi YH, Bernardi R, Pandolfi PP, Benveniste EN (2006) The promyelocytic leukemiaprotein functions as a negative regulator of IFN-gamma signaling. Proc Natl Acad SciUSA 103(49):18715–18720.

29. Kim YE, Ahn JH (2015) Positive role of promyelocytic leukemia protein in type I in-terferon response and its regulation by human cytomegalovirus. PLoS Pathog 11(3):e1004785.

30. Kahyo T, Nishida T, Yasuda H (2001) Involvement of PIAS1 in the sumoylation oftumor suppressor p53. Mol Cell 8(3):713–718.

31. Kwek SS, Derry J, Tyner AL, Shen Z, Gudkov AV (2001) Functional analysis and in-tracellular localization of p53 modified by SUMO-1. Oncogene 20(20):2587–2599.

32. Guo A, et al. (2000) The function of PML in p53-dependent apoptosis. Nat Cell Biol2(10):730–736.

33. Bernardi R, et al. (2004) PML regulates p53 stability by sequestering Mdm2 to thenucleolus. Nat Cell Biol 6(7):665–672.

34. Ghosh M, Weghorst K, Berberich SJ (2005) MdmX inhibits ARF mediated Mdm2 su-moylation. Cell Cycle 4(4):604–608.

35. Kashuba E, et al. (2003) EBV-encoded EBNA-5 associates with P14ARF in extra-nucleolar inclusions and prolongs the survival of P14ARF-expressing cells. Int J Cancer105(5):644–653.

36. Kashuba E, Mattsson K, Klein G, Szekely L (2003) p14ARF induces the relocation ofHDM2 and p53 to extranucleolar sites that are targeted by PML bodies and protea-somes. Mol Cancer 2:18.

37. Lallemand-Breitenbach V, et al. (2008) Arsenic degrades PML or PML-RARalphathrough a SUMO-triggered RNF4/ubiquitin-mediated pathway. Nat Cell Biol 10(5):547–555.

38. Fang NN, et al. (2014) Rsp5/Nedd4 is the main ubiquitin ligase that targets cytosolicmisfolded proteins following heat stress. Nat Cell Biol 16(12):1227–1237.

39. Scherer M, et al. (2014) Crystal structure of cytomegalovirus IE1 protein reveals tar-geting of TRIM family member PML via coiled-coil interactions. PLoS Pathog 10(11):e1004512.

40. Condemine W, Takahashi Y, Le Bras M, de Thé H (2007) A nucleolar targeting signalin PML-I addresses PML to nucleolar caps in stressed or senescent cells. J Cell Sci120(Pt 18):3219–3227.

41. Mauri F, et al. (2008) Modification of Drosophila p53 by SUMO modulates its trans-activation and pro-apoptotic functions. J Biol Chem 283(30):20848–20856.

42. Psakhye I, Jentsch S (2012) Protein group modification and synergy in the SUMOpathway as exemplified in DNA repair. Cell 151(4):807–820.

43. Koken MH, et al. (1995) The PML growth-suppressor has an altered expression inhuman oncogenesis. Oncogene 10(7):1315–1324.

44. Bischof O, Dejean A (2007) SUMO is growing senescent. Cell Cycle 6(6):677–681.45. Ivanschitz L, De Thé H, Le Bras M (2013) PML, SUMOylation, and senescence. Front

Oncol 3:171.46. Neo SH, et al. (2015) TRIM28 is an E3 ligase for ARF-mediated NPM1/B23 SUMOylation

that represses centrosome amplification. Mol Cell Biol 35(16):2851–2863.

Ivanschitz et al. PNAS | November 17, 2015 | vol. 112 | no. 46 | 14283

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Mar

ch 1

5, 2

020