Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Cell, Tumor, and Stem Cell Biology

DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway

Orli Yogev, Keren Saadon, Shira Anzi, Kazushi Inoue and Eitan Shaulian
Orli Yogev
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Keren Saadon
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shira Anzi
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kazushi Inoue
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Eitan Shaulian
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/0008-5472.CAN-07-2865 Published March 2008
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

The dynamic behavior of the nucleolus plays a role in the detection of and response to DNA damage of cells. Two nucleolar proteins, p14ARF/p19ARF and B23, were shown to translocate out of the nucleolus after exposure of cells to DNA-damaging agents. This translocation affects multiple cellular functions, such as DNA repair, proliferation, and survival. In this study, we identify a pathway and scrutinize the mechanisms leading to the translocation of these proteins after exposure of cells to DNA-damaging agents. We show that redistribution of B23 and p19ARF after the exposure to genotoxic stress occurs preferentially when the c-Jun-NH2-kinase (JNK) pathway is activated and is inhibited when the JNK pathway is impaired. The stress-induced translocation of alternative reading frame (ARF) is JNK dependent and mediated by two activator proteins, c-Jun and JunB. Thr91 and Thr93 of c-Jun are required for the translocation, but the transcriptional activity of c-Jun is dispensable. Instead, c-Jun interacts with B23 in a dose-dependent manner. c-Jun itself is excluded from the nucleolus in a JNK-dependent manner. Hence, we suggest that c-Jun translocates B23 and ARF from the nucleolus after JNK activation by means of protein interactions. In senescent cells, JNK activity and c-Jun levels are reduced concomitantly with ARF nucleolar accumulation, and UV radiation does not cause the translocation of ARF. [Cancer Res 2008;68(5):1398–406]

  • JNK
  • B23
  • p19ARF
  • redistribution
  • AP-1

Introduction

A growing body of evidence attributes a new role to the nucleolus, extending beyond its traditional activity of ribosome production, stress response ( 1, 2). Among the proteins that comprise it, some are important regulators of the cell cycle and stress responses. The localization of two of these proteins, p14ARF/p19ARF and B23 (nucleophosmin, numartin, or NO38), is regulated by genotoxic stress ( 3– 5).

The human p14ARF/mouse p19ARF are important tumor suppressors. Knockout of p19ARF underscored its importance in cancer prevention, as p19ARF-deficient animals are more prone to cancer ( 6). The tumor suppressor effects of alternative reading frame (ARF) are mainly relayed by regulation of the p53 pathway ( 7, 8). ARF interacts and inactivates Mdm2, thereby increasing the stability and activity of the tumor suppressor p53 ( 9, 10). Hence, increased expression of ARF results in a p53-dependent growth arrest or apoptosis. However, increased tumor development in triple knockout mice nullizygous for ARF, p53, and Mdm2 showed that ARF acts also in a p53-independent manner ( 11). Some of the p53-independent activity is attributed to its ability to reduce rRNA processing ( 12, 13) and inhibit oncogene-induced transcription ( 14, 15).

ARF augments p53 activity mainly in response to oncogenic stress. Its expression is up-regulated in response to deregulated oncogenic activity due to elevated transcription governed by several transcription factors, such as E2F ( 16), c-myc ( 17), AP-1 ( 18), or by oncogenic Ras ( 19). A second mechanism regulating ARF activity involves the control of its subcellular localization ( 3). The nucleolar localization of ARF raised a model suggesting that ARF sequesters Mdm2 to the nucleolus and thereby releases p53 from its inhibitory effects ( 11). However, later work ( 20) suggested that ARF may also posses growth-suppressing activity when it is located in the nucleoplasm. Moreover, recent studies have actually suggested that retention of ARF in the nucleolus inhibits its activity ( 3, 21), whereas its redistribution to the nucleoplasm enables extended interaction with Hdm2 ( 3). Furthermore, this last study showed that ARF is redistributed in the nucleus in response to DNA damage ( 3).

ARF localization is mostly governed by interactions with the abundant nucleolar protein B23 ( 22, 23). B23 functions as a molecular chaperone that shuttles between the nucleus and the cytoplasm. It affects cellular proliferation, resistance to DNA damage–dependent cell death, and maintenance of genomic stability, in addition to its activities in ribosome biogenesis and rRNA processing ( 24). Its interactions with ARF direct ARF to the nucleolus. In acute myeloid leukemia cells containing cytoplasmic mutants of B23, ARF is cytoplasmic as well ( 25, 26), and in cells deficient in B23, ARF is excluded from the nucleolus ( 27). Independently of ARF, B23 interacts with two key proteins in cell cycle regulation, Hdm2 and p53 ( 5, 28). Both of these interactions seem to lead to p53 stabilization ( 5, 28), in agreement with this hypothesized role of the protein in the maintenance of genomic stability. Moreover, increased expression of B23 has been associated with improved DNA repair ( 29), and B23 1–deficient cells exhibit increased H2AXγ phosphorylation ( 27). Its redistribution from the nucleolus in response to cytotoxic drugs and genotoxic stresses, such as the inhibition of RNA polymerase I and II or exposure to DNA intercalating agents or UV radiation, further supports its role in the response to DNA damage ( 3– 5, 30, 31). It seems that the translocation from the nucleolus is an important regulatory step that controls the effects of B23 on DNA repair.

Several members of the AP-1 transcription factor family are highly responsive to genotoxic stress. c-Jun and JunB are strongly induced by DNA-damaging agents such as UV radiation ( 32). Induction of c-Jun by DNA damage is mostly dependent on its upstream kinase, the c-Jun amino-terminal kinase (JNK; refs. 33, 34). However, JNK-independent inductions of c-Jun and JunB were also observed ( 35). JNK phosphorylates c-Jun at Ser63 and Ser73 and Thr91 and Thr93, and regulates its transcriptional capacity and ability to interact with other proteins ( 36– 38). Induction of c-Jun is important for UV-driven apoptosis ( 39). Reports on the effects of JNK on JunB are mixed. Although early reports suggested that JunB does not possess JNK phosphorylation sites ( 40), a later report showed phosphorylation of Thr102 and Thr104 by JNK ( 41). However, the significance of the induction of JunB by UV is not clear.

Currently, the pathways and mechanisms that regulate the redistribution of B23 and ARF after exposure to DNA-damaging agents are unknown. In light of the importance of these translocations, we explored the molecular mechanisms that regulate them. We show that activation of the JNK pathway is required for dispersal of B23 and p19ARF from the nucleolus. The stress-induced translocation of ARF is mediated by JunB and c-Jun whose phosphorylation at Thr91 and Thr93 is required for the translocation. Moreover, we show that B23 directly interacts with c-Jun, and that JNK-dependent translocation of c-Jun translocates associated B23. We also show that when JNK activity is reduced due to senescence, UV radiation does not trigger the translocation of ARF.

Materials and Methods

Cell lines and tissue culture. All the cells were grown in DMEM supplemented with 10% fetal bovine serum. The cell lines used in this work have been previously described as follows: c-jun−/−p53−/− by Shaulian et al. ( 39), 63/73 Ala by Behrens et al. ( 42), and 91/93 Ala by Laderoute et al. ( 43).

Plasmids. Expression vectors for c-jun, junB, and junD were described previously ( 44). cDNAs for wild-type c-Jun and 63/73Ala were cloned into pcDNA3 to generate cytomegalovirus (CMV)-derived expression vectors. CMV 91/93 Ala was obtained from G. Li and R. Johnson (University of California, San Diego, California). The conversion of Arg 272 and Lys 273 to glutamic acids was performed using the QuikChange site-directed mutagenesis kit II, according to the manufacturer's instructions (Stratagene). Expression vectors for green fluorescent protein (GFP)-fibrillarin and JDP2 were obtained from Y. Shav-Tal and A. Aronheim (Bar Ilan University, Israel and the Technion-Israel Institute of Technology, Israel, respectively). ShRNA for JunB was previously described ( 18).

Antibodies. Polyclonal anti–c-Jun antibody (H-79; Santa Cruz Biotech) was used for c-Jun immunoprecipitation, immunofluorescence staining, and immunoblotting. Monoclonal antibodies KM-1 (Santa Cruz Biotech) and C-J 4C4 (Abcam) were used to detect phospho-Ser63 and phospho-Thr91-c-Jun, respectively by immunoblotting. Polyclonal anti–p19ARF ab80 (Abcam) was used for p19ARF staining and immunoblotting. Monoclonal mouse anti-B23 FC-61991 (Zymed; Invitrogen) was used for immunostaining, immunoprecipitation, and immunoblotting. Monoclonal anti-actin C4 (MP Biomedical) was used for immunoblotting. For detection of JunB, polyclonal anti-JunB N-17 (Santa Cruz Biotech) was used. HA-Tag 6E2 (Cell Signaling Technology) was used for the detection of HA-tagged proteins by immunofluorescence. For detection of JNK activation, anti–phospho-Thr183/Tyr185 JNK (Cell Signaling Technology) was used. Total JNK levels were determined using the monoclonal antibody G151-333 (BD Biosciences PharMingen).

Immunofluorescence staining. Cells were plated on coverslips and usually harvested 3 h after UV irradiation. Cells were fixed by a 30-min incubation in 4% paraformaldehyde at room temperature. After washes with PBS, cells were incubated with the primary antibody for 1 h at room temperature in PBS containing 3% bovine serum albumin and 0.3% triton. The slides were then washed with PBS with 0.1% Tween and incubated with fluorescently labeled secondary antibody for 40 min at room temperature. Slides were mounted using dibutylpthalate xylene mounting solution (Fluka Biochemika). Images were visualized using an Olympus fluorescence microscope. At least 200 randomly selected cells were counted at each point. We defined redistribution of ARF and B23 when cells exhibiting normal staining of 4′,6-diamidino-2-phenylindole (DAPI) did not exhibit visible nuclear foci of ARF and B23. In all cases, the fraction of cells expressing nucleolar ARF after treatment was scored. The Y axis title and the figure legends specify the 100% reference point of each experiment. Each experiment was repeated at least thrice.

Immunoprecipitations and immunoblotting. For immunoprecipitations, cell were lysed with NP40 lysis buffer [50 mmol/L Tris-HCl (pH 8.0), 120 mmol/L NaCl, 0.5% NP40, 1 mmol/L EDTA plus proteinase inhibitors (1:200 Cocktail III; Calbiochem), 0.1 mmol/L sodium orthovanadate, and 0.1 mmol/L sodium fluoride]. Two milligrams of total cell extract were used per reaction, and standard immunoprecipitation protocols were used throughout. Nuclear extracts were used for detection of JunB by immunoblotting analysis. Each experiment was repeated at least thrice.

Results

JNK activity regulates B23 and p19ARF distribution. The recent studies describing the translocation of the nucleolar proteins B23 and p14ARF after exposure of cells to genotoxic stresses ( 3– 5) prompted us to examine whether signaling specificity exists in the redistribution of these proteins. To examine the phenomena in relatively normal cells, we irradiated mouse embryo fibroblasts (MEF; passage 3) with 30 J/m2 UV and 20 Gy of ionizing radiation and then stained for p19ARF and B23 at 3 h after irradiation. As previously reported, in untreated cells, the majority of p19ARF colocalized with B23 in the nucleoli ( Fig. 1A ), but the nucleolar staining of p19ARF and B23 was significantly reduced after exposure to UV radiation. Interestingly, no change in the distribution of p19ARF and B23 was observed after exposure of the cells to ionizing radiation. To determine whether the reduction in the staining after exposure to UV radiation reflects a reduction in the levels of p19ARF or B23 that anchors p19ARF to the nucleolus, we measured the level of both proteins before and 3 h after UV exposure. Within this time frame, the changes in the levels of p19ARF and B23 were minor ( Fig. 1B). This suggests that the reduced nucleolar staining reflects changes in p19ARF distribution and not a change in the total amounts of p19ARF or B23.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Translocation of p19ARF from the nucleolus correlates with JNK activation. A, early passage MEFs were irradiated with 30 J/m2 UV or 20 Gy of ionizing radiation and stained for p19ARF, B23, and with DAPI 3 h after irradiation. Pictures were taken through a fluorescent microscope. B, the protein levels of p19ARF in the irradiated cells were determined 3 h after irradiation with 30 J/m2 UV. Actin served as a loading control. C, early passage MEFs were irradiated with 30 J/m2 UV or 20 Gy of ionizing radiation or treated with MMC or CDDP and harvested at the indicated times (hours after treatment). The level of phosphorylated JNK was determined using anti–phospho-Thr183/Tyr185-JNK antibody. JNK1 was used as a loading control. D, p19ARF localization was determined by immunostaining the cells treated with UV, ionizing radiation, MMC, and CDDP; and the portion of cells retaining nucleolar p19ARF at 4 h after treatment was plotted. Nucleolar ARF, the fraction of cells expressing nucleolar ARF from the total cell population.

One of the most significant differences between UV and ionizing radiations is the pathways that they activate. Specifically, there is a difference in their abilities to activate the JNK pathway ( 39). Therefore, we explored a possible correlation between JNK activation and translocation of p19ARF in response to the different genotoxic stresses that activate JNK. To this end, we exposed MEFs to 30 J/m2 UV, 20 Gy ionizing radiation, 50 μmol/L cisplatin (CDDP), or 50 μmol/L mitomycin C (MMC) and examined their ability to activate JNK and induce p19ARF translocation. JNK activation was determined by measuring the extent of Thr183/Tyr185 phosphorylation, which is known to activate it (reviewed by ref. 45).

As depicted in Fig. 1C, UV is the most efficient activator of JNK and ionizing radiation is the weakest one. At the concentration examined, MMC activates JNK better than ionizing radiation but less efficiently than CDDP. The stress-dependent translocation by the different agents was closely correlated to the level of JNK activation. Nucleolar staining was retained in 76% of ionizing radiation–treated cells, 47% of MMC-treated cells, 18% of CDDP-treated cells, and in only 15% of the UV-treated cells ( Fig. 1D). To directly examine the involvement of JNK in the distribution of p19ARF and B23, we used two cellular models. As p19ARF expression is elevated in p53-deficient cells, it is easily detected by staining. Irradiation of p53−/− fibroblasts with 30J/m2 UV almost completely dispersed both B23 and p19ARF from the nucleolus ( Fig. 2A–B ). However, a preincubation with 100 μmol/L of JNK inhibitor SP600125 at 1 h before treatment inhibited the translocation of p19ARF from the nucleolus, and the percentage of cells in which nucleolar p19ARF was detected increased >15-fold, from 3.98 to 61.8 ( Fig. 2A–B). As depicted in Fig. 2C, the significant increase in p19ARF staining in the nucleolus is not a result of any increase in its overall quantity. In fact, SP600125 treatment slightly reduced the levels of p19ARF. Significant repression of c-Jun phosphorylation served as a control for the inhibitor activity. To corroborate the notion that JNK controls p19ARF and B23 nuclear localization, we also took a genetic approach and examined ARF translocation after UV exposure in JNK+/+ mouse fibroblasts and JNK 1 and 2 double-knockout fibroblasts (JNK1,2−/−), in which c-Jun is not phosphorylated by JNK in response to UV, and its induction is relatively limited (Supplementary Fig. S1A). As depicted in Fig. 2D, in the absence of JNK expression, the nucleolar staining of both B23 and p19ARF is morphologically distinct, the nucleoli are smaller, and more B23 is located in the nucleoplasm. However, UV irradiation does not trigger the export of p19ARF and B23 from the nucleolus in these cells; thus proving that JNK activity is involved in dispersing them from the nucleolus. Moreover, the dispersal of p19ARF in JNK1,2−/− after UV exposure was comparable with that of and JNK+/+ cells treated with SP600125. Both were significantly higher than in SP600125-untreated, UV-irradiated JNK+/+ cells (Supplementary Fig. S1B).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Export of p19ARF from the nucleolus is JNK-dependent. A, p53−/− mouse fibroblasts were irradiated with 30 J/m2 UV. Control fibroblasts were left untreated. Some of the cells were preincubated with a JNK inhibitor SP600125 (SP) 1 h before irradiation. The cells were than stained for p19ARF and B23. B, quantitative analysis of p19ARF dispersal, which is shown in A. Y-axis, percentage of cells containing nucleolar p19ARF from the total cell population. Empty bars, SP600125-treated cells; black bars, untreated cells. C, protein extracts were prepared from p53−/− fibroblasts treated as indicated in A; and the levels of p19ARF, B23, c-Jun, and phospho-c-Jun were determined by immunoblotting with specific antibodies. Actin was used as a loading control. D, JNK+/+ and JNK1,2−/− double-knockouts were exposed to 30 J/m2 UV, and p19ARF and B23 localization was determined by immunostaining 3 h after irradiation. c, control.

c-Jun and JunB mediate the JNK-dependent redistribution of p19ARF by UV. The results presented above show that the JNK pathway regulates the translocation of p19ARF from the nucleolus. Therefore, it was of interest to identify what JNK substrates mediate p19ARF redistribution. The plethora of JNK substrates complicates this task; however, AP-1 genes are clear candidates. As depicted in Figs. 2 and 3B , UV induces c-Jun and JunB in a kinetic that correlates with the p19ARF redistribution. c-Jun is a primary JNK substrate. Therefore, we initially examined whether the status of c-Jun affects the redistribution of p19ARF after exposure to UV. To that end, we irradiated p53−/−c-jun+/+ and p53−/−c-jun−/− mouse fibroblasts with 30 J/m2 and examined p19ARF localization 3 h later. As depicted in Fig. 3A, p19ARF was also dispersed from the nucleoli of p53−/−c-jun−/− cells. However, the translocation of p19ARF was more extensive in c-jun–expressing cells, as only 4% of the p53−/−c-jun+/+ cells were stained for nucleolar ARF, whereas nucleolar staining of ARF was observed in 49% of the p53−/−c-jun−/− ( Fig. 3A). Preincubation with the JNK inhibitor increased the fraction of cells stained for nucleolar p19ARF by 64% and 27% in p53−/−c-jun+/+ and p53−/−c-jun−/− cells, respectively, suggesting that c-Jun is required for p19ARF translocation, but an additional factor contributes as well. Thus, we examined other AP-1 proteins that may be involved in p19ARF translocation. We knocked down JunB expression with specific small hairpin RNA (shRNA; ref. 18) in low-passage MEFs and examined the redistribution of p19ARF after UV irradiation ( Fig. 3C–D). Indeed, repression of JunB expression also inhibited the translocation of p19ARF from the nucleolus in MEFs ( Fig. 3D). Hence, we conclude that JunB cooperates with c-Jun to translocate p19ARF. To test if the effect of c-Jun and JunB on p19ARF translocation is additive, we knocked down junB in p53−/−c-jun−/− cells to generate c-jun, junB–deficient cells. junB shRNA significantly reduced the levels of JunB (Supplementary Fig. S2A). Irradiation of p53−/−c-jun−/− reduced the portion of cells expressing nucleolar p19ARF to 14%, and inhibition of the JNK pathway prevented translocation and increased the fraction of cells expressing nucleolar p19ARF to 84% (Supplementary Fig. S2B). In contrast, the translocation of p19ARF in p53−/−c-jun−/− shjunB cells was significantly reduced, retaining nucleolar p19ARF in 64% of the cells (Supplementary Fig. S2B). In these cells, the pretreatment with SP600125 only mildly increased the proportion of cells exhibiting nucleolar p19ARF. This result suggests that the effects of c-Jun and JunB are additive.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

c-Jun and JunB are required for UV-induced p19ARF translocation. A, p53−/−c-jun+/+ (striped bars) and p53−/−c-jun−/− (black bars) mouse fibroblasts were exposed to 30 J/m2 UV, with or without preincubation with JNK inhibitor (SP). The cells were stained for p19ARF 3 h after UV exposure, and the percentage of cells containing nucleolar p19ARF at that time is plotted above. The fraction of cells containing nucleolar p19ARF in the untreated control was considered as 100%. B, JunB protein levels were determined by immunoblotting in MEFs exposed to UV and then harvested at the indicated times (hours after treatment). Actin was used as a loading control. C, MEFs were infected with a retrovirus-expressing shRNA against junB or the vector virus alone, and selected. Six days after infection, the cells were exposed to 30 J/m2 UV, and the level of JunB at 3 h after irradiation was determined by immunoblotting. Lamin A and B served as the loading controls. D, the MEFs whose JunB levels are exhibited in C were also stained for p19ARF, and the percentage of cells containing nucleolar p19ARF is plotted. Dark bars, cells expressing the vector; empty bars, cells expressing shRNA for junB.

Overexpression of Jun proteins translocates p19ARF from the nucleolus. UV radiation activates multiple pathways and triggers intensive transcriptional responses. To examine the net effects of Jun proteins on the redistribution of p19ARF, we transfected p53−/−c-jun−/− cells with Ha-tagged c-jun, junB, junD, or JDP2 and then localized p19ARF in the transfected cells. As depicted in Fig. 4A , all Jun proteins dispersed p19ARF from the nucleolus. However, significant differences in their potency were observed. c-Jun redistributed p19ARF most efficiently, and only 33% of the transfected cells retained nucleolar p19ARF. JunB was 1.5-fold less efficient and JunD was relatively inefficient, with p19ARF retained in the nucleoli of 77% of the transfected cells ( Fig. 4A). Jun specificity in p19ARF redistribution is supported by the fact that in addition to GFP, JDP2, another JNK target, did not redistribute p19ARF when overexpressed under the same conditions ( Fig. 4A). The reduced nucleolar staining of p19ARF is not due to reduction in its level, as transduction of c-Jun by viral expression in these cells did not repress p19ARF expression (Supplementary Fig. S3).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Redistribution of p19ARF by Jun proteins. A, p53−/−c-jun−/− cells were transfected with expression vectors coding for HA-tagged c-Jun, JunB, JunD, or with expression vectors coding for GFP or JDP2. Transfected cells were identified by staining against the HA tag at 36 h after transfection, and the localization of p19ARF in Jun-expressing cells was determined using coimmunofluorescent staining. The proportion of cells containing nucleolar p19ARF after GFP transfection was regarded as 100%. Bands above the bars exhibit the level of each of the transfected proteins as determined by immunoblotting. B, c-jun +/+ fibroblasts were preincubated with JNK inhibitor or not, exposed to 30 J/m2 UV, and harvested at the indicated times (h). The levels of phospho-Thr91 (p Thr 91) and phospho-Ser63 (p Ser 63)as well as the total level of c-Jun were determined by immunoblotting. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) served as a loading control. C, p53−/−c-jun−/− cells were transfected with plasmids coding for HA-tagged wild-type (wt) and mutated c-Jun, as indicated (left). The cells were stained and subjected to microscopic determination of p19ARF localization as in A. Levels of transfected c-Jun proteins exhibited above were determined by immunoblotting. Right, p19ARF localization was determined in mouse fibroblasts expressing the indicated c-Jun before or 3 h after exposure to 30 J/m2 UV. The fraction of cells expressing nucleolar ARF from the total cell population is presented. D, expression vectors for wild-type c-jun, c-jun mutant 272/273E, and for GFP were transfected to p53−/−c-jun−/− cells. Costaining for c-Jun and p19ARF was used to determine the localization of the last-in Jun-transfected cells. The proportion of cells containing nucleolar p19ARF after GFP transfection was regarded as 100%.

Two clusters in c-Jun are phosphorylated by JNK after exposure of cells to UV, Ser63 and Ser73, and Thr91 and Thr93 ( Fig. 4B). We determined which JNK phosphorylation sites are required for c-Jun–dependent redistribution of p19ARF by transfecting wild-type c-jun or mutants, in which JNK phosphorylation clusters were substituted to alanines (Ser63/73Ala and Thr91/93Ala) into p53−/−c-jun−/−, and subsequent examination of p19ARF localization ( Fig. 4C, left). Interestingly, the canonical JNK phosphorylation sites, Ser63 and Ser73, were dispensable for p19ARF redistribution as Ser63/73Ala dispersed p19ARF comparably to wild-type c-Jun. On the other hand, Thr91 and Thr93 are required as the Thr91/93Ala mutant is impaired in its ability to disperse p19ARF. To avoid any possible transfection artifacts, we also examined the localization of p19ARF in mouse fibroblasts in which Ser63/73Ala or Thr91/93Ala were substituted for the endogenous wild-type c-jun ( 42, 43). In agreement with the lack of ability of Thr91/93Ala mutant to redistribute p19ARF when overexpressed, p19ARF was not significantly dispersed in the Thr91/93Ala cells 3 h after exposure to UV, whereas in cells expressing wild-type c-Jun, a significant redistribution was observed ( Fig. 4C, right). The p19ARF was redistributed less in Ser63/73Ala cells probably due to the lower level of c-Jun induction in these cells shortly after UV exposure (Supplementary Fig. S4A; ref. 42). These results suggest that Thr91 and Thr93 of c-Jun are important for redistribution of p19ARF. c-Jun is a transcription factor that regulates the expression of numerous genes ( 32). To examine whether the transcriptional activity of c-Jun is required for p19ARF redistribution, we generated a transcriptionally inactive c-Jun mutant by replacing two basic residues, Arg272 and Lys273, in its basic DNA-binding domain with two glutamic acids to generate a mutant incapable of binding DNA. Its ability to induce the transcription of AP-1–controlled reporter gene (3XTRE-Luc) was compared with that of wild-type c-Jun (Supplementary Fig. S4B). Unlike wild-type c-Jun, which enhances the transcription of the reporter gene in a dose-dependent manner and whose transcriptional capacity is augmented by coexpression of its upstream MAP3K, MEKK1, the mutant 272/273E did not activate transcription (Supplementary Fig. S4B). Nevertheless, its transfection into p53−/−c-jun−/− culminated in the dispersal of p19ARF from the nucleolus, comparable with that of wild-type c-Jun ( Fig. 4D). This result suggests that the transcriptional activity of c-Jun is dispensable for the redistribution of p19ARF. To further corroborate this notion, we examined the induction of the c-jun gene, which is subject to autoregulation by the endogenous Jun protein and can therefore serve as an indicator for its activity, in wild-type and Thr91/93Ala-expressing cells after exposure to UV (Supplementary Fig. S4C). Again, the ability to regulate gene expression does not correlate with the ability to redistribute p19ARF, as c-jun mRNA expression increased to the same extent in both cell lines 3 h after exposure to 30 J/m2 UV (Supplementary Fig. S4C), whereas p19ARF was dispersed only in wild-type c-jun–expressing cells ( Fig. 4C).

c-Jun interacts with B23 in a JNK-independent manner and changes its subnuclear localization in a JNK-dependent manner. As the transcriptional activity of c-Jun is dispensable for redistribution of p19ARF, we tested whether its interactions with other proteins contribute to the ability of c-Jun to disperse p19ARF from the nucleolus. We explored two possible interactions: interactions between c-Jun and p19ARF and interactions of c-Jun with B23. Coimmunoprecipitation experiments were performed to determine the validity of the hypothetical interactions. We directly examined interactions of endogenous, not overexpressed, proteins in p53−/− fibroblasts that were exposed to UV (or left untreated) and harvested 3 h later. In addition, we preincubated the same cells with SP600125 to determine the dependency of the interactions on JNK activity. In these experiments, immunoprecipitates of c-Jun did not contain p19ARF (data not shown). However, precipitated c-Jun coprecipitated B23 reproducibly ( Fig. 5A ). The weak interactions observed between c-Jun and B23 in untreated cells were strengthened in UV-irradiated cells. However, these interactions do not correlate with c-Jun phosphorylation by JNK, as preincubation with SP600125, which significantly reduced c-Jun phosphorylation after UV exposure, actually increased the association of c-Jun with B23 (compare UV − SP600125 to UV + SP600125). These interactions are independent of p19ARF expression as they were also observed in extracts from p19ARF-deficient cells (Supplementary Fig. S5). These results suggest that c-Jun interacts with B23 and that this interaction is not dependent on JNK phosphorylation. Indirect interactions between c-Jun and p19ARF were observed in NIH3T3 cells, containing metallothionein promoter–regulated p19ARF 24 h after induction with ZN2+. Immunoprecipitation of p19ARF revealed the presence of B23 as well as low levels of c-Jun in the precipitate, thus, suggesting the presence of a complex containing p19ARF/B23 and c-Jun ( Fig. 5B, left). The hypothesis that c-Jun directly displaces p19ARF from the complex with B23 was directly tested by transfecting HEK293 cells with expression vectors for B23, p19ARF, and c-Jun and examining the amounts of p19ARF that coprecipitated with B23 in the presence of increasing amounts of c-Jun. As depicted in Fig. 5B (right), B23 was precipitated even in nontransfected cells, and comparable high levels were precipitated by the antibody after transfection. Increasing the amount of transfected c-Jun resulted in an increase in the association between B23 and c-Jun ( Fig. 5B, right). Surprisingly, p19ARF interactions with B23 increased rather than decreased, in parallel to the increase in B23 to c-Jun interactions ( Fig. 5B, right). These results show that c-Jun does not prevent p19ARF from interacting with B23. Therefore, we examined the alternative possibility that JNK might control the subnuclear localization of c-Jun and, thus, the localization of interacting B23 as well. It should be mentioned that the localization of several AP-1 proteins was previously found to be regulated by UV ( 46). We, therefore, stained untreated and UV-irradiated JNK+/+ and JNK1,2−/− mouse fibroblasts for c-Jun to detect JNK-dependent changes in its subnuclear localization after UV exposure ( Fig. 5C). In JNK+/+ mouse fibroblasts, c-Jun is distributed throughout the nucleus, including in the nucleolus where B23 is concentrated. After exposure to UV, however, c-Jun is excluded from certain areas suspected to be the nucleoli ( Fig. 5C), in correlation with B23 dispersal. To determine whether the unstained areas are indeed the nucleoli, we transfected the JNK+/+ mouse fibroblasts with an expression vector for GFP-tagged fibrillarin, which is often used as a nucleolar marker, and detected its expression relative to c-Jun in UV-irradiated cells ( Fig. 5C, bottom). This analysis confirmed that the areas from which c-Jun is excluded are indeed the nucleoli. c-Jun staining is also observed in the entire nuclear area of untreated JNK1,2−/− cells. Moreover, regions of intense c-Jun staining that coreside with the nucleolar B23 were also observed ( Fig. 5C, arrows). However, unlike in JNK-expressing cells, c-Jun was not excluded from the nucleolus of irradiated JNK-deficient cells. Quantitative analysis revealed a good correlation between c-Jun exclusion and the export of B23 from the nucleoli ( Fig. 5D). These results suggest that JNK-dependent translocation of c-Jun is correlated with the exclusion of B23 from the nucleolus. Additional experiments showed that B23 is redistributed after UV exposure also in cells expressing Ser63/73Ala c-Jun. However, it is not dispersed in the Thr91/93Ala cells (Supplementary Fig. S6).

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

c-Jun interacts with B23 in a JNK-independent manner and is excluded from the nucleolus in a JNK-dependent manner. A, p53−/− fibroblasts were either exposed to 30 J/m2 UV or not, in the presence or absence of JNK inhibitor SP600125. Endogenous c-Jun was immunoprecipitated from cells harvested 3 h after irradiation, and the levels of immunoprecipitated c-Jun and coprecipitated B23 were detected by immunoblotting using specific antibodies. The right part of the panel shows the level, Ser63 phosphorylated c-Jun, and B23 in the extracts subjected to immunoprecipitation. NC, negative control immunoprecipitate from UV-exposed cells that were incubated with irrelevant antibody. B, p19ARF was precipitated from extracts of control or UV-irradiated NIH3T3 containing metallothionein promoter–regulated p19ARF, 24 h after induction with ZN2+, and levels of coprecipitated endogenous, B23 and c-Jun were determined using specific antibodies (left). Right, HEK293 cells were cotransfected with constant levels of B23 and p19ARF (2 and 4 μg, respectively) and the indicated amounts of c-Jun (in μg). Twenty-four hours after transfection, the cells were harvested, and B23 was immunoprecipitated with a specific antibody. The levels of precipitated B23, c-Jun, and p19ARF were determined by immunoblotting with a specific antibody. C, JNK+/+ and JNK1,2−/− mouse fibroblasts were exposed to UV and stained for B23 and c-Jun 3 h after irradiation. Bottom, JNK+/+ cells were transfected with an expression vector for GFP-fibrillarin. The cells were irradiated at 24 h after transfection and then stained for c-Jun at 3 h after irradiation, using a specific antibody. D, quantitative analysis of nucleolar c-Jun (left graph) and B23 (right graph) before and 3 h after UV exposure. Black bars, staining in JNK+/+ cells; white bars, staining in JNK1,2−/− cells.

p19ARF is not dispersed from the nucleoli of senescent cells. ARF levels increase during progressive passages of explanted MEFs in culture. JNK activity, on the other hand, is reduced in senesced fibroblasts, resulting in defective stress responses in these cells ( 47). To show the importance of the JNK pathway for p19ARF redistribution in a physiologic system in which JNK activity is reduced without the application of experimental manipulation, we simultaneously examined JNK activity and the location of p19ARF in early and late passage MEFs before and after exposure to UV radiation. As depicted in Fig. 6A , both basal and UV-induced JNK-dependent c-Jun and phospho–c-Jun levels are higher in early passage MEFs. Concordantly, in early passage MEFs, p19ARF was efficiently translocated from the nucleoli after UV exposure. In contrast, and in agreement with the hypothesized effects of low JNK activity, little dispersal of p19ARF from the nucleoli of late passage MEFs was observed after UV exposure ( Fig. 6B). This result shows that low JNK activity can result in the prevention of p19ARF redistribution after UV exposure.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

UV does not affect p19ARF localization in senesced cells. A, levels of c-Jun and Ser63-phosphorylated c-Jun were examined in passage 2 and passage 6 MEFs by immunoblotting with specific antibodies. Actin was used as a loading control. B, early passage (P2) and late passage (P6) MEFs were irradiated with UV and the localization of p19ARF was detected by fluorescent immunostaining. C and D, a model describing the activity of the JNK pathway in stress-induced translocation of B23 and ARF. C, cells not exposed to UV. D, UV-irradiated cells.

Discussion

The nucleolus functions in the stress response ( 1, 2, 4) and redistribution of B23 and p19ARF may relay this response ( 3, 4). In this study, we define for the first time a pathway and a mechanism that underlies B23 and p19ARF redistribution. Both are redistributed after the exposure of cells to DNA damage, but this redistribution is selective and occurs efficiently when the JNK pathway is activated. UV radiation, the most potent JNK activator, triggers the redistribution of p19ARF and B23 most effectively, whereas exposure to ionizing radiation, which is a poor activator of JNK, does not result in efficient p19ARF redistribution. Furthermore, inhibition of JNK activity by the synthetic inhibitor SP600125, or in cells deficient in JNK, efficiently prevented the redistribution of p19ARF and B23, thus, proving that the DNA damage-dependent redistribution is JNK-dependent. However, our data suggest that although the JNK pathway is a major regulator of DNA damage–dependent localization of p19ARF and B23, it is not the only one. We show that impairment of JNK activity prevents only 60% to 80% of the UV-dependent redistribution p19ARF ( Figs. 2– 3). In addition, at later times after UV-irradiation, p19ARF may exit the nucleolus in a JNK-independent manner (data not shown). Hence, we suggest that the JNK-dependent redistribution is the early response to the stress.

Out of the plethora of JNK substrates, the Jun proteins regulate the JNK-dependent B23 and p19ARF translocations. p19ARF redistribution in the absence of c-Jun or JunB is reduced, demonstrating that both are required for the redistribution. However, reduced expression of each of these individual proteins partially prevented the redistribution ( Fig. 3A and D), and combined inhibition of both prevents the redistribution after exposure to UV almost as efficiently as inhibition of JNK activity, suggesting that most of the effect is mediated by these proteins (Supplementary Fig. S2). Inhibition of the JNK-dependent activity of JunD may also be required for a complete inhibition of p19ARF redistribution. This notion is supported by two experimental findings: transient expression of JunD into mouse fibroblasts resulted in moderate redistribution of the endogenous p19ARF ( Fig. 4A). In addition, a previous study reported an intense nucleolar staining of p19ARF in junD−/− MEFs ( 48). This was intuitively attributed to the ability of JunD to down-regulate p19ARF expression ( 48). Alternatively, the strong inhibition of translocation when JNK activity is impaired may also stem from direct JNK phosphorylation of either p19ARF or B23 as a precedence for the involvement of JNK in the trafficking of other nucleolar proteins was described in other systems ( 49). This notion, however, is not supported by the fact that in cells expressing c-Jun mutated at the JNK phosphorylation sites Thr91/93 no translocation is observed after UV exposure. We also showed the requirement of the JNK pathway for the distribution of p19ARF in a physiologically relevant system of senesced cells, in which JNK activity and c-Jun levels are low, and p19ARF was not translocated after UV treatment ( Fig. 6A and B).

To study the mechanism of the redistribution, we focused our attention on the c-Jun protein, due to the presence of extensive data and reagents related to its modulation by JNK. Interestingly, our results show for the first time a specific role for the phosphorylation at Thr91/93. These sites are phosphorylated by UV, with a kinetic similar to that of the phosphorylation of Ser63/73 ( Fig. 4B). However, as of today, no biological activity has been assigned to this phosphorylation. Using both cell lines expressing knock-in c-Jun mutants and transient overexpression experiments, we show that the presence of Thr91/93 is important for the redistribution, whereas the activity of Ser63/73 is dispensable. We also show that c-Jun–dependent transcriptional regulation of target genes is not involved in the redistribution of p19ARF. DNA-binding and transcriptionally inactive mutant c-Jun (272/273E) can efficiently redistribute p19ARF ( Fig. 4D). Instead, we identified a more direct mechanism: c-Jun interacts with B23. Surprisingly, nonphosphorylated c-Jun interacts with B23 better than phosphorylated c-Jun ( Fig. 5A). Therefore, our results suggest that the interactions between B23 and c-Jun are JNK-independent. Hence, the activity of JNK may be important for augmenting the reservoir of c-Jun ( 32, 34, 50). The interaction between B23 and c-Jun is p19ARF-independent (Supplementary Fig. S5) and does not enhance the dissociation between B23 and p19ARF ( Fig. 5B). On the other hand, we show that the translocation of Jun and B23 out of the nucleolus is JNK dependent. Therefore, we suggest that, just as for the effects on transcription initiation factor-IA ( 49), JNK is involved in the shuttling of the c-Jun–associated B23. In contrast to the situation in JNK-expressing cells, c-Jun is not excluded from the nucleoli of JNK-deficient cells. This is in perfect correlation with B23 translocation ( Fig. 5C–D). The possibility that the nucleolus could be totally disrupted after irradiation is not valid as fibrillarin, often used as nucleolar marker, is concentrated at nucleolar-shaped structures from which c-Jun is excluded after UV exposure. In addition, when p19ARF was ectopically overexpressed to high levels, UV irradiation caused only partial redistribution of p19ARF (data not shown), thus, suggesting that p19ARF redistribution is a stoichiometric process. In this respect, the involvement of at least two UV-inducible Jun proteins can provide a reasonable stoichiometric relationship to explain the B23 translocation. A recent study by Liu et al. ( 46) offered a possible mechanism by which c-Jun localization might be changed by UV radiation; Jun homodimers are concentrated in nucleolar-like structures; and upon exposure to UV, c-Jun interacts with activating transcription factor 2 and changes its subnuclear position.

A recent model suggested that upon UV exposure, p14ARF dissociates from B23 and interacts with Hdm2 ( 3). B23 itself is also translocated to the nucleoplasm after UV exposure by a mechanism obscure before this study ( 4, 5). Whether B23-associated p19ARF is translocated along with c-Jun and the dissociation occurs at later stage is currently studied. Our model suggests that the induction of the AP-1 pathway is the initial and essential step. We suggest that the JNK activity elevates c-Jun levels, thus, increasing the association between c-Jun and B23, and that the phosphorylation of Thr91/93 by JNK enhances translocation of c-Jun and the associated B23 out of the nucleolus. As B23 anchors p19ARF to the nucleolus, its translocation will inevitably result in the exit of p19ARF from the nucleolus as illustrated in our model ( Fig. 6C and D).

Acknowledgments

Grant support: Grant number 0397583 from the Israeli Science Foundation and grant number 0394234 from the Public Committee for Allocation of Estate Funds, Ministry of Justice, Israel.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank G. Li, R. Johnson, C. Sherr, and A. Behrens for cell lines; Y. Shav-Tal, A. Aroneim, and M.Oren for plasmids; and G. Peters for critical reading and comments.

Footnotes

  • Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

  • Received July 26, 2007.
  • Revision received December 4, 2007.
  • Accepted December 27, 2007.
  • ©2008 American Association for Cancer Research.

References

  1. ↵
    Olson MO. Sensing cellular stress: another new function for the nucleolus? Sci STKE 2004; 2004: 10.
    OpenUrl
  2. ↵
    Mayer C, Grummt I. Cellular stress and nucleolar function. Cell Cycle 2005; 4: 1036–8.
    OpenUrlCrossRefPubMed
  3. ↵
    Lee C, Smith BA, Bandyopadhyay K, et al. DNA damage disrupts the p14ARF-B23(nucleophosmin) interaction and triggers a transient subnuclear redistribution of p14ARF. Cancer Res 2005; 65: 9834–42.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Rubbi CP, Milner J. Disruption of the nucleolus mediates stabilization of p53 in response to DNA damage and other stresses. EMBO J 2003; 22: 6068–77.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Kurki S, Peltonen K, Latonen L, et al. Nucleolar protein NPM interacts with HDM2 and protects tumor suppressor protein p53 from HDM2-mediated degradation. Cancer Cell 2004; 5: 465–75.
    OpenUrlCrossRefPubMed
  6. ↵
    Kamijo T, Zindy F, Roussel MF, et al. Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF. Cell 1997; 91: 649–59.
    OpenUrlCrossRefPubMed
  7. ↵
    Kim WY, Sharpless NE. The regulation of INK4/ARF in cancer and aging. Cell 2006; 127: 265–75.
    OpenUrlCrossRefPubMed
  8. ↵
    Sherr CJ. Divorcing ARF and p53: an unsettled case. Nat Rev Cancer 2006; 6: 663–73.
    OpenUrlCrossRefPubMed
  9. ↵
    Pomerantz J, Schreiber-Agus N, Liegeois NJ, et al. The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2's inhibition of p53. Cell 1998; 92: 713–23.
    OpenUrlCrossRefPubMed
  10. ↵
    Stott FJ, Bates S, James MC, et al. The alternative product from the human CDKN2A locus, p14(ARF), participates in a regulatory feedback loop with p53 and MDM2. EMBO J 1998; 17: 5001–14.
    OpenUrlAbstract
  11. ↵
    Weber JD, Jeffers JR, Rehg JE, et al. p53-independent functions of the p19(ARF) tumor suppressor. Genes Dev 2000; 14: 2358–65.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Itahana K, Bhat KP, Jin A, et al. Tumor suppressor ARF degrades B23, a nucleolar protein involved in ribosome biogenesis and cell proliferation. Mol Cell 2003; 12: 1151–64.
    OpenUrlCrossRefPubMed
  13. ↵
    Sugimoto M, Kuo ML, Roussel MF, et al. Nucleolar Arf tumor suppressor inhibits ribosomal RNA processing. Mol Cell 2003; 11: 415–24.
    OpenUrlCrossRefPubMed
  14. ↵
    Qi Y, Gregory MA, Li Z, et al. p19ARF directly and differentially controls the functions of c-Myc independently of p53. Nature 2004; 431: 712–7.
    OpenUrlCrossRefPubMed
  15. ↵
    Datta A, Nag A, Pan W, et al. Myc-ARF (alternate reading frame) interaction inhibits the functions of Myc. J Biol Chem 2004; 279: 36698–707.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Bates S, Phillips AC, Clark PA, et al. p14ARF links the tumour suppressors RB and p53 [letter]. Nature 1998; 395: 124–5.
    OpenUrlCrossRefPubMed
  17. ↵
    Zindy F, Eischen CM, Randle DH, et al. Myc signaling via the ARF tumor suppressor regulates p53-dependent apoptosis and immortalization. Genes Dev 1998; 12: 2424–33.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Sreeramaneni R, Chaudhry A, McMahon M, et al. Ras-Raf-Arf signaling critically depends on the Dmp1 transcription factor. Mol Cell Biol 2005; 25: 220–32.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Palmero I, Pantoja C, Serrano M. p19ARF links the tumour suppressor p53 to Ras [letter]. Nature 1998; 395: 125–6.
    OpenUrlCrossRefPubMed
  20. ↵
    Llanos S, Clark PA, Rowe J, et al. Stabilization of p53 by p14ARF without relocation of MDM2 to the nucleolus. Nat Cell Biol 2001; 3: 445–52.
    OpenUrlCrossRefPubMed
  21. ↵
    Korgaonkar C, Hagen J, Tompkins V, et al. Nucleophosmin (B23) targets ARF to nucleoli and inhibits its function. Mol Cell Biol 2005; 25: 1258–71.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Bertwistle D, Sugimoto M, Sherr CJ. Physical and functional interactions of the Arf tumor suppressor protein with nucleophosmin/B23. Mol Cell Biol 2004; 24: 985–96.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Brady SN, Yu Y, Maggi LB, Jr., et al. ARF impedes NPM/B23 shuttling in an Mdm2-sensitive tumor suppressor pathway. Mol Cell Biol 2004; 24: 9327–38.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Grisendi S, Mecucci C, Falini B, et al. Nucleophosmin and cancer. Nat Rev Cancer 2006; 6: 493–505.
    OpenUrlCrossRefPubMed
  25. ↵
    den Besten W, Kuo ML, Williams RT, et al. Myeloid leukemia-associated nucleophosmin mutants perturb p53-dependent and independent activities of the Arf tumor suppressor protein. Cell Cycle 2005; 4: 1593–8.
    OpenUrlPubMed
  26. ↵
    Colombo E, Martinelli P, Zamponi R, et al. Delocalization and destabilization of the Arf tumor suppressor by the leukemia-associated NPM mutant. Cancer Res 2006; 66: 3044–50.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Colombo E, Bonetti P, Lazzerini Denchi E, et al. Nucleophosmin is required for DNA integrity and p19Arf protein stability. Mol Cell Biol 2005; 25: 8874–86.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Colombo E, Marine JC, Danovi D, et al. Nucleophosmin regulates the stability and transcriptional activity of p53. Nat Cell Biol 2002; 4: 529–33.
    OpenUrlCrossRefPubMed
  29. ↵
    Wu MH, Chang JH, Yung BY. Resistance to UV-induced cell-killing in nucleophosmin/B23 over-expressed NIH 3T3 fibroblasts: enhancement of DNA repair and up-regulation of PCNA in association with nucleophosmin/B23 over-expression. Carcinogenesis 2002; 23: 93–100.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Wu MH, Chang JH, Chou CC, et al. Involvement of nucleophosmin/B23 in the response of HeLa cells to UV irradiation. Int J Cancer 2002; 97: 297–305.
    OpenUrlCrossRefPubMed
  31. ↵
    Chan PK. Characterization and cellular localization of nucleophosmin/B23 in HeLa cells treated with selected cytotoxic agents (studies of B23-translocation mechanism). Exp Cell Res 1992; 203: 174–81.
    OpenUrlCrossRefPubMed
  32. ↵
    Shaulian E, Karin M. AP-1 as a regulator of cell life and death. Nat Cell Biol 2002; 4: 131–6.
    OpenUrlCrossRef
  33. ↵
    Shaulian E, Karin M. AP-1 in cell proliferation and survival. Oncogene 2001; 20: 2390–400.
    OpenUrlCrossRefPubMed
  34. ↵
    Davis RJ. Signal transduction by the JNK group of MAP kinases. Cell 2000; 103: 239–52.
    OpenUrlCrossRefPubMed
  35. ↵
    Yogev O, Anzi S, Inoue K, et al. Induction of transcriptionally active Jun proteins regulates drug-induced senescence. J Biol Chem 2006; 281: 34475–83.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Binetruy B, Smeal T, Karin M. Ha-Ras augments c-Jun activity and stimulates phosphorylation of its activation domain. Nature 1991; 351: 122–7.
    OpenUrlCrossRefPubMed
  37. Derijard B, Hibi M, Wu IH, et al. JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain. Cell 1994; 76: 1025–37.
    OpenUrlCrossRefPubMed
  38. ↵
    Hibi M, Lin A, Smeal T, et al. Identification of an oncoprotein- and UV-responsive protein kinase that binds and potentiates the c-Jun activation domain. Genes Dev 1993; 7: 2135–48.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Shaulian E, Schreiber M, Piu F, et al. The mammalian UV response: c-Jun induction is required for exit from p53-imposed growth arrest. Cell 2000; 103: 897–907.
    OpenUrlCrossRefPubMed
  40. ↵
    Kallunki T, Deng T, Hibi M, et al. c-Jun can recruit JNK to phosphorylate dimerization partners via specific docking interactions. Cell 1996; 87: 929–39.
    OpenUrlCrossRefPubMed
  41. ↵
    Li B, Tournier C, Davis RJ, et al. Regulation of IL-4 expression by the transcription factor JunB during T helper cell differentiation. EMBO J 1999; 18: 420–32.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Behrens A, Sibilia M, Wagner EF. Amino-terminal phosphorylation of c-Jun regulates stress-induced apoptosis and cellular proliferation. Nat Genet 1999; 21: 326–9.
    OpenUrlCrossRefPubMed
  43. ↵
    Laderoute KR, Calaoagan JM, Gustafson-Brown C, et al. The response of c-jun/AP-1 to chronic hypoxia is hypoxia-inducible factor 1 α dependent. Mol Cell Biol 2002; 22: 2515–23.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Deng T, Karin M. JunB differs from c-Jun in its DNA-binding and dimerization domains, and represses c-Jun by formation of inactive heterodimers. Genes Dev 1993; 7: 479–90.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Davis RJ. Signal transduction by the c-Jun N-terminal kinase. Biochem Soc Symp 1999; 64: 1–12.
    OpenUrlPubMed
  46. ↵
    Liu H, Deng X, Shyu YJ, et al. Mutual regulation of c-Jun and ATF2 by transcriptional activation and subcellular localization. EMBO J 2006; 25: 1058–69.
    OpenUrlCrossRefPubMed
  47. ↵
    Adler V, Dolan LR, Kim J, et al. Changes in jun N-terminal kinase activation by stress during aging of cultured normal human fibroblasts. Mol Carcinog 1996; 17: 8–12.
    OpenUrlCrossRefPubMed
  48. ↵
    Weitzman JB, Fiette L, Matsuo K, et al. JunD protects cells from p53-dependent senescence and apoptosis [In Process Citation]. Mol Cell 2000; 6: 1109–19.
    OpenUrlCrossRefPubMed
  49. ↵
    Mayer C, Bierhoff H, Grummt I. The nucleolus as a stress sensor: JNK2 inactivates the transcription factor TIF-IA and down-regulates rRNA synthesis. Genes Dev 2005; 19: 933–41.
    OpenUrlAbstract/FREE Full Text
  50. ↵
    Jaeschke A, Karasarides M, Ventura JJ, et al. JNK2 is a positive regulator of the cJun transcription factor. Mol Cell 2006; 23: 899–911.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
Cancer Research: 68 (5)
March 2008
Volume 68, Issue 5
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
Orli Yogev, Keren Saadon, Shira Anzi, Kazushi Inoue and Eitan Shaulian
Cancer Res March 1 2008 (68) (5) 1398-1406; DOI: 10.1158/0008-5472.CAN-07-2865

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
Orli Yogev, Keren Saadon, Shira Anzi, Kazushi Inoue and Eitan Shaulian
Cancer Res March 1 2008 (68) (5) 1398-1406; DOI: 10.1158/0008-5472.CAN-07-2865
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Diversity in Proadhesive Signaling Mechanisms in B-CLL Lymphocytes
  • Role of ATM Phosphorylation in Exon 11 of BRCA1
  • Methylated Genes in TGF-β Signaling in HNSCC
Show more Cell, Tumor, and Stem Cell Biology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement