Arsenic trioxide reactivates proteasome-dependent degradation of mutant p53 protein in cancer cells in part via enhanced expression of Pirh2 E3 ligase

Wensheng Yan, Yong-Sam Jung, Yanhong Zhang, Xinbin Chen, Wensheng Yan, Yong-Sam Jung, Yanhong Zhang, Xinbin Chen

Abstract

The p53 gene is mutated in more than 50% of human tumors. Mutant p53 exerts an oncogenic function and is often highly expressed in cancer cells due to evasion of proteasome-dependent degradation. Thus, reactivating proteasome-dependent degradation of mutant p53 protein is an attractive strategy for cancer management. Previously, we found that arsenic trioxide (ATO), a drug for acute promyelocytic leukemia, degrades mutant p53 protein through a proteasome pathway. However, it remains unclear what is the E3 ligase that targets mutant p53 for degradation. In current study, we sought to identify an E3 ligase necessary for ATO-mediated degradation of mutant p53. We found that ATO induces expression of Pirh2 E3 ligase at the transcriptional level. We also found that knockdown of Pirh2 inhibits, whereas ectopic expression of Pirh2 enhances, ATO-induced degradation of mutant p53 protein. Furthermore, we found that Pirh2 E3 ligase physically interacts with and targets mutant p53 for polyubiquitination and subsequently proteasomal degradation. Interestingly, we found that ATO cooperates with HSP90 or HDAC inhibitor to promote mutant p53 degradation and growth suppression in tumor cells. Together, these data suggest that ATO promotes mutant p53 degradation in part via induction of the Pirh2-dependent proteasome pathway.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Arsenic-induced degradation of mutant p53…
Figure 1. Arsenic-induced degradation of mutant p53 protein is inhibited by MG132, an inhibitor of 26S proteasome.
Western blots were prepared with extracts from HaCaT (A) and MIA PaCa-2 (B) cells, which were untreated or pretreated with 4 µM MG132 for 2 h, and then untreated or treated with ATO for 6 h.
Figure 2. Arsenic trioxide induces expression of…
Figure 2. Arsenic trioxide induces expression of Pirh2 E3 ligase.
(A) The level of Pirh2 transcript is increased by ATO. RT-PCR analysis was performed with total RNA isolated from MIA PaCa-2 and HaCaT cells untreated or treated with 7.5 µM ATO for 2–6 h. Actin mRNA was amplified as a loading control. (B) Western blots were prepared with extracts from MIA PaCa-2 and HaCaT cells untreated or treated as in (A), and then probed with antibodies against Pirh2 and actin, respectively.
Figure 3. Ectopic expression of Pirh2 promotes…
Figure 3. Ectopic expression of Pirh2 promotes arsenic-induced degradation of mutant p53 protein.
(A–B) Western blots were prepared with extracts from HaCaT (A) and MIA PaCa-2 (B) cells, which were transfected with pcDNA3 or pcDNA3-FLAG-Pirh2 for 24 h, and then treated with 5 or 7.5 µM ATO for 6 h. The blots were then probed with antibodies against FLAG tag, p53, and actin, respectively. (C) Schematic presentation of the Pirh2 protein along with locations of the Zn Finger and RING domains, two substitution mutations C145S and C148S in the RING domain (Pirh2-DN), and deletion of the RING domain in Pirh2 (Pirh2-ΔRING). (D–E) Ectopic expression of Pirh2-DN or Pirh2-ΔRING has little if any effect on the level of mutant p53. Western blots were prepared with extracts from HaCaT (D) and MIA PaCa-2 (E) cells, which were transfected with pcDNA3, pcDNA3-FLAG-Pirh2-DN, pcDNA3-FLAG-Pirh2-ΔRING, or pcDNA3-FLAG-Pirh2 for 24 h. The blots were then probed with antibodies against FLAG tagged Pirh2, p53, and actin, respectively.
Figure 4. Knockdown of Pirh2 inhibits arsenic-induced…
Figure 4. Knockdown of Pirh2 inhibits arsenic-induced degradation of mutant p53 protein.
Western blots were prepared with extracts from HaCaT (A) and MIA PaCa-2 (B) cells, which were transfected with scrambled siRNA #1 (Scr-1) (lanes 1–2), Scr-2 (lanes 5–6), siRNA against Pirh2-1 (siPirh2-1) (lanes 3–4), or siPirh2-2 (lanes 7–8), and then treated with ATO for 6 h. The blots were then probed with antibodies against Pirh2, p53, and actin, respectively.
Figure 5. Pirh2 physically associates with mutant…
Figure 5. Pirh2 physically associates with mutant p53 protein for polyubiquitination.
(A) HaCaT and MIA PaCa-2 cells were treated with 5 or 7.5 µM ATO for 6 h. Cell extracts from HaCaT (left) and MIA PaCa-2 (right) cells were immunoprecipitated with anti-p53 or control IgG. The immunocomplexes were then used to detect mutant p53 and Pirh2 along with whole-cell lysates as input control. (B) The experiment was performed as described in (A), except that anti-Pirh2 antibody was used in immunoprecipitation. (C–E) In vitro synthesized 35S-labeled wild-type p53 (C), R175H (D), and R273H (E) were mixed with GST, GST-tagged Pirh2, Pirh2-DN, or Pirh2-ΔRING. The complexes were then mixed with a buffer containing E1, E2 (UbcH5b), and Ub and then incubated at 30°C for 2 h. Ubiquitinated p53 was analyzed by SDS-PAGE and detected by autoradiography. (F) A model of Pirh2-mediated degradation of mutant p53 induced by ATO.
Figure 6. Arsenic trioxide cooperates with HSP90…
Figure 6. Arsenic trioxide cooperates with HSP90 or HDAC inhibitor to decrease mutant p53 expression and tumor cell proliferation.
(A–B) Western blots were prepared with extracts from HaCaT (A) and MIA PaCa-2 (B) cells, which were untreated or treated with 1 µM 17AAG or 2 µM SAHA for 12 h. The blots were then probed with antibodies against Pirh2 and actin, respectively. (C–D) Western blots were prepared with extracts from HaCaT (C) and MIA PaCa-2 (D) cells, which were untreated or treated with 7.5 µM ATO, 1 µM 17AAG or 2 µM SAHA, alone or in combination for 12 h. The blots were then probed with antibodies against p53 and actin, respectively. (E–F) HaCaT (E) and MIA PaCa-2 (F) cells were treated as in (C–D) for 24 h. Surviving cells from both control and treated groups were counted and presented as Mean ± SD from three separate experiments. *, p<0.05.

References

    1. Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, et al. (2007) Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat 28: 622–629.
    1. Harms K, Nozell S, Chen X (2004) The common and distinct target genes of the p53 family transcription factors. Cell Mol Life Sci 61: 822–842.
    1. Dittmer D, Pati S, Zambetti G, Chu S, Teresky AK, et al. (1993) Gain of function mutations in p53. Nat Genet 4: 42–46.
    1. Hanel W, Moll UM (2012) Links between mutant p53 and genomic instability. J Cell Biochem 113: 433–439.
    1. Brosh R, Rotter V (2009) When mutants gain new powers: news from the mutant p53 field. Nat Rev Cancer 9: 701–713.
    1. Olive KP, Tuveson DA, Ruhe ZC, Yin B, Willis NA, et al. (2004) Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell 119: 847–860.
    1. Lang GA, Iwakuma T, Suh YA, Liu G, Rao VA, et al. (2004) Gain of function of a p53 hot spot mutation in a mouse model of Li-Fraumeni syndrome. Cell 119: 861–872.
    1. Morton JP, Timpson P, Karim SA, Ridgway RA, Athineos D, et al. (2010) Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer. Proc Natl Acad Sci U S A 107: 246–251.
    1. Petitjean A, Achatz MI, Borresen-Dale AL, Hainaut P, Olivier M (2007) TP53 mutations in human cancers: functional selection and impact on cancer prognosis and outcomes. Oncogene 26: 2157–2165.
    1. Levy CB, Stumbo AC, Ano Bom AP, Portari EA, Cordeiro Y, et al. (2011) Co-localization of mutant p53 and amyloid-like protein aggregates in breast tumors. Int J Biochem Cell Biol 43: 60–64.
    1. Blandino G, Levine AJ, Oren M (1999) Mutant p53 gain of function: differential effects of different p53 mutants on resistance of cultured cells to chemotherapy. Oncogene 18: 477–485.
    1. Li R, Sutphin PD, Schwartz D, Matas D, Almog N, et al. (1998) Mutant p53 protein expression interferes with p53-independent apoptotic pathways. Oncogene 16: 3269–3277.
    1. Frazier MW, He X, Wang J, Gu Z, Cleveland JL, et al. (1998) Activation of c-myc gene expression by tumor-derived p53 mutants requires a discrete C-terminal domain. Mol Cell Biol 18: 3735–3743.
    1. Scian MJ, Stagliano KE, Anderson MA, Hassan S, Bowman M, et al. (2005) Tumor-derived p53 mutants induce NF-kappaB2 gene expression. Mol Cell Biol 25: 10097–10110.
    1. Zalcenstein A, Stambolsky P, Weisz L, Muller M, Wallach D, et al. (2003) Mutant p53 gain of function: repression of CD95(Fas/APO-1) gene expression by tumor-associated p53 mutants. Oncogene 22: 5667–5676.
    1. Yan W, Liu G, Scoumanne A, Chen X (2008) Suppression of inhibitor of differentiation 2, a target of mutant p53, is required for gain-of-function mutations. Cancer Res 68: 6789–6796.
    1. Yan W, Chen X (2009) Identification of GRO1 as a critical determinant for mutant p53 gain of function. J Biol Chem 284: 12178–12187.
    1. Yeudall WA, Vaughan CA, Miyazaki H, Ramamoorthy M, Choi MY, et al. (2012) Gain-of-function mutant p53 upregulates CXC chemokines and enhances cell migration. Carcinogenesis 33: 442–451.
    1. Li Y, Prives C (2007) Are interactions with p63 and p73 involved in mutant p53 gain of oncogenic function? Oncogene 26: 2220–2225.
    1. Di Como CJ, Gaiddon C, Prives C (1999) p73 function is inhibited by tumor-derived p53 mutants in mammalian cells. Mol Cell Biol 19: 1438–1449.
    1. Gaiddon C, Lokshin M, Ahn J, Zhang T, Prives C (2001) A subset of tumor-derived mutant forms of p53 down-regulate p63 and p73 through a direct interaction with the p53 core domain. Mol Cell Biol 21: 1874–1887.
    1. Strano S, Fontemaggi G, Costanzo A, Rizzo MG, Monti O, et al. (2002) Physical interaction with human tumor-derived p53 mutants inhibits p63 activities. J Biol Chem 277: 18817–18826.
    1. Strano S, Munarriz E, Rossi M, Cristofanelli B, Shaul Y, et al. (2000) Physical and functional interaction between p53 mutants and different isoforms of p73. J Biol Chem 275: 29503–29512.
    1. Whitesell L, Sutphin PD, Pulcini EJ, Martinez JD, Cook PH (1998) The physical association of multiple molecular chaperone proteins with mutant p53 is altered by geldanamycin, an hsp90-binding agent. Mol Cell Biol 18: 1517–1524.
    1. Wiech M, Olszewski MB, Tracz-Gaszewska Z, Wawrzynow B, Zylicz M, et al. (2012) Molecular mechanism of mutant p53 stabilization: the role of HSP70 and MDM2. PLoS One 7: e51426.
    1. Li D, Marchenko ND, Schulz R, Fischer V, Velasco-Hernandez T, et al. (2011) Functional inactivation of endogenous MDM2 and CHIP by HSP90 causes aberrant stabilization of mutant p53 in human cancer cells. Mol Cancer Res 9: 577–588.
    1. Ano Bom AP, Rangel LP, Costa DC, de Oliveira GA, Sanches D, et al. (2012) Mutant p53 aggregates into prion-like amyloid oligomers and fibrils: implications for cancer. J Biol Chem 287: 28152–28162.
    1. Terzian T, Suh YA, Iwakuma T, Post SM, Neumann M, et al. (2008) The inherent instability of mutant p53 is alleviated by Mdm2 or p16INK4a loss. Genes Dev 22: 1337–1344.
    1. Vakifahmetoglu-Norberg H, Kim M, Xia HG, Iwanicki MP, Ofengeim D, et al. (2013) Chaperone-mediated autophagy degrades mutant p53. Genes Dev 27: 1718–1730.
    1. Li D, Marchenko ND, Moll UM (2011) SAHA shows preferential cytotoxicity in mutant p53 cancer cells by destabilizing mutant p53 through inhibition of the HDAC6-Hsp90 chaperone axis. Cell Death Differ 18: 1904–1913.
    1. Rangel LP, Costa DC, Vieira TC, Silva JL (2014) The aggregation of mutant p53 produces prion-like properties in cancer. Prion 8 In press.
    1. Yan W, Zhang Y, Zhang J, Liu S, Cho SJ, et al. (2011) Mutant p53 protein is targeted by arsenic for degradation and plays a role in arsenic-mediated growth suppression. J Biol Chem 286: 17478–17486.
    1. Emadi A, Gore SD (2010) Arsenic trioxide - An old drug rediscovered. Blood Rev 24: 191–199.
    1. Yan W, Chen X, Zhang Y, Zhang J, Jung YS (2013) Arsenic suppresses cell survival via Pirh2-mediated proteasomal degradation of DeltaNp63 protein. J Biol Chem 288: 2907–2913.
    1. Yan W, Liu S, Xu E, Zhang J, Zhang Y, et al. (2013) Histone deacetylase inhibitors suppress mutant p53 transcription via histone deacetylase 8. Oncogene 32: 599–609.
    1. Jung YS, Liu G, Chen X (2010) Pirh2 E3 ubiquitin ligase targets DNA polymerase eta for 20S proteasomal degradation. Mol Cell Biol 30: 1041–1048.
    1. Jung YS, Qian Y, Yan W, Chen X (2013) Pirh2 E3 ubiquitin ligase modulates keratinocyte differentiation through p63. J Invest Dermatol 133: 1178–1187.
    1. Zheng T, Wang J, Zhao Y, Zhang C, Lin M, et al. (2013) Spliced MDM2 isoforms promote mutant p53 accumulation and gain-of-function in tumorigenesis. Nat Commun 4: 2996.
    1. Karsy M, Albert L, Murali R, Jhanwar-Uniyal M (2014) The impact of arsenic trioxide and all-trans retinoic acid on p53 R273H-codon mutant glioblastoma. Tumour Biol 35: 4567–4580.
    1. Lopes UG, Erhardt P, Yao R, Cooper GM (1997) p53-dependent induction of apoptosis by proteasome inhibitors. J Biol Chem 272: 12893–12896.
    1. Choudhury S, Kolukula VK, Preet A, Albanese C, Avantaggiati ML (2013) Dissecting the pathways that destabilize mutant p53: the proteasome or autophagy? Cell Cycle 12: 1022–1029.
    1. Zheng G, Ning J, Yang YC (2007) PLAGL2 controls the stability of Pirh2, an E3 ubiquitin ligase for p53. Biochem Biophys Res Commun 364: 344–350.
    1. Lee JT, Gu W (2010) The multiple levels of regulation by p53 ubiquitination. Cell Death Differ 17: 86–92.
    1. Shloush J, Vlassov JE, Engson I, Duan S, Saridakis V, et al. (2011) Structural and functional comparison of the RING domains of two p53 E3 ligases, Mdm2 and Pirh2. J Biol Chem 286: 4796–4808.
    1. Peng Y, Chen L, Li C, Lu W, Chen J (2001) Inhibition of MDM2 by hsp90 contributes to mutant p53 stabilization. J Biol Chem 276: 40583–40590.
    1. Yan W, Liu S, Xu E, Zhang J, Zhang Y, et al. (2013) Histone deacetylase inhibitors suppress mutant p53 transcription via histone deacetylase 8. Oncogene 32: 599–609.
    1. Zhu HB, Yang K, Xie YQ, Lin YW, Mao QQ, et al. (2013) Silencing of mutant p53 by siRNA induces cell cycle arrest and apoptosis in human bladder cancer cells. World J Surg Oncol 11: 22.
    1. Suh YA, Post SM, Elizondo-Fraire AC, Maccio DR, Jackson JG, et al. (2011) Multiple stress signals activate mutant p53 in vivo. Cancer Res 71: 7168–7175.
    1. Shen SC, Lee WR, Yang LY, Tsai HH, Yang LL, et al. (2012) Quercetin enhancement of arsenic-induced apoptosis via stimulating ROS-dependent p53 protein ubiquitination in human HaCaT keratinocytes. Exp Dermatol 21: 370–375.
    1. Tatham MH, Geoffroy MC, Shen L, Plechanovova A, Hattersley N, et al. (2008) RNF4 is a poly-SUMO-specific E3 ubiquitin ligase required for arsenic-induced PML degradation. Nat Cell Biol 10: 538–546.
    1. Geoffroy MC, Jaffray EG, Walker KJ, Hay RT (2010) Arsenic-induced SUMO-dependent recruitment of RNF4 into PML nuclear bodies. Mol Biol Cell 21: 4227–4239.
    1. Lallemand-Breitenbach V, Jeanne M, Benhenda S, Nasr R, Lei M, et al. (2008) Arsenic degrades PML or PML-RARalpha through a SUMO-triggered RNF4/ubiquitin-mediated pathway. Nat Cell Biol 10: 547–555.

Source: PubMed

3
Sottoscrivi