Role of wild-type p53-induced phosphatase 1 in cancer (Review)

  • Authors:
    • Zhi‑Peng Wang
    • Ye Tian
    • Jun Lin
  • View Affiliations

  • Published online on: July 27, 2017     https://doi.org/10.3892/ol.2017.6685
  • Pages: 3893-3898
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Wild-type p53-induced phosphatase (Wip1) is a member of the protein phosphatase type 2C family and is an established oncogene due to its dephosphorylation of several tumor suppressors and negative control of the DNA damage response system. It has been reported to dephosphorylate p53, ataxia telangiectasia mutated, checkpoint kinase 1 and p38 mitogen activated protein kinases, forming negative feedback loops to inhibit apoptosis and cell cycle arrest. Wip1 serves a major role in tumorigenesis, progression, invasion, distant metastasis and apoptosis in various types of human cancer. Therefore, it may be a potential biomarker and therapeutic target in the diagnosis and treatment of cancer. Furthermore, previous evidence has revealed a new role for Wip1 in the regulation of chemotherapy resistance. In the present review, the current knowledge on the role of Wip1 in cancer is discussed, as well as its potential as a novel target for cancer treatment and its function in chemotherapy resistance.

Introduction

Genetic stability ensures the inheritance of the correct genetic information and preserves the function of normal physiological processes. However, cells living in a constantly changing environment are influenced by various stresses, which may alter DNA sequences and induce DNA damage (1). During the evolution of genes, cells have developed a DNA damage response system including cell cycle checkpoints, senescence and apoptosis (2). If that system is not able to repair DNA damage, DNA replication, transcription and recombination may be altered, leading to gene mutation and chromosomal rearrangements or loss, which promotes the development of cancer (3). Wild-type p53-induced phosphatase 1 (Wipl) is an oncogene that negatively regulates the DNA damage response system and serves a role in tumorigenesis, therapy and prognosis in various types of human cancer (4).

Wip1 was originally identified as a target protein in the p53-dependent response to ionizing radiation (5). Wip1 is a serine/threonine protein phosphatase that is encoded by the protein phosphatase magnesium-dependent 1 δ (gene, PPM1D) in the 17q22/q24 human chromosomal region, and is a member of the protein phosphatase type 2C (PP2C) family (6). It is 605 amino acids long and consists of a central phosphatase catalytic domain and a non-functional region (7). Wip1 is a monomeric enzyme, similar to other members of the PP2C family, the dephosphorylation of which requires catalysis by bivalent cations, including magnesium and manganese ions (5).

Previous studies have revealed that Wip1 dephosphorylates several key DNA damage response proteins, including p53, ataxia telangiectasia mutated, checkpoint kinase (Chk) 1, Chk2, murine double minute 2 (Mdm2) and p38 mitogen activated protein kinases (p38 MAPK), exercising negative feedback loops that lead to cell cycle arrest, increased tumorigenesis and the inhibition of apoptosis (Fig. 1) (812). Among these loops, negative regulation of p53 is vital. TP53 may be the most important tumor suppressor gene, the mutation or depletion of which is present in ~50% of all human tumors (13). However, Wip1 is not only able to directly dephosphorylate p53 protein at serine 15, but also indirectly inactivate p53 protein through p38 MAPK and Mdm2 (8,14,15), which attenuates the p53 function. Furthermore, dephosphorylation of p53 by Wip1 induces inappropriate re-initiation of mitosis and uncontrolled polyploid progression that may be a potential underlying mechanism of tumor progression (14). Previous studies have identified additional Wip1 targets, including murine double minute X, xeroderma pigmentosum complementation group A and C, nuclear factor kappa B (NF-κB) and DNA methylation, resulting in the promotion of proliferation, inhibition of inflammation and nucleotide excision repair (9,1619). On the other hand, cytotoxic drugs, including cisplatin and doxorubicin, are able to induce senescence and apoptosis in tumor cells, an effect that is dependent on p53 signaling pathway in vitro and in vivo (20,21). This indicates that Wip1 phosphatase activity may mediate the cytotoxicity of chemotherapeutic agents via targeting p53. The present review summarizes the regulatory mechanisms and functions of Wip1 as an oncogene in various types of cancer. In addition, the potential role of Wip1 as a tumor biomarker and therapeutic target in these cancer types was investigated.

Wip1 in breast cancer

The role of Wip1 in breast cancer is the most studied compared with all other types of human cancer (22). In 28% of primary breast cancer cases, the amplification of the 17q22/q24 chromosomal region has been demonstrated through cytogenetic analysis, a phenomenon that is more common in high-grade breast cancer (23). In addition, a number of studies have identified that the overexpression of Wip1 negatively regulates the p53, p38 MAPK and p16 signaling pathways, which may lead to breast cancer tumorigenesis, proliferation and poor prognosis (24,25). Previous reports have demonstrated that the upregulation of Wip1 may reverse the induction of apoptosis by microRNA (miRNA/miR)-16 and miRNA-34a, which are tumor suppressors of breast cancer (26,27). Therefore, high Wip1 expression levels may be a predisposing factor for breast cancer. Spike and Wahl (28) revealed that Wip1 regulated chemosensitivity by controlling the p53 signaling pathway. Downregulation of Wip1 enhanced the chemosensitivity of breast cancer to adriamycin via targeting wild-type p53 and reducing cell growth and cell survival; however, these effects were not present in cell lines with mutant-type p53 (Table I; Fig. 1) (29,30). Although the occurrence of breast cancer is regulated by various oncogenes, including ErbB2, Wnt1 and breast cancer susceptibility protein type 1 and 2 (22), these results suggested that Wip1 may be considered as a potential biomarker for tumorigenesis and index of prognosis in patients with breast cancer. In addition, decreasing its expression levels may have a therapeutic effect during the chemotherapy of breast cancer with wild-type p53.

Table I.

Roles of Wip1 in various types of cancer.

Table I.

Roles of Wip1 in various types of cancer.

Cancer type Overexpression/mutation level, %TargetsTumorigenesis Differentiation ChemoresistancePrognosis
Breast28p53, p16, p38 MAPK++++
Glioma37.5Chk2, p53+NDNDND
Neuroblastoma56Mdm2, p53++++
Medulloblastoma64Mdm2, p53, CXCR4, AKT++ND+
Ovarian clear cell carcinoma40Chk1, p53+ND++
Liver65P53++ND+
BladderNDP53+ND+ND
Kidney67ND++ND+
Nasopharyngeal carcinoma69ND++ND+

[i] Wip1, wild-type p53-induced phosphatase 1; P38 MAPK, p38 mitogen activated protein kinases; Chk1, checkpoint kinase 1; Chk2, checkpoint kinase 2; Mdm2, murine double minute 2; CXCR4, C-X-C chemokine receptor type 4; AKT, protein kinase B; +, positive correlation; ND, no data available.

Wip1 in childhood glioma

Overexpression of Wip1 and gain-of-function mutations of PPM1D have been detected in numerous types of pediatric cancer, including glioma (31), neuroblastoma (32) and medulloblastoma (33).

Wip1 in glioma

Zhang et al (34) identified that carboxy terminal truncating mutations of PPM1D occur in 37.5% of glioma cases, and these gain-of-function PPM1D mutants suppressed phosphorylation of Chk2 at threonine 68 and p53 at serine 15, resulting in dysfunction of the DNA damage response network (Table I) (34). This result may be associated with predisposition to and the tumorigenesis of glioma.

Wip1 in neuroblastoma

Overexpression of Wip1 in neuroblastoma may repress p53 function by two signaling pathways, one is the Wip1-p53 pathway, and the other is the Wip1-Mdm2-p53 pathway (35), resulting in tumorigenesis. In addition, a previous report demonstrated that Wip1 was significantly overexpressed in 56% of cancer tissues, and promoted tumor progression to a higher stage, poor prognosis and chemotherapy resistance (Table I) (32). Therefore, these data suggest that the inhibition of Wip1 expression levels may be a potential therapeutic target. GSK2830371 is a Wip1 selective antagonist able to significantly inhibit 96.5% of Wip1 activity in neuroblastoma cell lines, which promotes p53 function and apoptotic responses (32). Furthermore, GSK2830371 had a synergistic effect on the antiproliferative properties of the chemotherapeutic agents adriamycin and carboplatin (32).

Wip1 in medulloblastoma

Previous studies have reported that the amplification and overexpression of Wip1 occurred in 64% of human medulloblastomas, and it is more common in highly aggressive medulloblastomas (36,37). Buss et al (37) also identified that high levels of Wip1 expression were associated with increased expression of Mdm2, which may be an underlying mechanism of promoting medulloblastoma growth via targeting p53 (37). In addition, Pfister et al (38) demonstrated that the upregulation of Wip1 expression was associated with poor prognosis in medulloblastoma (Table I) (38). Furthermore, the results of a previous study have revealed that Wip1 is able to promote the progression and invasion of aggressive medulloblastoma by regulating C-X-C chemokine receptor type 4 and protein kinase B (Akt; Fig. 1) (33). These data suggest that Wip1 serves an important role in tumorigenesis and cancer progression and may be an indicator for the prognosis of medulloblastoma.

In pediatric types of cancer, overexpression of Wip1 contributes to a number of malignant characteristics, including tumor progression, aggressive phenotype and poor prognosis (32,34,37). In addition, a Wip1 inhibitor may be a promising novel candidate for targeted therapeutic strategies for these severe tumors.

Wip1 in ovarian clear cell carcinoma

A previous study identified that the amplification and overexpression of the PPM1D gene occurred in ≥40% of cases of ovarian clear cell carcinoma, which is higher compared with other ovarian tumor subtypes (39). A gene knockdown study revealed the viability of ovarian clear cell carcinoma cell lines depended on the phosphatase activity of Wip1, indicating that the Wip1 and PPM1D genes may be drivers of ovarian clear cell carcinoma (40). Another previous study reported that Wip1 is able to negatively regulate the Chk1 and p53 signaling pathway, resulting in tumorigenesis (41). However, cisplatin mediates tumor cell DNA damage and apoptotic function through these signaling pathways, suggesting that Wip1 may be responsible for the cisplatin resistance of ovarian clear cell carcinoma (41). In addition, a recent study demonstrated that Akt confers cisplatin resistance in part through the regulation of PPM1D protein stability, preventing its proteasomal degradation and consequently increasing its half-life (Table I) (42). Accumulating evidence has indicated that Wip1 is directly associated with tumor cell survival and chemoresistance (43). Due to its late diagnosis and the development of chemoresistance, ovarian clear cell carcinoma is characterized by the poorest prognosis among ovarian types of cancer (44). Therefore, Wip1 expression levels may be a biomarker of diagnosis and index of prognosis. In addition, targeting Wip1 may improve therapeutic outcomes in ovarian clear cell carcinoma.

Wip1 in liver cancer

miR-29c belongs to the miR-29 family, which are established tumor suppressors (45). A previous study demonstrated that miR-29c is downregulated in liver cancer and may affect the apoptosis, tumorigenesis, and prognosis of tumor cells (46). Previous studies have revealed that the overexpression of miR-29c inhibits cancer cell proliferation and metastasis, as well as inducing cell cycle arrest (47,48). Wip1 was also revealed to be significantly upregulated in hepatocellular carcinoma and may contribute to the development of this cancer (49). Wang et al (49) were the first to investigate the association between Wip1 and miR-29c, revealing an inverse correlation. In addition, the overexpression of Wip1 may suppress miR-29c-induced apoptosis and cell cycle arrest via dephosphorylating wild-type p53 (49). Although mutations of p53 occur in ~50% of human cancer cases, this rate is <30% for liver cancer cases, the majority of which express wild-type p53 (50). These findings suggest that Wip1 and miR-29c serve roles in the development of hepatocellular carcinoma. Furthermore, a previous study demonstrated that Wip1 was overexpressed in hepatocellular carcinoma tissues, compared with in non-cancerous tissues, and high Wip1 expression levels were associated with a more advanced tumor-node-metastasis stage, as well as being a significantly independent prognostic factor (51). Therefore, Wip1 not only participates in tumorigenesis but also indicates poor prognosis in liver cancer.

Wip1 in bladder cancer

Amplification and overexpression of Wip1 were also identified in bladder cancer (52). Wang et al (53) demonstrated that RNA interference of PPM1D inhibited bladder cancer cell proliferation and tumorigenesis in mice, potentially through targeting of the p53, p38 MAPK and Akt signaling pathways. This indicates that targeting PPM1D may be a potential therapeutic strategy for the treatment of bladder cancer. Furthermore, Lin et al (54) revealed that the level of Wip1 expression in cisplatin-resistant bladder cancer was high compared with the control tumor tissue (54). In addition, loss of homeodomain-interacting protein kinase-2 enhanced Wip1 expression, which subsequently increased tumor cell viability in cell lines with wild-type p53 during cisplatin treatment (54). These results suggest that Wip1 upregulation decreases the tumor response to cisplatin, resulting in tumor cell-survival and resistance to apoptosis that is induced by chemotherapeutic drugs. Therefore, Wip1 may lead to chemotherapy resistance in bladder cancer. Conversely, previous studies have revealed that chemotherapy resistance induced by Wip1 is dependent on the presence of wild-type p53; however, in p53-negative cell lines, Wip1 sensitizes tumor cells to chemotherapeutic drugs by regulating the B-cell lymphoma-2 associated X protein:B-cell lymphoma-extra large ratio and runt related transcription factor-2 and protects normal tissues (Fig. 1) (55,56). Therefore, Wip1 inhibition is a potential therapeutic target in bladder cancer with preserved wild-type p53, but the reverse effect may occur in p53-negative tumors; however, this remains to be elucidated.

Wip1 in kidney cancer

Two previous studies demonstrated that Wip1 is amplified and overexpressed in kidney cancer, the pathological types of which included clear cell type, granule cell type and papillary cell type (57,58). These reports revealed that Wip1 expression levels are correlated with the clinical characteristics of kidney cancer, including lymph node metastasis, distant metastasis, Fuhrman grade, clinical stage and pathological differentiation (57,58). In addition, these studies also identified that patients with high levels of Wip1 expression had significantly lower survival rates than those with low levels of Wip1 expression, and the downregulation of Wip1 promoted apoptosis and decreased migration and invasion in kidney cancer cell lines (Table I) (57,58). These results indicate that Wip1 may serve an important role in the tumorigenesis and the progression of kidney cancer.

Wip1 in nasopharyngeal carcinoma

Sun et al (59) also observed the tumorigenic action of Wip1 in nasopharyngeal carcinoma, leading to a more aggressive grade, distant metastasis and a poorer prognosis (Table I). Therefore, all the aforementioned evidence suggests that Wip1 overexpression promotes tumorigenesis in a number of solid tumors and indicates that Wip1 is a potential molecular target for tumor therapy.

Conclusion

Wip1 has received increasing attention since it was first identified in 1997 (5). A number of studies have demonstrated that Wip1 negatively regulates various signaling pathways and feedback loops, particularly p53-induced mechanisms, resulting in tumorigenesis of multiple tissues and organs. In addition, Wip1 expression serves a critical role in the progression, migration, invasion and apoptosis of cancer. As an oncogene, its expression levels indicate a poor prognosis of disease. Therefore, Wip1 may act as a potential tumor biomarker, therapeutic target and index of prognosis in various types of cancer.

In conclusion, a number of studies have demonstrated that Wip1 is an attractive chemotherapeutic target. Its overexpression and amplification increases chemotherapy resistance in tumors with wild-type p53, but the reverse of this effect is observed in p53-negative tumor cells. However, the underlying mechanisms by which Wip1 affects chemotherapy remain to be investigated.

Acknowledgements

The present study was supported by the Capital Health Research and Development of Special Grants (grant no. 2011-2002-05).

References

1 

Jackson SP and Bartek J: The DNA-damage response in human biology and disease. Nature. 461:1071–1078. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Harper JW and Elledge SJ: The DNA damage response: Ten years after. Mol Cell. 28:739–745. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Lord CJ and Ashworth A: The DNA damage response and cancer therapy. Nature. 481:287–294. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Le Guezennec X and Bulavin DV: WIP1 phosphatase at the crossroads of cancer and aging. Trends Biochem Sci. 35:109–114. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Fiscella M, Zhang H, Fan S, Sakaguchi K, Shen S, Mercer WE, Vande Woude GF, O'Connor PM and Appella E: Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad Sci USA. 94:6048–6053. 1997. View Article : Google Scholar : PubMed/NCBI

6 

Bulavin DV, Demidov ON, Saito S, Kauraniemi P, Phillips C, Amundson SA, Ambrosino C, Sauter G, Nebreda AR, Anderson CW, et al: Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity. Nat Genet. 31:210–215. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Nakagawa H, Wardell CP, Furuta M, Taniguchi H and Fujimoto A: Cancer whole-genome sequencing: Present and future. Oncogene. 34:5943–5950. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Lu X, Nguyen TA, Moon SH, Darlington Y, Sommer M and Donehower LA: The type 2C phosphatase Wip1: An oncogenic regulator of tumor suppressor and DNA damage response pathways. Cancer Metastasis Rev. 27:123–135. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Lowe J, Cha H, Lee MO, Mazur SJ, Appella E and Fornace AJ Jr: Regulation of the Wip1 phosphatase and its effects on the stress response. Front Biosci (Landmark Ed). 17:1480–1498. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Shreeram S, Demidov ON, Hee WK, Yamaguchi H, Onishi N, Kek C, Timofeev ON, Dudgeon C, Fornace AJ, Anderson CW, et al: Wip1 phosphatase modulates ATM-dependent signaling pathways. Mol Cell. 23:757–764. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Lu X, Nannenga B and Donehower LA: PPM1D dephosphorylates Chk1 and p53 and abrogates cell cycle checkpoints. Genes Dev. 19:1162–1174. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Fujimoto H, Onishi N, Kato N, Takekawa M, Xu XZ, Kosugi A, Kondo T, Imamura M, Oishi I, Yoda A and Minami Y: Regulation of the antioncogenic Chk2 kinase by the oncogenic Wip1 phosphatase. Cell Death Differ. 13:1170–1180. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Hollstein M, Sidransky D, Vogelstein B and Harris CC: p53 mutations in human cancers. Science. 253:49–53. 1991. View Article : Google Scholar : PubMed/NCBI

14 

Crescenzi E, Raia Z, Pacifico F, Mellone S, Moscato F, Palumbo G and Leonardi A: Down-regulation of wild-type p53-induced phosphotase 1 (Wip1) plays a critical role in regulating several p53-dependent functions in premature senescent tumor cells. J Biol Chem. 288:16212–16224. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Lu X, Ma O, Nguyen TA, Jones SN, Oren M and Donehower LA: The Wip1 phosphatase acts as a gatekeeper in the p53-Mdm2 autoregulatory loop. Cancer Cell. 12:342–354. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Zhang X, Lin L, Guo H, Yang J, Jones SN, Jochemsen A and Lu X: Phosphorylation and degradation of MdmX is inhibited by Wip1 phosphatase in the DNA damage response. Cancer Res. 69:7960–7968. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Nguyen TA, Slattery SD, Moon SH, Darlington YF, Lu X and Donehower LA: The oncogenic phosphatase Wip1 negatively regulates nucleotide excision repair. DNA Repair (Amst). 9:813–823. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Chew J, Biswas S, Shreeram S, Humaidi M, Wong ET, Dhillion MK, Teo H, Hazra A, Fang CC, López-Collazo E, et al: Wip1 phosphatase is a negative regulator of NF-kappaB signaling. Nat Cell Biol. 11:659–666. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Filipponi D, Muller J, Emelyanov A and Bulavin DV: Wip1 controls global heterochromatin silencing via ATM/BRCA1-dependent DNA methylation. Cancer Cell. 24:528–541. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Chang BD, Broude EV, Dokmanovic M, Zhu H, Ruth A, Xuan Y, Kandel ES, Lausch E, Christov K and Roninson IB: A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res. 59:3761–3767. 1999.PubMed/NCBI

21 

te Poele RH, Okorokov AL, Jardine L, Cummings J and Joel SP: DNA damage is able to induce senescence in tumor cells in vitro and in vivo. Cancer Res. 62:1876–1883. 2002.PubMed/NCBI

22 

Emelyanov A and Bulavin DV: Wip1 phosphatase in breast cancer. Oncogene. 34:4429–4438. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Rennstam K, Ahlstedt-Soini M, Baldetorp B, Bendahl PO, Borg A, Karhu R, Tanner M, Tirkkonen M and Isola J: Patterns of chromosomal imbalances defines subgroups of breast cancer with distinct clinical features and prognosis. A study of 305 tumors by comparative genomic hybridization. Cancer Res. 63:8861–8868. 2003.PubMed/NCBI

24 

Demidov ON, Kek C, Shreeram S, Timofeev O, Fornace AJ, Appella E and Bulavin DV: The role of the MKK6/p38 MAPK pathway in Wip1-dependent regulation of ErbB2-driven mammary gland tumorigenesis. Oncogene. 26:2502–2506. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Yu E, Ahn YS, Jang SJ, Kim MJ, Yoon HS, Gong G and Choi J: Overexpression of the Wip1 gene abrogates the p38 MAPK/p53/Wip1 pathway and silences p16 expression in human breast cancers. Breast Cancer Res Treat. 101:269–278. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Zhang X, Wan G, Mlotshwa S, Vance V, Berger FG, Chen H and Lu X: Oncogenic Wip1 phosphatase is inhibited by miR-16 in the DNA damage signaling pathway. Cancer Res. 70:7176–7186. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Wang B, Li D and Kovalchuk O: p53 Ser15 phosphorylation and histone modifications contribute to IR-induced miR-34a transcription in mammary epithelial cells. Cell Cycle. 12:2073–2083. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Spike BT and Wahl GM: P53, stem cells, and reprogramming: Tumor suppression beyond guarding the genome. Genes Cancer. 2:404–419. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Kong W, Jiang X and Mercer WE: Downregulation of Wip-1 phosphatase expression in MCF-7 breast cancer cells enhances doxorubicin-induced apoptosis through p53-mediated transcriptional activation of Bax. Cancer Biol Ther. 8:555–563. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Pärssinen J, Alarmo EL, Karhu R and Kallioniemi A: PPM1D silencing by RNA interference inhibits proliferation and induces apoptosis in breast cancer cell lines with wild-type p53. Cancer Genet Cytogenet. 182:33–39. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Liang CH, Jiao BH, Lu SK, Guo EK and Zhang GY: The expression of proto-oncogene Wip1 in human glioblastoma multiforme and cell lines. Chin J Neuro Oncol. 9:1–6. 2011.

32 

Richter M, Dayaram T, Gilmartin AG, Ganji G, Pemmasani SK, Van Der Key H, Shohet JM, Donehower LA and Kumar R: Wip1 phosphatase as a potential therapeutic target in neuroblastoma. PLoS One. 10:e01156352015. View Article : Google Scholar : PubMed/NCBI

33 

Buss MC, Remke M, Lee J, Gandhi K, Schniederjan MJ, Kool M, Northcott PA, Pfister SM, Taylor MD and Castellino RC: The Wip1 oncogene promotes progression and invasion of aggressive medulloblastoma variants. Oncogene. 34:1126–1140. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Zhang L, Chen LH, Wan H, Yang R, Wang Z, Feng J, Yang S, Jones S, Wang S, Zhou W, et al: Exome sequencing identifies somatic gain-of-function PPM1D mutations in brainstem gliomas. Nat Genet. 46:726–730. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Barone G, Tweddle A, Shohet JM, Chesler L, Moreno L, Pearson AD and Van Maerken T: MDM2-p53 interaction in paediatric solid tumours: Preclinical rationale, biomarkers and resistance. Curr Drug Targets. 15:114–123. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Northcott PA, Shih DJ, Peacock J, Garzia L, Morrissy AS, Zichner T, Stütz AM, Korshunov A, Reimand J, Schumacher SE, et al: Subgroup-specific structural variation across 1,000 medulloblastoma genomes. Nature. 488:49–56. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Buss MC, Read TA, Schniederjan MJ, Gandhi K and Castellino RC: HDM2 promotes Wip1-mediated medulloblastoma growth. Neuro Oncol. 14:440–458. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Pfister S, Remke M, Benner A, Mendrzyk F, Toedt G, Felsberg J, Wittmann A, Devens F, Gerber NU, Joos S, et al: Outcome prediction in pediatric medulloblastoma based on DNA copy-number aberrations of chromosomes 6q and 17q and the MYC and MYCN loci. J Clin Oncol. 27:1627–1636. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Hirasawa A, Saito-Ohara F, Inoue J, Aoki D, Susumu N, Yokoyama T, Nozawa S, Inazawa J and Imoto I: Association of 17q21-q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM1D and APPBP2 as likely amplification targets. Clin Cancer Res. 9:1995–2004. 2003.PubMed/NCBI

40 

Tan DS, Lambros MB, Rayter S, Natrajan R, Vatcheva R, Gao Q, Marchiò C, Geyer FC, Savage K, Parry S, et al: PPM1D is a potential therapeutic target in ovarian clear cell carcinomas. Clin Cancer Res. 15:2269–2280. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Ali AY, Abedini MR and Tsang BK: The oncogenic phosphatase PPM1D confers cisplatin resistance in ovarian carcinoma cells by attenuating checkpoint kinase 1 and p53 activation. Oncogene. 31:2175–2186. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Ali AY, Kim JY, Pelletier JF, Vanderhyden BC, Bachvarov DR and Tsang BK: Akt confers cisplatin chemoresistance in human gynecological carcinoma cells by modulating PPM1D stability. Mol Carcinog. 54:1301–1314. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Ali AY, Farrand L, Kim JY, Byun S, Suh JY, Lee HJ and Tsang BK: Molecular determinants of ovarian cancer chemoresistance: New insights into an old conundrum. Ann NY Acad Sci. 1271:58–67. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Tan DS and Kaye S: Ovarian clear cell adenocarcinoma: A continuing enigma. J Clin Pathol. 60:355–360. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Bae HJ, Noh JH, Kim JK, Eun JW, Jung KH, Kim MG, Chang YG, Shen Q, Kim SJ, Park WS, et al: MicroRNA-29c functions as a tumor suppressor by direct targeting oncogenic SIRT1 in hepatocellular carcinoma. Oncogene. 33:2557–2567. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Xiong Y, Fang JH, Yun JP, Yang J, Zhang Y, Jia WH and Zhuang SM: Effects of microRNA-29 on apoptosis, tumorigenecity, and prognosis of hepatocellular carcinoma. Hepatology. 51:836–845. 2010.PubMed/NCBI

47 

Wang H, Zhu Y, Zhao M, Wu C, Zhang P, Tang L, Zhang H, Chen X, Yang Y and Liu G: miRNA-29c suppresses lung cancer cell adhesion to extracellular matrix and metastasis by targeting integrin β1 and matrix metalloproteinase 2 (MMP2). PLoS One. 8:e701922013. View Article : Google Scholar : PubMed/NCBI

48 

Ding DP, Chen ZL, Zhao XH, Wang JW, Sun J, Wang Z, Tan FW, Tan XG, Li BZ, Zhou F, et al: miR-29c induces cell cycle arrest in esophageal squamous cell carcinoma by modulating cyclin E expression. Carcinogenesis. 32:1025–1032. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Wang B, Li D, Sidler C, Rodriguez-Juarez R, Singh N, Heyns M, Ilnytskyy Y, Bronson RT and Kovalchuk O: A suppressive role of ionizing radiation-responsive miR-29c in the development of liver carcinoma via targeting WIP1. Oncotarget. 6:9937–9950. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Oda T, Tsuda H, Scarpa A, Sakamoto M and Hirohashi S: p53 gene mutation spectrum in hepatocellular carcinoma. Cancer Res. 52:6358–6364. 1992.PubMed/NCBI

51 

Li GB, Zhang XL, Yuan L, Jiao QQ, Liu DJ and Liu J: Protein phosphatase magnesium-dependent 1 δ (PPM1D) mRNA expression is a prognosis marker for hepatocellular carcinoma. PLoS One. 8:e607752013. View Article : Google Scholar : PubMed/NCBI

52 

Koo SH, Kwon KC, Ihm CH, Jeon YM, Park JW and Sul CK: Detection of genetic alterations in bladder tumors by comparative genomic hybridization and cytogenetic analysis. Cancer Genet Cytogenet. 110:87–93. 1999. View Article : Google Scholar : PubMed/NCBI

53 

Wang W, Zhu H, Zhang H, Zhang L, Ding Q and Jiang H: Targeting PPM1D by lentivirus-mediated RNA interference inhibits the tumorigenicity of bladder cancer cells. Braz J Med Bio Res. 47:1044–1049. 2014. View Article : Google Scholar

54 

Lin J, Zhang Q, Lu Y, Xue W, Xu Y and Hu X: Downregulation of HIPK2 increases resistance of bladder cancer cell to cisplatin by regulating wip1. PLoS One. 9:e984182014. View Article : Google Scholar : PubMed/NCBI

55 

Goloudina AR, Mazur SJ, Appella E, Garrido C and Demidov ON: Wip1 sensitizes p53-negative tumors to apoptosis by regulating the Bax/Bcl-xl ratio. Cell Cycle. 11:1883–1887. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Goloudina AR, Tanoue K, Hammann A, Fourmaux E, Le Guezennec X, Bulavin DV, Mazur SJ, Appella E, Garrido C and Demidov ON: Wip1 promotes RUNX2-dependent apoptosis in p53-negative tumors and protects normal tissues during treatment with anticancer agents. Proc Natl Acad Sci USA. 109:E68–E75. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Sun GG, Wang YD, Liu Q and Hu WN: Expression of Wip1 in kidney carcinoma and its correlation with tumor metastasis and clinical significance. Pathol Oncol Res. 21:219–224. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Liu S, Qi L, Han W, Wan X, Jiang S, Li Y, Xie Y, Liu L, Zeng F, Liu Z and Zu X: Overexpression of Wip1 is associated with biologic behavior in human clear cell renal cell carcinoma. PLoS One. 9:e1102182014. View Article : Google Scholar : PubMed/NCBI

59 

Sun GG, Zhang J, Ma XB, Wang YD, Cheng YJ and Hu WN: Overexpression of wild-type p53-induced phosphatase1 confers poor prognosis of patients with nasopharyngeal carcinoma. Pathol Oncol Res. 21:283–291. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2017
Volume 14 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang ZP, Tian Y and Lin J: Role of wild-type p53-induced phosphatase 1 in cancer (Review). Oncol Lett 14: 3893-3898, 2017
APA
Wang, Z., Tian, Y., & Lin, J. (2017). Role of wild-type p53-induced phosphatase 1 in cancer (Review). Oncology Letters, 14, 3893-3898. https://doi.org/10.3892/ol.2017.6685
MLA
Wang, Z., Tian, Y., Lin, J."Role of wild-type p53-induced phosphatase 1 in cancer (Review)". Oncology Letters 14.4 (2017): 3893-3898.
Chicago
Wang, Z., Tian, Y., Lin, J."Role of wild-type p53-induced phosphatase 1 in cancer (Review)". Oncology Letters 14, no. 4 (2017): 3893-3898. https://doi.org/10.3892/ol.2017.6685