Oncogenic Wip1 phosphatase is inhibited by miR-16 in the DNA damage signaling pathway.

Wild-type p53-induced phosphatase 1 (Wip1) was identified as an oncogene amplified and overexpressed in several human cancers. Recent evidence suggested that Wip1 is a critical inhibitor in the ATM/ATR-p53 DNA damage signaling pathway. Wip1 dephosphorylates several key DNA damage-responsive proteins and reverses DNA damage-induced cell cycle checkpoints. Previous reports showed that Wip1 was transcriptionally induced by p53 at the early stage of the DNA damage response. To investigate the temporal and functional regulation of Wip1, we identified a microRNA, miR-16, that specifically targets the mRNA of Wip1 and thus negatively regulates the expression level of Wip1. miR-16 itself is induced immediately after DNA damage. Therefore, the increase in Wip1 protein level is significantly postponed compared with that of its mRNA level, preventing a premature inactivation of ATM/ATR signaling and allowing a functional completion of the early DNA damage response. To better understand miR-16 biological functions in the context of cancer cells, we examined its expression in mammary tumor stem cells and found it to be markedly downregulated in mammary tumor stem cells. Overexpression of miR-16 or inhibition of Wip1 suppresses the self-renewal and growth of mouse mammary tumor stem cells and sensitizes MCF-7 human breast cancer cells to the chemotherapeutic drug doxorubicin. Together, our results suggest an important role of miR-16 in the regulation of Wip1 phosphatase in the DNA damage response and mammary tumorigenesis.

[1]  A. Ashworth,et al.  An integrative genomic and transcriptomic analysis reveals molecular pathways and networks regulated by copy number aberrations in basal-like, HER2 and luminal cancers , 2010, Breast Cancer Research and Treatment.

[2]  R. Medema,et al.  Wip1 phosphatase is associated with chromatin and dephosphorylates γH2AX to promote checkpoint inhibition , 2010, Oncogene.

[3]  E. Yu,et al.  Expression of a Homeostatic Regulator, Wip1 (Wild-type p53-induced Phosphatase), Is Temporally Induced by c-Jun and p53 in Response to UV Irradiation* , 2010, The Journal of Biological Chemistry.

[4]  R. Gatti,et al.  ATM is down-regulated by N-Myc–regulated microRNA-421 , 2010, Proceedings of the National Academy of Sciences.

[5]  Xiongbin Lu,et al.  Phosphorylation and degradation of MdmX is inhibited by Wip1 phosphatase in the DNA damage response. , 2009, Cancer research.

[6]  Hiroshi I. Suzuki,et al.  Modulation of microRNA processing by p53 , 2009, Nature.

[7]  W. V. van IJcken,et al.  MicroRNA‐mediated gene silencing modulates the UV‐induced DNA‐damage response , 2009, The EMBO journal.

[8]  A. Ashworth,et al.  Tiling Path Genomic Profiling of Grade 3 Invasive Ductal Breast Cancers , 2009, Clinical Cancer Research.

[9]  Ettore Appella,et al.  Induction of PPM1D following DNA-damaging treatments through a conserved p53 response element coincides with a shift in the use of transcription initiation sites , 2008, Nucleic acids research.

[10]  Mauro Biffoni,et al.  The miR-15a–miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities , 2008, Nature Medicine.

[11]  Daniel Medina,et al.  Identification of tumor-initiating cells in a p53-null mouse model of breast cancer. , 2008, Cancer research.

[12]  A. Ashworth,et al.  A chemical inhibitor of PPM1D that selectively kills cells overexpressing PPM1D , 2008, Oncogene.

[13]  Yolanda F. Darlington,et al.  The type 2C phosphatase Wip1: An oncogenic regulator of tumor suppressor and DNA damage response pathways , 2008, Cancer and Metastasis Reviews.

[14]  Xiongbin Lu,et al.  The Wip1 Phosphatase acts as a gatekeeper in the p53-Mdm2 autoregulatory loop. , 2007, Cancer cell.

[15]  L. Donehower,et al.  Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D , 2007, Journal of Neuro-Oncology.

[16]  S. Semba,et al.  Increased wild‐type p53‐induced phosphatase 1 (Wip1 or PPM1D) expression correlated with downregulation of checkpoint kinase 2 in human gastric carcinoma , 2007, Pathology international.

[17]  E. Appella,et al.  The role of the MKK6/p38 MAPK pathway in Wip1-dependent regulation of ErbB2-driven mammary gland tumorigenesis , 2007, Oncogene.

[18]  S. Hirohashi,et al.  Genome‐wide array‐based comparative genomic hybridization analysis of pancreatic adenocarcinoma: Identification of genetic indicators that predict patient outcome , 2007, Cancer science.

[19]  E. Appella,et al.  Wip1 phosphatase modulates ATM-dependent signaling pathways. , 2006, Molecular cell.

[20]  A. Hoischen,et al.  Comprehensive genomic analysis of desmoplastic medulloblastomas: identification of novel amplified genes and separate evaluation of the different histological components , 2006, The Journal of pathology.

[21]  Päivikki Kauraniemi,et al.  The serine-threonine protein phosphatase PPM1D is frequently activated through amplification in aggressive primary breast tumours , 2006, Breast Cancer Research and Treatment.

[22]  C. Croce,et al.  miR-15 and miR-16 induce apoptosis by targeting BCL2. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[23]  L. Donehower,et al.  PPM1D dephosphorylates Chk1 and p53 and abrogates cell cycle checkpoints. , 2005, Genes & development.

[24]  L. Donehower,et al.  Reversal of the ATM/ATR-Mediated DNA Damage Response by the Oncogenic Phosphatase PPM1D , 2005, Cell cycle.

[25]  L. Donehower,et al.  The p53-induced oncogenic phosphatase PPM1D interacts with uracil DNA glycosylase and suppresses base excision repair. , 2004, Molecular cell.

[26]  G. Dontu,et al.  Role of Notch signaling in cell-fate determination of human mammary stem/progenitor cells , 2004, Breast Cancer Research.

[27]  L. Donehower,et al.  Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis through p38 MAPK–mediated activation of the p16Ink4a-p19Arf pathway , 2004, Nature Genetics.

[28]  G. Dontu,et al.  Stem cells in normal breast development and breast cancer , 2003, Cell proliferation.

[29]  J. Inazawa,et al.  Association of 17q21-q24 gain in ovarian clear cell adenocarcinomas with poor prognosis and identification of PPM1D and APPBP2 as likely amplification targets. , 2003, Clinical cancer research : an official journal of the American Association for Cancer Research.

[30]  G. Dontu,et al.  In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. , 2003, Genes & development.

[31]  Akira Nakagawara,et al.  PPM1D is a potential target for 17q gain in neuroblastoma. , 2003, Cancer research.

[32]  S. Morrison,et al.  Prospective identification of tumorigenic breast cancer cells , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[33]  Albert J. Fornace,et al.  Amplification of PPM1D in human tumors abrogates p53 tumor-suppressor activity , 2002, Nature Genetics.

[34]  Jeffrey R. Marks,et al.  Oncogenic properties of PPM1D located within a breast cancer amplification epicenter at 17q23 , 2002, Nature Genetics.

[35]  Lawrence A. Donehower,et al.  Mice Deficient for the Wild-Type p53-Induced Phosphatase Gene (Wip1) Exhibit Defects in Reproductive Organs, Immune Function, and Cell Cycle Control , 2002, Molecular and Cellular Biology.

[36]  Masaaki Adachi,et al.  p53‐inducible Wip1 phosphatase mediates a negative feedback regulation of p38 MAPK‐p53 signaling in response to UV radiation , 2000, The EMBO journal.

[37]  N. D. Kim,et al.  Stem cell characteristics of transplanted rat mammary clonogens. , 2000, Experimental cell research.

[38]  M. Fiscella,et al.  Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. , 1997, Proceedings of the National Academy of Sciences of the United States of America.