1
|
Kumar JP, Kosek D, Durell SR, Miller Jenkins LM, Debnath S, Coussens NP, Hall MD, Appella DH, Dyda F, Mazur SJ, Appella E. Crystal structure and mechanistic studies of the PPM1D serine/threonine phosphatase catalytic domain. J Biol Chem 2024; 300:107561. [PMID: 39002674 PMCID: PMC11342775 DOI: 10.1016/j.jbc.2024.107561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024] Open
Abstract
Protein phosphatase 1D (PPM1D, Wip1) is induced by the tumor suppressor p53 during DNA damage response signaling and acts as an oncoprotein in several human cancers. Although PPM1D is a potential therapeutic target, insights into its atomic structure were challenging due to flexible regions unique to this family member. Here, we report the first crystal structure of the PPM1D catalytic domain to 1.8 Å resolution. The structure reveals the active site with two Mg2+ ions bound, similar to other structures. The flap subdomain and B-loop, which are crucial for substrate recognition and catalysis, were also resolved, with the flap forming two short helices and three short β-strands that are followed by an irregular loop. Unexpectedly, a nitrogen-oxygen-sulfur bridge was identified in the catalytic domain. Molecular dynamics simulations and kinetic studies provided further mechanistic insights into the regulation of PPM1D catalytic activity. In particular, the kinetic experiments demonstrated a magnesium concentration-dependent lag in PPM1D attaining steady-state velocity, a feature of hysteretic enzymes that show slow transitions compared with catalytic turnover. All combined, these results advance the understanding of PPM1D function and will support the development of PPM1D-targeted therapeutics.
Collapse
Affiliation(s)
- Jay Prakash Kumar
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Dalibor Kosek
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Stewart R Durell
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Lisa M Miller Jenkins
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Subrata Debnath
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Nathan P Coussens
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, United States
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, United States
| | - Daniel H Appella
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Fred Dyda
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Ettore Appella
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States.
| |
Collapse
|
2
|
Belotserkovskaya E, Golotin V, Uyanik B, Demidov ON. Clonal haematopoiesis - a novel entity that modifies pathological processes in elderly. Cell Death Discov 2023; 9:345. [PMID: 37726289 PMCID: PMC10509183 DOI: 10.1038/s41420-023-01590-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 06/02/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023] Open
Abstract
Progress in the development of new sequencing techniques with wider accessibility and higher sensitivity of the protocol of deciphering genome particularities led to the discovery of a new phenomenon - clonal haematopoiesis. It is characterized by the presence in the bloodstream of elderly people a minor clonal population of cells with mutations in certain genes, but without any sign of disease related to the hematopoietic system. Here we will review this recent advancement in the field of clonal haematopoiesis and how it may affect the disease's development in old age.
Collapse
Affiliation(s)
| | - Vasily Golotin
- Institute of Cytology RAS, 4 Tikhoretskii prospect, St. Petersburg, 194064, Russia
- Saint Petersburg bra-nch of "VNIRO" ("Gos-NOIRH" named after L.S. Berg), Saint Petersburg, Russia
| | - Burhan Uyanik
- INSERM UMR1231, Laboratory of Excellence LipSTIC and label Ligue Nationale contre le Cancer, 7 Boulevard Jeanne d'Arc, Dijon, 21000, France
| | - Oleg N Demidov
- Institute of Cytology RAS, 4 Tikhoretskii prospect, St. Petersburg, 194064, Russia.
- INSERM UMR1231, Laboratory of Excellence LipSTIC and label Ligue Nationale contre le Cancer, 7 Boulevard Jeanne d'Arc, Dijon, 21000, France.
- Sirius University of Science and Technology, 1 Olimpiiskii pr-t, Sochi, 354340, Russian Federation.
| |
Collapse
|
3
|
Storchova R, Palek M, Palkova N, Veverka P, Brom T, Hofr C, Macurek L. Phosphorylation of TRF2 promotes its interaction with TIN2 and regulates DNA damage response at telomeres. Nucleic Acids Res 2023; 51:1154-1172. [PMID: 36651296 PMCID: PMC9943673 DOI: 10.1093/nar/gkac1269] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/25/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Protein phosphatase magnesium-dependent 1 delta (PPM1D) terminates the cell cycle checkpoint by dephosphorylating the tumour suppressor protein p53. By targeting additional substrates at chromatin, PPM1D contributes to the control of DNA damage response and DNA repair. Using proximity biotinylation followed by proteomic analysis, we identified a novel interaction between PPM1D and the shelterin complex that protects telomeric DNA. In addition, confocal microscopy revealed that endogenous PPM1D localises at telomeres. Further, we found that ATR phosphorylated TRF2 at S410 after induction of DNA double strand breaks at telomeres and this modification increased after inhibition or loss of PPM1D. TRF2 phosphorylation stimulated its interaction with TIN2 both in vitro and at telomeres. Conversely, induced expression of PPM1D impaired localisation of TIN2 and TPP1 at telomeres. Finally, recruitment of the DNA repair factor 53BP1 to the telomeric breaks was strongly reduced after inhibition of PPM1D and was rescued by the expression of TRF2-S410A mutant. Our results suggest that TRF2 phosphorylation promotes the association of TIN2 within the shelterin complex and regulates DNA repair at telomeres.
Collapse
Affiliation(s)
- Radka Storchova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Matous Palek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Natalie Palkova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Pavel Veverka
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Tomas Brom
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Ctirad Hofr
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| |
Collapse
|
4
|
Development of Antibody-like Proteins Targeting the Oncogenic Ser/Thr Protein Phosphatase PPM1D. Processes (Basel) 2022. [DOI: 10.3390/pr10081501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PPM1D, a protein Ser/Thr phosphatase, is overexpressed in various cancers and functions as an oncogenic protein by inactivating the p53 pathway. Therefore, molecules that bind PPM1D are expected to be useful anti-cancer agents. In this study, we constructed a phage display library based on the antibody-like small molecule protein adnectin and screened for PPM1D-specific binding molecules. We identified two adnectins, PMDB-1 and PMD-24, that bind PPM1D specific B-loop and PPM1D430 as targets, respectively. Specificity analyses of these recombinant proteins using other Ser/Thr protein phosphatases showed that these molecules bind to only PPM1D. Expression of PMDB-1 in breast cancer-derived MCF-7 cells overexpressing endogenous PPM1D stabilized p53, indicating that PMDB-1 functions as an inhibitor of PPM1D. Furthermore, MTT assay exhibited that MCF-7 cells expressing PMDB-1 showed inhibition of cell proliferation. These data suggest that the adnectin PMDB-1 identified in this study can be used as a lead compound for anti-cancer drugs targeting intracellular PPM1D.
Collapse
|
5
|
Inhibition of the DNA damage response phosphatase PPM1D reprograms neutrophils to enhance anti-tumor immune responses. Nat Commun 2021; 12:3622. [PMID: 34131120 PMCID: PMC8206133 DOI: 10.1038/s41467-021-23330-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
PPM1D/Wip1 is a negative regulator of the tumor suppressor p53 and is overexpressed in several human solid tumors. Recent reports associate gain-of-function mutations of PPM1D in immune cells with worse outcomes for several human cancers. Here we show that mice with genetic knockout of Ppm1d or with conditional knockout of Ppm1d in the hematopoietic system, in myeloid cells, or in neutrophils all display significantly reduced growth of syngeneic melanoma or lung carcinoma tumors. Ppm1d knockout neutrophils infiltrate tumors extensively. Chemical inhibition of Wip1 in human or mouse neutrophils increases anti-tumor phenotypes, p53-dependent expression of co-stimulatory ligands, and proliferation of co-cultured cytotoxic T cells. These results suggest that inhibition of Wip1 in neutrophils enhances immune anti-tumor responses.
Collapse
|
6
|
Al Hinai ASA, Grob T, Rijken M, Kavelaars FG, Zeilemaker A, Erpelinck-Verschueren CAJ, Sanders MA, Löwenberg B, Jongen-Lavrencic M, Valk PJM. PPM1D mutations appear in complete remission after exposure to chemotherapy without predicting emerging AML relapse. Leukemia 2021; 35:2693-2697. [PMID: 33589749 DOI: 10.1038/s41375-021-01155-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/05/2020] [Accepted: 01/25/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Adil S A Al Hinai
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands.,National Genetic Center, Ministry of Health, Muscat, Sultanate of Oman
| | - Tim Grob
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Melissa Rijken
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - François G Kavelaars
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Annelieke Zeilemaker
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Mathijs A Sanders
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bob Löwenberg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mojca Jongen-Lavrencic
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Peter J M Valk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Methods for Identification of Substrates/Inhibitors of FCP/SCP Type Protein Ser/Thr Phosphatases. Processes (Basel) 2020. [DOI: 10.3390/pr8121598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Protein phosphorylation is the most widespread type of post-translational modification and is properly controlled by protein kinases and phosphatases. Regarding the phosphorylation of serine (Ser) and threonine (Thr) residues, relatively few protein Ser/Thr phosphatases control the specific dephosphorylation of numerous substrates, in contrast with Ser/Thr kinases. Recently, protein Ser/Thr phosphatases were reported to have rigid substrate recognition and exert various biological functions. Therefore, identification of targeted proteins by individual protein Ser/Thr phosphatases is crucial to clarify their own biological functions. However, to date, information on the development of methods for identification of the substrates of protein Ser/Thr phosphatases remains scarce. In turn, substrate-trapping mutants are powerful tools to search the individual substrates of protein tyrosine (Tyr) phosphatases. This review focuses on the development of novel methods for the identification of Ser/Thr phosphatases, especially small C-terminal domain phosphatase 1 (Scp1), using peptide-displayed phage library with AlF4−/BeF3−, and discusses the identification of putative inhibitors.
Collapse
|
8
|
Development of Specific Inhibitors for Oncogenic Phosphatase PPM1D by Using Ion-Responsive DNA Aptamer Library. Catalysts 2020. [DOI: 10.3390/catal10101153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
(1) Background: Ser/Thr protein phosphatase PPM1D is an oncogenic protein. In normal cells, however, PPM1D plays essential roles in spermatogenesis and immune response. Hence, it is necessary to develop novel PPM1D inhibitors without side effects on normal cells. Stimuli-responsive molecules are suitable for the spatiotemporal regulation of inhibitory activity. (2) Methods: In this study, we designed an ion-responsive DNA aptamer library based on G-quadruplex DNA that can change its conformation and function in response to monovalent cations. (3) Results: Using this library, we identified the PPM1D specific inhibitor M1D-Q5F aptamer. The M1D-Q5F aptamer showed anti-cancer activity against breast cancer MCF7 cells. Interestingly, the induction of the structural change resulting in the formation of G-quadruplex upon stimulation by monovalent cations led to the enhancement of the inhibitory activity and binding affinity of M1D-Q5F. (4) Conclusions: These data suggest that the M1D-Q5F aptamer may act as a novel stimuli-responsive anti-cancer agent.
Collapse
|
9
|
Shi L, Tian Q, Feng C, Zhang P, Zhao Y. The biological function and the regulatory roles of wild-type p53-induced phosphatase 1 in immune system. Int Rev Immunol 2020; 39:280-291. [PMID: 32696682 DOI: 10.1080/08830185.2020.1795153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Wild-type p53-induced phosphatase 1 (WIP1) belongs to the protein phosphatase 2C (PP2C) family and is a mammalian serine/threonine specific protein phosphatase to dephosphorylate numerous signaling molecules. Mammalian WIP1 regulates a wide array of targeting molecules and plays key regulatory roles in many cell processes such as DNA damage and repair, cell proliferation, differentiation, apoptosis, and senescence. WIP1 promotes the formation and development of tumors as an oncogene and a negative regulator of p53. It is also involved in the regulation of aging, neurological diseases and immune diseases. Recent studies demonstrated the critical roles of WIP1 in the differentiation and function of immune cells including T cells, neutrophils and macrophages. In the present manuscript, we briefly summarized the expression patterns, biological function and the target molecules and signal pathways of WIP1 and mainly discussed the latest advances on the regulatory effects of WIP1 in the immune system. WIP1 may be a potential target molecule to treat cancers and immune diseases such as allergic asthma.
Collapse
Affiliation(s)
- Lu Shi
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qianchuan Tian
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chang Feng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Peng Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
10
|
Metal-dependent Ser/Thr protein phosphatase PPM family: Evolution, structures, diseases and inhibitors. Pharmacol Ther 2020; 215:107622. [PMID: 32650009 DOI: 10.1016/j.pharmthera.2020.107622] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Protein phosphatases and kinases control multiple cellular events including proliferation, differentiation, and stress responses through regulating reversible protein phosphorylation, the most important post-translational modification. Members of metal-dependent protein phosphatase (PPM) family, also known as PP2C phosphatases, are Ser/Thr phosphatases that bind manganese/magnesium ions (Mn2+/Mg2+) in their active center and function as single subunit enzymes. In mammals, there are 20 isoforms of PPM phosphatases: PPM1A, PPM1B, PPM1D, PPM1E, PPM1F, PPM1G, PPM1H, PPM1J, PPM1K, PPM1L, PPM1M, PPM1N, ILKAP, PDP1, PDP2, PHLPP1, PHLPP2, PP2D1, PPTC7, and TAB1, whereas there are only 8 in yeast. Phylogenetic analysis of the DNA sequences of vertebrate PPM isoforms revealed that they can be divided into 12 different classes: PPM1A/PPM1B/PPM1N, PPM1D, PPM1E/PPM1F, PPM1G, PPM1H/PPM1J/PPM1M, PPM1K, PPM1L, ILKAP, PDP1/PDP2, PP2D1/PHLPP1/PHLPP2, TAB1, and PPTC7. PPM-family members have a conserved catalytic core region, which contains the metal-chelating residues. The different isoforms also have isoform specific regions within their catalytic core domain and terminal domains, and these regions may be involved in substrate recognition and/or functional regulation of the phosphatases. The twenty mammalian PPM phosphatases are involved in regulating diverse cellular functions, such as cell cycle control, cell differentiation, immune responses, and cell metabolism. Mutation, overexpression, or deletion of the PPM phosphatase gene results in abnormal cellular responses, which lead to various human diseases. This review focuses on the structures and biological functions of the PPM-phosphatase family and their associated diseases. The development of specific inhibitors against the PPM phosphatase family as a therapeutic strategy will also be discussed.
Collapse
|
11
|
Ogasawara S, Chuman Y, Michiba T, Kamada R, Imagawa T, Sakaguchi K. Inhibition of protein phosphatase PPM1D enhances retinoic acid-induced differentiation in human embryonic carcinoma cell line. J Biochem 2018; 165:471-477. [PMID: 30576481 DOI: 10.1093/jb/mvy119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- Sari Ogasawara
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo, Japan
| | - Yoshiro Chuman
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo, Japan
| | - Takahiro Michiba
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo, Japan
| | - Rui Kamada
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo, Japan
| | - Toshiaki Imagawa
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo, Japan
| | - Kazuyasu Sakaguchi
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo, Japan
| |
Collapse
|
12
|
Inoue Y, Yamashita N, Kitao H, Tanaka K, Saeki H, Oki E, Oda Y, Tokunaga E, Maehara Y. Clinical Significance of the Wild Type p53-Induced Phosphatase 1 Expression in Invasive Breast Cancer. Clin Breast Cancer 2017; 18:e643-e650. [PMID: 29275106 DOI: 10.1016/j.clbc.2017.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/19/2017] [Accepted: 11/11/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND Wild type p53-induced phosphatase 1 (Wip1), encoded by the protein phosphatase magnesium dependent 1 delta (PPM1D), inhibits p53. PPM1D amplification has been reported in breast cancer. Breast cancer can sometimes develop without a tumor protein 53 (TP53) mutation. In these cases, the p53 pathway might be disrupted by alternative mechanisms, and Wip1 is reported to be a key molecule involved. MATERIALS AND METHODS Primary invasive ductal carcinoma specimens were obtained from 201 cases, for which archival tissue samples for immunohistochemistry were available. We evaluated Wip1 and p21 protein expression (201 cases), Wip1 mRNA expression (63 cases), PPM1D DNA copy number (71 cases) and TP53 status (36 cases) using available samples among the 201 cases, and analyzed their relationships with clinicopathological factors and prognosis. RESULTS The nuclear expression of Wip1 protein was positive in 21 cases (10.4%). The PPM1D DNA copy number was significantly correlated with Wip1 protein expression. All cases with PPM1D amplification by single-nucleotide polymorphism comparative genomic hybridization array showed positive nuclear Wip1 expression. Wip1 protein expression was positively correlated with p21 expression. The tumors with positive Wip1 and negative p21 expression showed the poorest prognosis among all tumor types. CONCLUSION The protein expression of Wip1 might be regulated by PPM1D amplification, independent of TP53 status. Positive Wip1 and negative p21 expression was associated with the poorest prognosis and suggests the loss of p53 function.
Collapse
Affiliation(s)
- Yuka Inoue
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Nami Yamashita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Hiroyuki Kitao
- Department of Molecular Cancer Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka City, Japan
| | - Kimihiro Tanaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Hiroshi Saeki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Eriko Tokunaga
- Departments of Breast Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka City, Japan.
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| |
Collapse
|
13
|
Jeong HJ, Woo CG, Lee B, Khang SK, Nam SJ, Choi J. Protein Phosphatase Magnesium-Dependent 1δ (PPM1D) Expression as a Prognostic Marker in Adult Supratentorial Diffuse Astrocytic and Oligodendroglial Tumors. J Pathol Transl Med 2017; 52:71-78. [PMID: 29046514 PMCID: PMC5859240 DOI: 10.4132/jptm.2017.10.21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 12/19/2022] Open
Abstract
Background Protein phosphatase magnesium-dependent 1δ (PPM1D) is a p53-induced serine/threonine phosphatase, which is overexpressed in various human cancers. A recent study reported that a mutation in the PPM1D gene is associated with poor prognosis in brainstem gliomas. In this study, we evaluated the utility of PPM1D as a prognostic biomarker of adult supratentorial diffuse astrocytic and oligodendroglial tumors. Methods To investigate PPM1D protein expression, mRNA expression, and copy number changes, immunohistochemistry, RNAscope in situ hybridization, and fluorescence in situ hybridization were performed in 84 adult supratentorial diffuse gliomas. We further analyzed clinical characteristics and overall survival (OS) according to PPM1D protein expression, and examined its correlation with other glioma biomarkers such as isocitrate dehydrogenase (IDH) mutation, and p53 expression. Results Forty-six cases (54.8%) were PPM1D-positive. PPM1D expression levels were significantly correlated with PPM1D transcript levels (p= .035), but marginally with PPM1D gene amplification (p=.079). Patients with high-grade gliomas showed a higher frequency of PPM1D expression than those with low-grade gliomas (p <.001). Multivariate analysis demonstrated that PPM1D expression (hazard ratio [HR], 2.58; p=.032), age over 60 years (HR, 2.55; p=.018), and IDH1 mutation (HR, 0.18; p=.002) were significantly independent prognostic factors; p53 expression had no prognostic significance (p=.986). The patients with tumor expressing PPM1D showed a shorter OS (p=.003). Moreover, patients with tumor harboring wild-type IDH1 and PPM1D expression had the worst OS (p<.001). Conclusions Our data suggest that a subset of gliomas express PPM1D; PPM1D expression is a significant marker of poor prognosis in adult supratentorial diffuse astrocytic and oligodendroglial tumors.
Collapse
Affiliation(s)
- Hui Jeong Jeong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chang Gok Woo
- Department of Pathology, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Bora Lee
- Department of Biostatistics, Clinical Trial Center, Soonchunhyang Medical Center, Bucheon, Korea
| | - Shin Kwang Khang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Jeong Nam
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jene Choi
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Lambert CA, Garbacki N, Colige AC. Chemotherapy induces alternative transcription and splicing: Facts and hopes for cancer treatment. Int J Biochem Cell Biol 2017; 91:84-97. [DOI: 10.1016/j.biocel.2017.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/04/2017] [Accepted: 04/15/2017] [Indexed: 01/14/2023]
|
15
|
Pecháčková S, Burdová K, Macurek L. WIP1 phosphatase as pharmacological target in cancer therapy. J Mol Med (Berl) 2017; 95:589-599. [PMID: 28439615 PMCID: PMC5442293 DOI: 10.1007/s00109-017-1536-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/13/2017] [Accepted: 04/19/2017] [Indexed: 12/31/2022]
Abstract
DNA damage response (DDR) pathway protects cells from genome instability and prevents cancer development. Tumor suppressor p53 is a key molecule that interconnects DDR, cell cycle checkpoints, and cell fate decisions in the presence of genotoxic stress. Inactivating mutations in TP53 and other genes implicated in DDR potentiate cancer development and also influence the sensitivity of cancer cells to treatment. Protein phosphatase 2C delta (referred to as WIP1) is a negative regulator of DDR and has been proposed as potential pharmaceutical target. Until recently, exploitation of WIP1 inhibition for suppression of cancer cell growth was compromised by the lack of selective small-molecule inhibitors effective at cellular and organismal levels. Here, we review recent advances in development of WIP1 inhibitors and discuss their potential use in cancer treatment.
Collapse
Affiliation(s)
- Soňa Pecháčková
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220, Prague, Czech Republic
| | - Kamila Burdová
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220, Prague, Czech Republic
| | - Libor Macurek
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220, Prague, Czech Republic.
| |
Collapse
|
16
|
Kozakai Y, Kamada R, Furuta J, Kiyota Y, Chuman Y, Sakaguchi K. PPM1D controls nucleolar formation by up-regulating phosphorylation of nucleophosmin. Sci Rep 2016; 6:33272. [PMID: 27619510 PMCID: PMC5020408 DOI: 10.1038/srep33272] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/24/2016] [Indexed: 01/01/2023] Open
Abstract
An increase of nucleolar number and size has made nucleoli essential markers for cytology and tumour development. However, the underlying basis for their structural integrity and abundance remains unclear. Protein phosphatase PPM1D was found to be up-regulated in different carcinomas including breast cancers. Here, we demonstrate for the first time that PPM1D regulates nucleolar formation via inducing an increased phosphorylation of the nucleolar protein NPM. We show that PPM1D overexpression induces an increase in the nucleolar number regardless of p53 status. We also demonstrated that specific sequential phosphorylation of NPM is important for nucleolar formation and that PPM1D is a novel upstream regulator of this phosphorylation pathway. These results enhance our understanding of the molecular mechanisms that govern nucleoli formation by demonstrating that PPM1D regulates nucleolar formation by regulating NPM phosphorylation status through a novel signalling pathway, PPM1D-CDC25C-CDK1-PLK1.
Collapse
Affiliation(s)
- Yuuki Kozakai
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Rui Kamada
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Junya Furuta
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Yuhei Kiyota
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Yoshiro Chuman
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Kazuyasu Sakaguchi
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
17
|
Esfandiari A, Hawthorne TA, Nakjang S, Lunec J. Chemical Inhibition of Wild-Type p53-Induced Phosphatase 1 (WIP1/PPM1D) by GSK2830371 Potentiates the Sensitivity to MDM2 Inhibitors in a p53-Dependent Manner. Mol Cancer Ther 2016; 15:379-91. [PMID: 26832796 DOI: 10.1158/1535-7163.mct-15-0651] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 12/23/2015] [Indexed: 01/10/2023]
Abstract
Sensitivity to MDM2 inhibitors is widely different among responsive TP53 wild-type cell lines and tumors. Understanding the determinants of MDM2 inhibitor sensitivity is pertinent for their optimal clinical application. Wild-type p53-inducible phosphatase-1 (WIP1) encoded by PPM1D, is activated, gained/amplified in a range of TP53 wild-type malignancies, and is involved in p53 stress response homeostasis. We investigated cellular growth/proliferation of TP53 wild-type and matched mutant/null cell line pairs, differing in PPM1D genetic status, in response to Nutlin-3/RG7388 ± a highly selective WIP1 inhibitor, GSK2830371. We also assessed the effects of GSK2830371 on MDM2 inhibitor-induced p53(Ser15) phosphorylation, p53-mediated global transcriptional activity, and apoptosis. The investigated cell line pairs were relatively insensitive to single-agent GSK2830371. However, a non-growth-inhibitory dose of GSK2830371 markedly potentiated the response to MDM2 inhibitors in TP53 wild-type cell lines, most notably in those harboring PPM1D-activating mutations or copy number gain (up to 5.8-fold decrease in GI50). Potentiation also correlated with significant increase in MDM2 inhibitor-induced cell death endpoints that were preceded by a marked increase in a WIP1 negatively regulated substrate, phosphorylated p53(Ser15), known to increase p53 transcriptional activity. Microarray-based gene expression analysis showed that the combination treatment increases the subset of early RG7388-induced p53 transcriptional target genes. These findings demonstrate that potent and selective WIP1 inhibition potentiates the response to MDM2 inhibitors in TP53 wild-type cells, particularly those with PPM1D activation or gain, while highlighting the mechanistic importance of p53(Ser15) and its potential use as a biomarker for response to this combination regimen.
Collapse
Affiliation(s)
- Arman Esfandiari
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Thomas A Hawthorne
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Sirintra Nakjang
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom. Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John Lunec
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
18
|
Reitman ZJ. Smaller protein, larger therapeutic potential: PPM1D as a new therapeutic target in brainstem glioma. Pharmacogenomics 2015; 15:1639-41. [PMID: 25410889 DOI: 10.2217/pgs.14.123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
19
|
Ogasawara S, Kiyota Y, Chuman Y, Kowata A, Yoshimura F, Tanino K, Kamada R, Sakaguchi K. Novel inhibitors targeting PPM1D phosphatase potently suppress cancer cell proliferation. Bioorg Med Chem 2015; 23:6246-9. [PMID: 26358280 DOI: 10.1016/j.bmc.2015.08.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 08/27/2015] [Accepted: 08/29/2015] [Indexed: 01/07/2023]
Abstract
Protein phosphatase magnesium-dependent 1δ (PPM1D, Wip1) is a p53 inducible serine/threonine phosphatase. PPM1D is a promising target protein in cancer therapy since overexpression, missense mutations, truncating mutations, and gene amplification of PPM1D are reported in many tumors, including breast cancer and neuroblastoma. Herein, we report that a specific inhibitor, SL-176 that can be readily synthesized in 10 steps, significantly inhibits proliferation of a breast cancer cell line overexpressing PPM1D and induces G2/M arrest and apoptosis. SL-176 decreases PPM1D enzyme activity potently and specifically in vitro. These results demonstrate that SL-176 could be a useful lead compound in the development of effective anti-cancer agents.
Collapse
Affiliation(s)
- Sari Ogasawara
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | - Yuhei Kiyota
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | - Yoshiro Chuman
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | - Ayano Kowata
- Laboratory of Organic Chemistry II, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Fumihiko Yoshimura
- Laboratory of Organic Chemistry II, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Keiji Tanino
- Laboratory of Organic Chemistry II, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Rui Kamada
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | - Kazuyasu Sakaguchi
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan.
| |
Collapse
|
20
|
Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, Chambert K, Mick E, Neale BM, Fromer M, Purcell SM, Svantesson O, Landén M, Höglund M, Lehmann S, Gabriel SB, Moran JL, Lander ES, Sullivan PF, Sklar P, Grönberg H, Hultman CM, McCarroll SA. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014; 371:2477-87. [PMID: 25426838 PMCID: PMC4290021 DOI: 10.1056/nejmoa1409405] [Citation(s) in RCA: 2403] [Impact Index Per Article: 240.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cancers arise from multiple acquired mutations, which presumably occur over many years. Early stages in cancer development might be present years before cancers become clinically apparent. METHODS We analyzed data from whole-exome sequencing of DNA in peripheral-blood cells from 12,380 persons, unselected for cancer or hematologic phenotypes. We identified somatic mutations on the basis of unusual allelic fractions. We used data from Swedish national patient registers to follow health outcomes for 2 to 7 years after DNA sampling. RESULTS Clonal hematopoiesis with somatic mutations was observed in 10% of persons older than 65 years of age but in only 1% of those younger than 50 years of age. Detectable clonal expansions most frequently involved somatic mutations in three genes (DNMT3A, ASXL1, and TET2) that have previously been implicated in hematologic cancers. Clonal hematopoiesis was a strong risk factor for subsequent hematologic cancer (hazard ratio, 12.9; 95% confidence interval, 5.8 to 28.7). Approximately 42% of hematologic cancers in this cohort arose in persons who had clonality at the time of DNA sampling, more than 6 months before a first diagnosis of cancer. Analysis of bone marrow-biopsy specimens obtained from two patients at the time of diagnosis of acute myeloid leukemia revealed that their cancers arose from the earlier clones. CONCLUSIONS Clonal hematopoiesis with somatic mutations is readily detected by means of DNA sequencing, is increasingly common as people age, and is associated with increased risks of hematologic cancer and death. A subset of the genes that are mutated in patients with myeloid cancers is frequently mutated in apparently healthy persons; these mutations may represent characteristic early events in the development of hematologic cancers. (Funded by the National Human Genome Research Institute and others.).
Collapse
|
21
|
Kozakai Y, Kamada R, Kiyota Y, Yoshimura F, Tanino K, Sakaguchi K. Inhibition of C-terminal truncated PPM1D enhances the effect of doxorubicin on cell viability in human colorectal carcinoma cell line. Bioorg Med Chem Lett 2014; 24:5593-5596. [PMID: 25466181 DOI: 10.1016/j.bmcl.2014.10.093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 10/27/2014] [Accepted: 10/29/2014] [Indexed: 12/15/2022]
Abstract
PPM1D is a p53-inducible Ser/Thr phosphatase. One of the main functions of PPM1D in normal cells is to act as a negative regulator of the p53 tumor suppressor by dephosphorylating p53 and several kinases. PPM1D is considered an oncoprotein owing to both its functions and the fact that gene amplification and overexpression of PPM1D are reported in several tumors. Recently, PPM1D mutations resulting in C-terminal truncated alterations were found in brainstem gliomas and colorectal cancers, and these mutations enhanced the activity of PPM1D. Therefore, C-terminal truncated PPM1D should be also considered as a potential candidate target of anticancer drugs. Here we showed that combination treatment with PPM1D-specific inhibitor SPI-001 and doxorubicin suppressed cell viability of HCT-116 cells overexpressing C-terminal truncated PPM1D through p53 activation compared with doxorubicin alone. Our results suggest that combination treatment with PPM1D inhibitor and doxorubicin may be a potential anti-cancer treatment in PPM1D-mutated cancer cells.
Collapse
Affiliation(s)
- Yuuki Kozakai
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | - Rui Kamada
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | - Yuhei Kiyota
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | - Fumihiko Yoshimura
- Laboratory of Organic Chemistry II, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | - Keiji Tanino
- Laboratory of Organic Chemistry II, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | - Kazuyasu Sakaguchi
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan.
| |
Collapse
|
22
|
Ali AY, Kim JY, Pelletier JF, Vanderhyden BC, Bachvarov DR, Tsang BK. Akt confers cisplatin chemoresistance in human gynecological carcinoma cells by modulating PPM1D stability. Mol Carcinog 2014; 54:1301-14. [PMID: 25154814 DOI: 10.1002/mc.22205] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/11/2014] [Accepted: 06/18/2014] [Indexed: 12/18/2022]
Abstract
Ovarian cancer (OVCA) and cervical cancer (CECA) are lethal gynecological malignancies. Cisplatin (CDDP) and platinum derivatives are first line chemotherapeutics and their resistance impedes successful treatment. Understanding the molecular dysregulation underlying chemoresistance is important in developing rational therapeutic strategies. We have established that Protein Phosphatase Magnesium-dependent 1 D (PPM1D) confers CDDP resistance in gynecological cancer cells by deactivating p53. However, whether CDDP regulates intra-cellular PPM1D localization and whether this regulation is different between chemosensitive and chemoresistant cancer cells is unknown. Moreover, whether Akt regulates PPM1D in the context of CDDP resistance has not been studied. To illustrate the role of PPM1D in gynecological cancer cell chemoresistance and its regulation by Akt we have demonstrated that: (a) CDDP induced PPM1D down-regulation through proteasomal degradation in sensitive CECA cells; (b) CDDP induced PPM1D nuclear localization in resistant CECA cells, and nuclear exclusion in sensitive CECA cells and OVCA xenografts; (c) Over-expression of active Akt in sensitive CECA cells stabilized PPM1D content through inhibition of CDDP-induced PPM1D down-regulation; (d) Inhibition of Akt activity in resistant OVCA cells leads to decreased PPM1D stability and CDDP-induced down-regulation in resistant CECA cells; and (e) PPM1D is highly expressed in human ovarian tumor subtypes and in a tissue microarray panel of human ovarian tumors. In conclusion, we have established that PPM1D plays an important role in promoting CDDP resistance and as a novel downstream target of Akt, PPM1D mediates its action in conferring CDDP resistance in gynecological cancer cells.
Collapse
Affiliation(s)
- Ahmed Y Ali
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Ji-Young Kim
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jean-François Pelletier
- Département de Médecine Moleculaire, Faculté de Médecine, Université Laval, Québec City, Québec, Canada.,Centre de Recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec City, Québec, Canada
| | - Barbara C Vanderhyden
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Obstetrics & Gynecology, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Dimcho R Bachvarov
- Département de Médecine Moleculaire, Faculté de Médecine, Université Laval, Québec City, Québec, Canada.,Centre de Recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec City, Québec, Canada
| | - Benjamin K Tsang
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Obstetrics & Gynecology, University of Ottawa, Ottawa, Ontario, Canada.,Department of Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada.,World Class University (WCU) Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
23
|
Akbari MR, Lepage P, Rosen B, McLaughlin J, Risch H, Minden M, Narod SA. PPM1D mutations in circulating white blood cells and the risk for ovarian cancer. J Natl Cancer Inst 2013; 106:djt323. [PMID: 24262437 DOI: 10.1093/jnci/djt323] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We compared the frequency of PPM1D mutation in the white blood cells from 1295 ovarian cancer case patients and 834 control subjects. We found a truncating mutation in 20 case patients vs 1 control subject (odds ratio [OR] = 13.07; 95% confidence interval [CI] = 1.75 to 97.55; P < .001). The 12-year mortality of the PPM1D-positive case patients was higher than that of the PPM1D-negative case patients (hazard ratio = 2.02; 95% CI = 1.21 to 3.39; P = .007). Three of the 20 PPM1D carrier case patients had a past history of breast cancer compared with 29 of 1129 noncarriers (OR = 6.69; 95% CI = 1.86 to 24.11; P = .007). The lifetime risks for breast or ovarian cancer among female first-degree relatives of PPM1D mutation carriers were not increased compared with that of case patients without mutations. These observations suggest PPM1D mutations in the mosaic state predispose women to breast and ovarian cancer in the absence of a family history of cancer.
Collapse
Affiliation(s)
- Mohammad R Akbari
- Affiliations of authors: Womens College Research Institute, Womens College Hospital (MRA, SAN), Dalla Lana School of Public Health (MRA, JM, SAN), Samuel Lunenfeld Research Institute (JM), University of Toronto, Toronto, Canada; Genome Quebec Innovation Centre, McGill University, Montreal, Canada (PL); Department of Gynecology-Oncology (BR), and Department of Medical Oncology (MM), Princess Margaret Hospital, Toronto, Canada; Department of Epidemiology and Public Health, School of Public Health, School of Medicine, Yale University, New Haven, CT (HR)
| | | | | | | | | | | | | |
Collapse
|
24
|
Nutlin-3a, an MDM2 antagonist and p53 activator, helps to preserve the replicative potential of cancer cells treated with a genotoxic dose of resveratrol. Mol Biol Rep 2013; 40:5013-26. [PMID: 23666059 PMCID: PMC3723979 DOI: 10.1007/s11033-013-2602-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 04/29/2013] [Indexed: 10/30/2022]
Abstract
Resveratrol is a natural compound that has been intensely studied due to its role in cancer prevention and potential as an anti-cancer therapy. Its effects include induction of apoptosis and senescence-like growth inhibition. Here, we report that two cancer cell lines (U-2 OS and A549) differ significantly in their molecular responses to resveratrol. Specifically, in U-2 OS cells, the activation of the p53 pathway is attenuated when compared to the activation in A549 cells. This attenuation is accompanied by a point mutation (458: CGA→TGA) in the PPM1D gene and overexpression of the encoded protein, which is a negative regulator of p53. Experimentally induced knockdown of PPM1D in U-2 OS cells resulted in slightly increased activation of the p53 pathway, most clearly visible as stronger phosphorylation of p53 Ser37. When treated with nutlin-3a, a non-genotoxic activator of p53, U-2 OS and A549 cells both responded with substantial activation of the p53 pathway. Nutlin-3a improved the clonogenic survival of both cell lines treated with resveratrol. This improvement was associated with lower activation of DNA-damage signaling (phosphorylation of ATM, CHK2, and histone H2AX) and higher accumulation of cells in the G1 phase of the cell cycle. Thus, the hyperactivation of p53 by nutlin-3a helps to preserve the replicative potential of cells exposed to resveratrol.
Collapse
|
25
|
Ali AY, Farrand L, Kim JY, Byun S, Suh JY, Lee HJ, Tsang BK. Molecular determinants of ovarian cancer chemoresistance: new insights into an old conundrum. Ann N Y Acad Sci 2013; 1271:58-67. [PMID: 23050965 PMCID: PMC3499654 DOI: 10.1111/j.1749-6632.2012.06734.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ovarian cancer is the most lethal gynecological malignancy. Cisplatin and its derivatives are first-line chemotherapeutics, and their resistance is a major hurdle in successful ovarian cancer treatment. Understanding the molecular dysregulation underlying chemoresistance is important for enhancing therapeutic outcome. Here, we review two established pathways in cancer chemoresistance. p53 is a major tumor suppressor regulating proliferation and apoptosis, and its mutation is a frequent event in human malignancies. The PI3K/Akt axis is a key oncogenic pathway regulating survival and tumorigenesis by controlling several tumor suppressors, including p53. The interplay between these pathways is well established, although the oncogenic phosphatase PPM1D adds a new layer to this intricate relationship and provides new insights into the processes determining cell fate. Inhibition of the PI3K/Akt pathway by functional food compounds as an adjunct to chemotherapeutics may tip the balance in favor of apoptosis rather than survival, enhancing therapeutic efficacy, and reducing side effects.
Collapse
Affiliation(s)
- Ahmed Y Ali
- Department of Cellular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
26
|
Ruark E, Snape K, Humburg P, Loveday C, Bajrami I, Brough R, Rodrigues DN, Renwick A, Seal S, Ramsay E, Duarte SDV, Rivas MA, Warren-Perry M, Zachariou A, Campion-Flora A, Hanks S, Murray A, Pour NA, Douglas J, Gregory L, Rimmer A, Walker NM, Yang TP, Adlard JW, Barwell J, Berg J, Brady AF, Brewer C, Brice G, Chapman C, Cook J, Davidson R, Donaldson A, Douglas F, Eccles D, Evans DG, Greenhalgh L, Henderson A, Izatt L, Kumar A, Lalloo F, Miedzybrodzka Z, Morrison PJ, Paterson J, Porteous M, Rogers MT, Shanley S, Walker L, Gore M, Houlston R, Brown MA, Caufield MJ, Deloukas P, McCarthy MI, Todd JA, Turnbull C, Reis-Filho JS, Ashworth A, Antoniou AC, Lord CJ, Donnelly P, Rahman N. Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer. Nature 2013; 493:406-10. [PMID: 23242139 PMCID: PMC3759028 DOI: 10.1038/nature11725] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 10/26/2012] [Indexed: 02/06/2023]
Abstract
Improved sequencing technologies offer unprecedented opportunities for investigating the role of rare genetic variation in common disease. However, there are considerable challenges with respect to study design, data analysis and replication. Using pooled next-generation sequencing of 507 genes implicated in the repair of DNA in 1,150 samples, an analytical strategy focused on protein-truncating variants (PTVs) and a large-scale sequencing case-control replication experiment in 13,642 individuals, here we show that rare PTVs in the p53-inducible protein phosphatase PPM1D are associated with predisposition to breast cancer and ovarian cancer. PPM1D PTV mutations were present in 25 out of 7,781 cases versus 1 out of 5,861 controls (P = 1.12 × 10(-5)), including 18 mutations in 6,912 individuals with breast cancer (P = 2.42 × 10(-4)) and 12 mutations in 1,121 individuals with ovarian cancer (P = 3.10 × 10(-9)). Notably, all of the identified PPM1D PTVs were mosaic in lymphocyte DNA and clustered within a 370-base-pair region in the final exon of the gene, carboxy-terminal to the phosphatase catalytic domain. Functional studies demonstrate that the mutations result in enhanced suppression of p53 in response to ionizing radiation exposure, suggesting that the mutant alleles encode hyperactive PPM1D isoforms. Thus, although the mutations cause premature protein truncation, they do not result in the simple loss-of-function effect typically associated with this class of variant, but instead probably have a gain-of-function effect. Our results have implications for the detection and management of breast and ovarian cancer risk. More generally, these data provide new insights into the role of rare and of mosaic genetic variants in common conditions, and the use of sequencing in their identification.
Collapse
Affiliation(s)
- Elise Ruark
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Katie Snape
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Peter Humburg
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Chey Loveday
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Ilirjana Bajrami
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Rachel Brough
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
- Cancer Research UK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Daniel Nava Rodrigues
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Anthony Renwick
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Sheila Seal
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Emma Ramsay
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | | | - Manuel A. Rivas
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7LD, UK
| | - Margaret Warren-Perry
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Anna Zachariou
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Adriana Campion-Flora
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Sandra Hanks
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Anne Murray
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Naser Ansari Pour
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Jenny Douglas
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Lorna Gregory
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Andrew Rimmer
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Neil M. Walker
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY, UK
| | - Tsun-Po Yang
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Julian W. Adlard
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds, LS7 4SA, UK
| | - Julian Barwell
- Leicestershire Genetics Centre, University Hospitals of Leicester NHS Trust, LE1 5WW, UK
| | - Jonathan Berg
- Human genetics, Division of Medical Sciences, University of Dundee, DD1 9SY, UK
| | - Angela F. Brady
- NW Thames Regional Genetics Service, Kennedy Galton Centre, London, HA1 3UJ, UK
| | - Carole Brewer
- Peninsula Regional Genetics Service, Royal Devon & Exeter Hospital, Exeter, EX1 2ED, UK
| | - Glen Brice
- SW Thames Regional Genetics Service, St George’s Hospital, London, SW17 0RE, UK
| | - Cyril Chapman
- West Midlands Regional Genetics Service, Birmingham Women’s Hospital, Birmingham, B15 2TG, UK
| | - Jackie Cook
- Sheffield Regional Genetics Service, Sheffield Children’s NHS Foundation Trust, S10 2TH, UK
| | - Rosemarie Davidson
- West of Scotland Regional Genetics Service, Laboratory Medicine, Southern General Hospital, Glasgow, G51 4TF, UK
| | - Alan Donaldson
- South Western Regional Genetics Service, University Hospitals of Bristol NHS Foundation Trust, BS2 8EG, UK
| | - Fiona Douglas
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, NE1 3BZ, UK
| | - Diana Eccles
- Faculty of Medicine, University of Southampton, Southampton University Hospitals NHS Trust, SO16 5YA, UK
| | - D. Gareth Evans
- Genetic Medicine, Manchester Academic Health Science Centre, St. Mary’s Hospital, Manchester M13 9WL, UK
| | - Lynn Greenhalgh
- Merseyside and Cheshire Clinical Genetics Service, Liverpool Women’s NHS Foundation Trust, Liverpool, L8 7SS, UK
| | - Alex Henderson
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, NE1 3BZ, UK
| | - Louise Izatt
- SE Thames Regional Genetics Service, Guy’s and St Thomas NHS Foundation Trust, London, SE1 9RT, UK
| | - Ajith Kumar
- NE Thames Regional Genetics Service, Great Ormond St Hospital, London, WC1N 3JH, UK
| | - Fiona Lalloo
- University Dept of Medical Genetics & Regional Genetics Service, St Mary’s Hospital, Manchester, M13 9WL, UK
| | - Zosia Miedzybrodzka
- University of Aberdeen and North of Scotland Clinical Genetics Service, Aberdeen Royal Infirmary, AB25 2ZA, UK
| | - Patrick J. Morrison
- Northern Ireland Regional Genetics Service, Belfast HSC Trust, Department of Medical Genetics, Queen’s University Belfast, BT9 7AB, UK
| | - Joan Paterson
- East Anglian Regional Genetics Service, Cambridge University Hospitals NHS Foundation Trust, CB2 0QQ, UK
| | - Mary Porteous
- South East of Scotland Clinical Genetics Service, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Mark T. Rogers
- All Wales Medical Genetics Service, University Hospital of Wales, Cardiff, CF14 4XW, UK
| | - Susan Shanley
- Dept of Cancer Genetics, Royal Marsden NHS Foundation Trust, Sutton, SM2 5PT, UK
| | - Lisa Walker
- Oxford Regional Genetics Service, Oxford University Hospitals NHS Trust, Oxford, OX3 7LJ, UK
| | - Martin Gore
- Dept of Gynaecologic Oncology, Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Richard Houlston
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Matthew A. Brown
- University of Queensland Diamantina Institute, University of Queensland, Princess Alexandra Hospital, Woolloongabba, Brisbane, 4102, Australia
| | - Mark J. Caufield
- Clinical Pharmacology and Barts and The London Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Panagiotis Deloukas
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Mark I. McCarthy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LI, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, OX3 7LI, UK
| | - John A. Todd
- Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY, UK
| | | | | | - Clare Turnbull
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
- Dept of Cancer Genetics, Royal Marsden NHS Foundation Trust, Sutton, SM2 5PT, UK
| | - Jorge S. Reis-Filho
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Alan Ashworth
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Antonis C. Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Christopher J. Lord
- The Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Peter Donnelly
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Department of Statistics, University of Oxford, Oxford, OX1 3TG, UK
| | - Nazneen Rahman
- Division of Genetics & Epidemiology, The Institute of Cancer Research, Sutton, SM2 5NG, UK
- Dept of Cancer Genetics, Royal Marsden NHS Foundation Trust, Sutton, SM2 5PT, UK
| |
Collapse
|
27
|
Zhu YH, Bulavin DV. Wip1-dependent signaling pathways in health and diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 106:307-25. [PMID: 22340722 DOI: 10.1016/b978-0-12-396456-4.00001-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Spatial and temporal regulation of protein phosphorylation is key to the control of different molecular networks. This regulation is achieved in part through dephosphorylation of numerous signaling molecules, and emerging evidence highlights the importance of a new member of the PP2C family of phosphatase, Wild-type p53 induced phosphatase 1 (Wip1), in regulating stress-induced and DNA damage-induced networks. In recent years, analysis of Wip1 has focused primarily on its role in tumorigenesis because of its overexpression in human tumors and a profound tumor-resistant phenotype of Wip1-deficient mice. Recently, Wip1 has also been shown to play an important role in several physiological processes including adult neurogenesis and organismal aging. This review addresses how Wip1 phosphatase regulates different signaling networks in a spatial and temporal manner and how these differences contribute to various biological outcomes in the context of physiological and pathological conditions.
Collapse
Affiliation(s)
- Yun-Hua Zhu
- Cell Cycle Control and Tumorigenesis Group, Institute of Molecular and Cell Biology, Proteos, Singapore
| | | |
Collapse
|
28
|
Yagi H, Chuman Y, Kozakai Y, Imagawa T, Takahashi Y, Yoshimura F, Tanino K, Sakaguchi K. A small molecule inhibitor of p53-inducible protein phosphatase PPM1D. Bioorg Med Chem Lett 2011; 22:729-32. [PMID: 22115592 DOI: 10.1016/j.bmcl.2011.10.084] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 10/05/2011] [Accepted: 10/07/2011] [Indexed: 12/29/2022]
Abstract
PPM1D is a p53-inducible Ser/Thr protein phosphatase. PPM1D gene amplification and overexpression have been reported in a variety of human tumors, including breast cancer and neuroblastoma. Because the phosphatase activity of PPM1D is essential for its oncogenic role, PPM1D inhibitors should be viable anti-cancer agents. In our current study, we showed that SPI-001 was a potent and specific PPM1D inhibitor. SPI-001 inhibited PPM1D phosphatase activity in PPM1D-overexpressing human breast cancer cells and increased phosphorylation of p53. Furthermore, SPI-001 suppressed cell proliferation by inducing apoptosis. Our present study suggested that SPI-001 was a potential lead compound in developing anti-cancer drugs.
Collapse
Affiliation(s)
- Hiroaki Yagi
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, North 10, West 8, Kita-ku, Hokkaido University, Sapporo 060-0810, Japan
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Chuman Y, Iizuka K, Honda T, Onoue H, Shimohigashi Y, Sakaguchi K. Phosphatase assay for multi-phosphorylated substrates using phosphatase specific-motif antibody. J Biochem 2011; 150:319-25. [PMID: 21558087 DOI: 10.1093/jb/mvr056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Protein phosphorylation plays central roles in a wide variety of signal transduction pathways and most phosphorylated proteins contain multi-phosphorylated sites. PPM1 type Ser/Thr protein phosphatase family is known to show rigid substrate specificity unlike other Ser/Thr phosphatase PPP family including PP1, PP2A and PP2B. PPM1 type phosphatases are reported to play important roles in growth regulation and in cellular stress signalling. In this study, we developed a phosphatase assay of PPM1D using phosphatase motif-specific antibody. PPM1D is a member of PPM1 type Ser/Thr phosphatase and known to dephosphorylate Ser(P)-Gln sequence. The gene amplification and overexpression of PPM1D were reported in many human cancers. We generated the monoclonal antibody specific for the Ser(P)-Gln sequence, named 3G9-H11. The specificity of this method using ELISA enables the convenient measurement of the dephosphorylation level of only PPM1D target residues of substrate peptides with multiple phosphorylated sites in the presence of multiple phosphatases. In addition, the antibody was applicable to immunoblotting assay for PPM1D function analysis. These results suggested that this method should be very useful for the PPM1D phosphatase assay, including high-throughput analysis and screening of specific inhibitors as anti-cancer drugs. The method using phosphatase motif-specific antibody can be applied to other PPM1 phosphatase family.
Collapse
Affiliation(s)
- Yoshiro Chuman
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, 060-0810, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Macůrek L, Lindqvist A, Voets O, Kool J, Vos HR, Medema RH. Wip1 phosphatase is associated with chromatin and dephosphorylates γH2AX to promote checkpoint inhibition. Oncogene 2010; 29:2281-91. [DOI: 10.1038/onc.2009.501] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
31
|
Le Guezennec X, Bulavin DV. WIP1 phosphatase at the crossroads of cancer and aging. Trends Biochem Sci 2009; 35:109-14. [PMID: 19879149 DOI: 10.1016/j.tibs.2009.09.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 09/10/2009] [Accepted: 09/10/2009] [Indexed: 01/07/2023]
Abstract
The PP2C family serine/threonine phosphatase WIP1 is characterized by distinctive oncogenic properties mediated by inhibitory functions on several tumor suppressor pathways, including ATM, CHK2, p38MAPK and p53. PPM1D, the gene encoding WIP1, is aberrantly amplified in different types of human primary cancers, and its deletion in mice results in a profound tumor-resistant phenotype. Numerous downstream targets of WIP1 have been identified, and genetic studies confirm that some play a part in tumorigenesis. Recent evidence highlights a new role for WIP1 in the regulation of a cell-autonomous decline in proliferation of certain self-renewing cell types, including pancreatic beta-cells, with advancing age. These emerging functions of WIP1 make it a potent therapeutic target against cancer and aging.
Collapse
Affiliation(s)
- Xavier Le Guezennec
- Institute of Molecular and Cell Biology, Cell Cycle Control and Tumorigenesis Group, 61 Biopolis Drive, Proteos, Singapore
| | | |
Collapse
|
32
|
Wip1, an oncogene targeting tumor suppressors expressed in intestinal stem cells. CURRENT COLORECTAL CANCER REPORTS 2009. [DOI: 10.1007/s11888-009-0027-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|