1
|
Lagorgette L, Bogdanova DA, Belotserkovskaya EV, Garrido C, Demidov ON. PP2C phosphatases-terminators of suicidal thoughts. Cell Death Dis 2024; 15:919. [PMID: 39702569 DOI: 10.1038/s41419-024-07269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/16/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024]
Abstract
Cell death and related signaling pathways are essential during development and in various physiological and pathological conditions. Post-translational modifications such as ubiquitination and phosphorylation play an important role in these signaling pathways. The involvement of kinases - enzymes that catalyze protein phosphorylation - in cell death signaling has been extensively studied. On the other hand, not many studies have been devoted to analyzing the role in cell death of phosphatases, enzymes involved in the removal of phosphorylated residues added to proteins by kinases. Obviously, the two opposite reactions, phosphorylation and dephosphorylation, are equally important in the regulation of protein functions and subsequently in the execution of the cell death program. Here, we have summarized recent work on the involvement of serine-threonine PP2C phosphatases in cell death pathways, senescence and autophagy, focusing in particular on the most studied phosphatase PPM1D (PP2Cδ) as an example of the regulatory role of PP2Cs in cell death. The review should help to draw attention to the importance of PP2C family phosphatases in cell death checkpoints and to discover new targets for drug development.
Collapse
Affiliation(s)
- Lisa Lagorgette
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
| | - Daria A Bogdanova
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia
- Institute of Cytology RAS, St. Petersburg, Russia
| | | | - Carmen Garrido
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France
- Center for Cancer Georges-François Leclerc, Dijon, France
| | - Oleg N Demidov
- INSERM, UMR 1231, Laboratoire d'Excellence LipSTIC and « Equipe labellisée par la Ligue Nationale contre le Cancer », University of Burgundy, Dijon, France.
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius University of Science and Technology, Sochi, Russia.
- Institute of Cytology RAS, St. Petersburg, Russia.
| |
Collapse
|
2
|
Zhang L, Hsu JI, Goodell MA. PPM1D in Solid and Hematologic Malignancies: Friend and Foe? Mol Cancer Res 2022; 20:1365-1378. [PMID: 35657598 PMCID: PMC9437564 DOI: 10.1158/1541-7786.mcr-21-1018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 01/07/2023]
Abstract
In the face of constant genomic insults, the DNA damage response (DDR) is initiated to preserve genome integrity; its disruption is a classic hallmark of cancer. Protein phosphatase Mg2+/Mn2+-dependent 1D (PPM1D) is a central negative regulator of the DDR that is mutated or amplified in many solid cancers. PPM1D overexpression is associated with increased proliferative and metastatic behavior in multiple solid tumor types and patients with PPM1D-mutated malignancies have poorer prognoses. Recent findings have sparked an interest in the role of PPM1D in hematologic malignancies. Acquired somatic mutations may provide hematopoietic stem cells with a competitive advantage, leading to a substantial proportion of mutant progeny in the peripheral blood, an age-associated phenomenon termed "clonal hematopoiesis" (CH). Recent large-scale genomic studies have identified PPM1D to be among the most frequently mutated genes found in individuals with CH. While PPM1D mutations are particularly enriched in patients with therapy-related myeloid neoplasms, their role in driving leukemic transformation remains uncertain. Here, we examine the mechanisms through which PPM1D overexpression or mutation may drive malignancy by suppression of DNA repair, cell-cycle arrest, and apoptosis. We also discuss the divergent roles of PPM1D in the oncogenesis of solid versus hematologic cancers with a view to clinical implications and new therapeutic avenues.
Collapse
Affiliation(s)
- Linda Zhang
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
| | - Joanne I. Hsu
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, Texas
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
| | - Margaret A. Goodell
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Corresponding Author: Margaret A. Goodell, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030. E-mail:
| |
Collapse
|
3
|
MiR-145-5p Inhibits the Invasion of Prostate Cancer and Induces Apoptosis by Inhibiting WIP1. JOURNAL OF ONCOLOGY 2021; 2021:4412705. [PMID: 34899906 PMCID: PMC8660234 DOI: 10.1155/2021/4412705] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 11/26/2022]
Abstract
Prostate cancer (PCa) is a common malignant tumor of the male genitourinary system that seriously affects the quality of life of patients. Studying the pathogenesis and therapeutic targets of PCa is important. In this study, we investigated the role of miR-145-5p in PCa and its potential molecular mechanisms. The expression levels of miR-145-5p in PCa tissues and adjacent control tissues were detected by real-time quantitative polymerase chain reaction. The effects of miR-145-5p overexpression on PCa were studied using cell proliferation, migration, and invasion experiments. Furthermore, WIP1 was the target gene of miR-145-5p through the bioinformatics website and dual-luciferase reporter gene experiment. Further studies found that WIP1 downregulation could inhibit the proliferation, invasion, and cloning of PCa cells. Overexpression of WIP1 reversed the anticancer effects of miR-145. The anticancer effect of miR-145 was achieved by inhibiting the PI3K/AKT signaling pathway and upregulating ChK2 and p-p38MAPK. Taken together, these results confirmed that miR-145-5p inhibited the growth and metastasis of PCa cells by inhibiting the expression of proto-oncogene WIP1, thereby playing a role in tumor suppression in PCa and may become a potential therapeutic target for the treatment of PCa.
Collapse
|
4
|
Pharmacological Inhibition of WIP1 Sensitizes Acute Myeloid Leukemia Cells to the MDM2 Inhibitor Nutlin-3a. Biomedicines 2021; 9:biomedicines9040388. [PMID: 33917342 PMCID: PMC8067413 DOI: 10.3390/biomedicines9040388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/23/2021] [Accepted: 03/27/2021] [Indexed: 12/11/2022] Open
Abstract
In acute myeloid leukemia (AML), the restoration of p53 activity through MDM2 inhibition proved efficacy in combinatorial therapies. WIP1, encoded from PPM1D, is a negative regulator of p53. We evaluated PPM1D expression and explored the therapeutic efficacy of WIP1 inhibitor (WIP1i) GSK2830371, in association with the MDM2 inhibitor Nutlin-3a (Nut-3a) in AML cell lines and primary samples. PPM1D transcript levels were higher in young patients compared with older ones and in core-binding-factor AML compared with other cytogenetic subgroups. In contrast, its expression was reduced in NPM1-mutated (mut, irrespective of FLT3-ITD status) or TP53-mut cases compared with wild-type (wt) ones. Either Nut-3a, and moderately WIP1i, as single agent decreased cell viability of TP53-wt cells (MV-4-11, MOLM-13, OCI-AML3) in a time/dosage-dependent manner, but not of TP53-mut cells (HEL, KASUMI-1, NOMO-1). The drug combination synergistically reduced viability and induced apoptosis in TP53-wt AML cell line and primary cells, but not in TP53-mut cells. Gene expression and immunoblotting analyses showed increased p53, MDM2 and p21 levels in treated TP53-wt cells and highlighted the enrichment of MYC, PI3K-AKT-mTOR and inflammation-related signatures upon WIP1i, Nut-3a and their combination, respectively, in the MV-4-11 TP53-wt model. This study demonstrated that WIP1 is a promising therapeutic target to enhance Nut-3a efficacy in TP53-wt AML.
Collapse
|
5
|
Zhang X, Park JE, Kim EH, Hong J, Hwang KT, Kim YA, Jang CY. Wip1 controls the translocation of the chromosomal passenger complex to the central spindle for faithful mitotic exit. Cell Mol Life Sci 2021; 78:2821-2838. [PMID: 33067654 PMCID: PMC11072438 DOI: 10.1007/s00018-020-03665-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 08/12/2020] [Accepted: 10/05/2020] [Indexed: 10/23/2022]
Abstract
Dramatic cellular reorganization in mitosis critically depends on the timely and temporal phosphorylation of a broad range of proteins, which is mediated by the activation of the mitotic kinases and repression of counteracting phosphatases. The mitosis-to-interphase transition, which is termed mitotic exit, involves the removal of mitotic phosphorylation by protein phosphatases. Although protein phosphatase 1 (PP1) and protein phosphatase 2A (PP2A) drive this reversal in animal cells, the phosphatase network associated with ordered bulk dephosphorylation in mitotic exit is not fully understood. Here, we describe a new mitotic phosphatase relay in which Wip1/PPM1D phosphatase activity is essential for chromosomal passenger complex (CPC) translocation to the anaphase central spindle after release from the chromosome via PP1-mediated dephosphorylation of histone H3T3. Depletion of endogenous Wip1 and overexpression of the phosphatase-dead mutant disturbed CPC translocation to the central spindle, leading to failure of cytokinesis. While Wip1 was degraded in early mitosis, its levels recovered in anaphase and the protein functioned as a Cdk1-counteracting phosphatase at the anaphase central spindle and midbody. Mechanistically, Wip1 dephosphorylated Thr-59 in inner centromere protein (INCENP), which, subsequently bound to MKLP2 and recruited other components to the central spindle. Furthermore, Wip1 overexpression is associated with the overall survival rate of patients with breast cancer, suggesting that Wip1 not only functions as a weak oncogene in the DNA damage network but also as a tumor suppressor in mitotic exit. Altogether, our findings reveal that sequential dephosphorylation of mitotic phosphatases provides spatiotemporal regulation of mitotic exit to prevent tumor initiation and progression.
Collapse
Affiliation(s)
- Xianghua Zhang
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ji Eun Park
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Catholic University of Daegu, Daegu, 42472, Republic of Korea
| | - Jihee Hong
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ki-Tae Hwang
- Department of Surgery, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Young A Kim
- Department of Pathology, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea.
| | - Chang-Young Jang
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
6
|
Martinikova AS, Burocziova M, Stoyanov M, Macurek L. Truncated PPM1D Prevents Apoptosis in the Murine Thymus and Promotes Ionizing Radiation-Induced Lymphoma. Cells 2020; 9:cells9092068. [PMID: 32927737 PMCID: PMC7565556 DOI: 10.3390/cells9092068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 12/20/2022] Open
Abstract
Genome integrity is protected by the cell-cycle checkpoints that prevent cell proliferation in the presence of DNA damage and allow time for DNA repair. The transient checkpoint arrest together with cellular senescence represent an intrinsic barrier to tumorigenesis. Tumor suppressor p53 is an integral part of the checkpoints and its inactivating mutations promote cancer growth. Protein phosphatase magnesium-dependent 1 (PPM1D) is a negative regulator of p53. Although its loss impairs recovery from the G2 checkpoint and promotes induction of senescence, amplification of the PPM1D locus or gain-of-function truncating mutations of PPM1D occur in various cancers. Here we used a transgenic mouse model carrying a truncating mutation in exon 6 of PPM1D (Ppm1dT). As with human cell lines, we found that the truncated PPM1D was present at high levels in the mouse thymus. Truncated PPM1D did not affect differentiation of T-cells in the thymus but it impaired their response to ionizing radiation (IR). Thymocytes in Ppm1dT/+ mice did not arrest in the checkpoint and continued to proliferate despite the presence of DNA damage. In addition, we observed a decreased level of apoptosis in the thymi of Ppm1dT/+ mice. Moreover, the frequency of the IR-induced T-cell lymphomas increased in Ppm1dT/+Trp53+/- mice resulting in decreased survival. We conclude that truncated PPM1D partially suppresses the p53 pathway in the mouse thymus and potentiates tumor formation under the condition of a partial loss of p53 function.
Collapse
Affiliation(s)
- Andra S. Martinikova
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, CZ14220 Prague, Czech Republic; (A.S.M.); (M.B.); (M.S.)
- Department of Developmental and Cell Biology, Faculty of Science, Charles University, Albertov 6, CZ12800 Prague, Czech Republic
| | - Monika Burocziova
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, CZ14220 Prague, Czech Republic; (A.S.M.); (M.B.); (M.S.)
| | - Miroslav Stoyanov
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, CZ14220 Prague, Czech Republic; (A.S.M.); (M.B.); (M.S.)
| | - Libor Macurek
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, CZ14220 Prague, Czech Republic; (A.S.M.); (M.B.); (M.S.)
- Correspondence: ; Tel.: +42-(0)2-4106-3210
| |
Collapse
|
7
|
Li B, Hu J, He D, Chen Q, Liu S, Zhu X, Yu M. PPM1D Knockdown Suppresses Cell Proliferation, Promotes Cell Apoptosis, and Activates p38 MAPK/p53 Signaling Pathway in Acute Myeloid Leukemia. Technol Cancer Res Treat 2020; 19:1533033820942312. [PMID: 32691668 PMCID: PMC7375723 DOI: 10.1177/1533033820942312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES This study was to explore the effect of protein phosphatase, Mg2+/Mn2+ dependent 1D knockdown on proliferation and apoptosis as well as p38 MAPK/p53 signaling pathway in acute myeloid leukemia. METHODS The expression of protein phosphatase, Mg2+/Mn2+ dependent 1D was detected in acute myeloid leukemia cell lines including SKM-1, KG-1, AML-193, and THP-1 cells, and normal bone marrow mononuclear cells isolated from healthy donors. The knockdown of protein phosphatase, Mg2+/Mn2+ dependent 1D was conducted by transfecting small interfering RNA into AML-193 cells and KG-1 cells. RESULTS The relative messenger RNA/protein expressions of protein phosphatase, Mg2+/Mn2+ dependent 1D were higher in SKM-1, KG-1, AML-193, and THP-1 cells compared with control cells (normal bone marrow mononuclear cells). After transfecting protein phosphatase, Mg2+/Mn2+ dependent 1D small interfering RNA into AML-193 cells and KG-1 cells, both messenger RNA and protein expressions of protein phosphatase, Mg2+/Mn2+ dependent 1D were significantly reduced, indicating the successful transfection. Most importantly, knockdown of protein phosphatase, Mg2+/Mn2+ dependent 1D suppressed cell proliferation and promoted cell apoptosis in AML-193 cells and KG-1 cells. In addition, knockdown of protein phosphatase, Mg2+/Mn2+ dependent 1D enhanced the expressions of p-p38 and p53 in AML-193 cells and KG-1 cells. The above observation suggested that protein phosphatase, Mg2+/Mn2+ dependent 1D knockdown suppressed cell proliferation, promoted cell apoptosis, and activated p38 MAPK/p53 signaling pathway in acute myeloid leukemia cells. CONCLUSION Protein phosphatase, Mg2+/Mn2+ dependent 1D is implicated in acute myeloid leukemia carcinogenesis, which illuminates its potential role as a treatment target for acute myeloid leukemia.
Collapse
Affiliation(s)
- Bin Li
- Department of Hematology, The Second People's Hospital of Yunnan Province, Yunnan, China
| | - Jie Hu
- Department of Hematology, The Second People's Hospital of Yunnan Province, Yunnan, China
| | - Di He
- Department of Hematology, The Second People's Hospital of Yunnan Province, Yunnan, China
| | - Qi Chen
- Department of Hematology, The Second People's Hospital of Yunnan Province, Yunnan, China
| | - Suna Liu
- Department of Hematology, The Second People's Hospital of Yunnan Province, Yunnan, China
| | - Xiaoling Zhu
- Department of Hematology, The Second People's Hospital of Yunnan Province, Yunnan, China
| | - Meijia Yu
- Department of Hematology, The Second People's Hospital of Yunnan Province, Yunnan, China
| |
Collapse
|
8
|
Deng W, Li J, Dorrah K, Jimenez-Tapia D, Arriaga B, Hao Q, Cao W, Gao Z, Vadgama J, Wu Y. The role of PPM1D in cancer and advances in studies of its inhibitors. Biomed Pharmacother 2020; 125:109956. [PMID: 32006900 PMCID: PMC7080581 DOI: 10.1016/j.biopha.2020.109956] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/08/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
A greater understanding of factors causing cancer initiation, progression and evolution is of paramount importance. Among them, the serine/threonine phosphatase PPM1D, also referred to as wild-type p53-induced phosphatase 1 (Wip1) or protein phosphatase 2C delta (PP2Cδ), is emerging as an important oncoprotein due to its negative regulation on a number of crucial cancer suppressor pathways. Initially identified as a p53-regulated gene, PPM1D has been afterwards found amplified and more recently mutated in many human cancers such as breast cancer. The latest progress in this field further reveals that selective inhibition of PPM1D to delay tumor onset or reduce tumor burden represents a promising anti-cancer strategy. Here, we review the advances in the studies of the PPM1D activity and its relevance to various cancers, and recent progress in development of PPM1D inhibitors and discuss their potential application in cancer therapy. Consecutive research on PPM1D and its relationship with cancer is essential, as it ultimately contributes to the etiology and treatment of cancer.
Collapse
Affiliation(s)
- Wenhong Deng
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Jieqing Li
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Kimberly Dorrah
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Denise Jimenez-Tapia
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Brando Arriaga
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Qiongyu Hao
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Wei Cao
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Zhaoxia Gao
- Department of General Surgery, 5th Hospital of Wuhan, Wuhan, 430050, China; Department of Surgery, Johns Hopkins Hospital Bayview Campus, Baltimore, MD, USA
| | - Jay Vadgama
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Gupta AA, Shekatkar M, Raj AT, Kheur S. Potential Role of Magnesium in Cancer Initiation and Progression. Pathol Oncol Res 2019; 26:2001-2002. [PMID: 31494817 DOI: 10.1007/s12253-019-00750-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Archana A Gupta
- Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, 411041, India.
| | - Madhura Shekatkar
- Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, 411041, India
| | - A Thirumal Raj
- Department of Oral Pathology and Microbiology, Sri Venkateswara Dental College and Hospital, Chennai, India
| | - Supriya Kheur
- Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, 411041, India
| |
Collapse
|
10
|
Mendes PMV, Bezerra DLC, Dos Santos LR, de Oliveira Santos R, de Sousa Melo SR, Morais JBS, Severo JS, Vieira SC, do Nascimento Marreiro D. Magnesium in Breast Cancer: What Is Its Influence on the Progression of This Disease? Biol Trace Elem Res 2018; 184:334-339. [PMID: 29198048 DOI: 10.1007/s12011-017-1207-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 11/15/2017] [Indexed: 12/22/2022]
Abstract
Breast cancer is a disease of high mortality, characterized by the progressive accumulation of mutations that contribute to the uncontrolled development of breast tissue cells. Literature shows that disturbances in mineral homeostasis, such as magnesium, may interfere with tumor progression. The objective of this study is to provide updated information about magnesium's role in the pathogenesis of breast cancer. A review of literature was carried out from a search for articles in the PubMed and CAPES Periodicals databases published between 1995 and 2016 with the cross-references of the descriptors "magnesium," "breast neoplasms," and "oxidative stress" and the corresponding words in Portuguese. We included studies on the metabolism and bioavailability of magnesium and studies related to breast cancer and excluded articles in which only the abstract was available, dissertations, theses, articles involving adjuvant and/or neoadjuvant therapies, and supplementation of minerals in breast cancer patients. Magnesium is a mineral that participates in the metabolism of various nutrients and nucleic acids. In the presence of breast cancer, neoplastic cells increase the expression of magnesium transport channels, which raises the intracellular concentration of the mineral, contributing to tumor growth through its function of increasing energy demand. The data obtained in this review illustrates the influence of magnesium on the progression of breast cancer. However, the existing data are scarce and inconsistent, which demonstrates a need for further studies on the subject with a goal to have better control of the disease.
Collapse
Affiliation(s)
- Priscyla Maria Vieira Mendes
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apt°. 2001, Jóquei, Teresina, PI, CEP 64048-320, Brazil
| | - Daila Leite Chaves Bezerra
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apt°. 2001, Jóquei, Teresina, PI, CEP 64048-320, Brazil
| | - Loanne Rocha Dos Santos
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apt°. 2001, Jóquei, Teresina, PI, CEP 64048-320, Brazil
| | - Raísa de Oliveira Santos
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apt°. 2001, Jóquei, Teresina, PI, CEP 64048-320, Brazil
| | - Stéfany Rodrigues de Sousa Melo
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apt°. 2001, Jóquei, Teresina, PI, CEP 64048-320, Brazil
| | - Jennifer Beatriz Silva Morais
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apt°. 2001, Jóquei, Teresina, PI, CEP 64048-320, Brazil
| | - Juliana Soares Severo
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apt°. 2001, Jóquei, Teresina, PI, CEP 64048-320, Brazil
| | - Sabas Carlos Vieira
- Department of Medicine, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Teresina, PI, Brazil
| | - Dilina do Nascimento Marreiro
- Department of Nutrition, Federal University of Piauí, Ministro Petrônio Portella Campus, Ininga, Rua Hugo Napoleão, 665, Ed. Palazzo Reale, Apt°. 2001, Jóquei, Teresina, PI, CEP 64048-320, Brazil.
| |
Collapse
|
11
|
Abstract
Cells undergoing oncogenic transformation frequently inactivate tumor suppressor pathways that could prevent their uncontrolled growth. Among those pathways p53 and p38MAPK pathways play a critical role in regulation of cell cycle, senescence and cell death in response to activation of oncogenes, stress and DNA damage. Consequently, these two pathways are important in determining the sensitivity of tumor cells to anti-cancer treatment. Wild type p53-induced phosphatase, Wip1, is involved in governance of both pathways. Recently, strategies directed to manipulation with Wip1 activity proposed to advance current day anticancer treatment and novel chemical compounds synthesized to improve specificity of manipulation with Wip1 activity. Here we reviewed the history of Wip1 studies in vitro and in vivo, in genetically modified animal models that support Wip1 role in tumorigenesis through regulation of p53 and p38MAPK pathways. Based on our knowledge we propose several recommendations for future more accurate studies of Wip1 interactions with other pathways involved in tumorigenesis using recently developed tools and for adoption of Wip1 manipulation strategies in anti-cancer therapy.
Collapse
|
12
|
Rasmussen MK, Nielsen J, Kjellerup RB, Andersen SM, Rittig AH, Johansen C, Iversen L, Gesser B. Protein phosphatase 2Cδ/Wip1 regulates phospho-p90RSK2 activity in lesional psoriatic skin. J Inflamm Res 2017; 10:169-180. [PMID: 29290690 PMCID: PMC5735993 DOI: 10.2147/jir.s152869] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objectives P90 ribosomal S6 kinase (RSK) 1 and 2 are serine/threonine protein kinases believed to mediate proliferation and apoptosis via the extracellular signal-regulated kinases (ERK1/2) signaling pathway. Macrophage migration inhibitory factor (MIF) and epidermal growth factor (EGF) are activators of this pathway and are elevated in the serum of patients with psoriasis compared with healthy controls. Studies on COS-7 cell cultures have shown that protein phosphatase 2Cδ (PP2Cδ) decreases the activity of RSK2 following EGF stimulation. We therefore hypothesize that PP2Cδ regulates RSK2 activity in psoriasis. Methods In paired biopsies from nonlesional (NL) and lesional (L) skins, we analyzed the level of RSK1, 2 phosphorylation and the expression of PP2Cδ isoforms, integrin-linked kinase-associated serine/threonine phosphatase (ILKAP) and wild-type p53-induced phosphatase 1 (Wip1) by Western blotting, immunofluorescence and coimmunoprecipitation with monoclonal antibody for RSK2. The induction of Wip1 by MIF or EGF was studied in cultured normal human keratinocytes. Results The protein level of RSK1, 2 phosphorylated at T573/T577 was significantly increased in L compared with NL psoriatic skin, while phosphorylation at S380/S386 was reduced in L compared with NL psoriatic skin when assayed by Western blotting and immunofluorescence microscopy. ILKAP expression was significantly higher in L than in NL skin, whereas Wip1 was expressed in similar amounts but showed increased coimmunoprecipitation with RSK2 in L compared with NL psoriatic skin. In cultured normal human keratinocytes stimulated with MIF, Wip1 phosphorylation and Wip1 expression were increased after 24 hours, but not when costimulated with dimethyl fumarate (DMF). The increased coimmunoprecipitation of Wip1 with RSK2 was significantly induced by EGF or MIF activation at 24 hours and could be significantly inhibited by DMF or the ERK1/2 inhibitor PD98059. Conclusion The complex formation of Wip1 with RSK2 indicates a direct interaction reducing P-RSK2 (S386) activation in L skin and indicates that Wip1 has a role in the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Mads K Rasmussen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Jakob Nielsen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Stine M Andersen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Anne H Rittig
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Claus Johansen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Borbala Gesser
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
13
|
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: 0.6] [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
|
14
|
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
|
15
|
Wang ZP, Tian Y, Lin J. Role of wild-type p53-induced phosphatase 1 in cancer. Oncol Lett 2017; 14:3893-3898. [PMID: 28959360 DOI: 10.3892/ol.2017.6685] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 12/16/2016] [Indexed: 12/23/2022] Open
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.
Collapse
Affiliation(s)
- Zhi-Peng Wang
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Ye Tian
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Jun Lin
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
16
|
Xia ZS, Wu D, Zhong W, Lu XJ, Yu T, Chen QK. Wip1 gene silencing enhances the chemosensitivity of human colon cancer cells. Oncol Lett 2017; 14:1875-1883. [PMID: 28781635 DOI: 10.3892/ol.2017.6361] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/13/2017] [Indexed: 12/31/2022] Open
Abstract
Colon cancer is one of the most common cancers in the world. Multidrug resistance is one of the main reasons for failure of therapy in patients with advanced colon cancer. In previous studies, multiple methods were investigated to reverse the multidrug resistance of colon cancer cells. However, to date, no clinical method has been identified to be satisfactory. Therefore, successful reversal of drug resistance in colon cancer cells still requires new therapeutic strategies or pharmaceuticals. Wild-type p53-induced phosphatase (Wip1), a member of the 2C type serine/threonine protein phosphatase family, is closely associated with the p53 gene, which is the most important tumor-suppressor gene. Wip1 was reported to be associated with the chemosensitivity of breast cancer cells. However, the correlation between the expression of Wip1 gene and the chemosensitivity of colon cancer cells has not been reported yet. In the present study, Wip1-811 small interfering RNA (siRNA) targeting Wip1 was investigated to reverse the multidrug resistance of colon cancer cells. The siRNA duplexes were transfected into RKO colon cancer cells. The messenger RNA (mRNA) expression of Wip1 was measured by reverse transcription-quantitative polymerase chain reaction. The protein level of Wip1 was detected by western blotting. The cell viability was measured by MTS assay. The cell apoptosis and cell cycle were analyzed by flow cytometry. Intracellular adriamycin cumulative concentration was determined using flow cytometry. Wip1-811 siRNA efficiently inhibited the expression of Wip1 at the mRNA and protein levels, and enhanced the sensitivity of RKO colon cancer cells towards chemotherapy, which was accompanied by increased cell apoptosis, following the inhibition of Wip1 gene expression. These results indicate that Wip1 gene silencing could enhance the chemosensitivity of colon cancer cells, which may provide a new potential approach for the reversal of multidrug resistance in colon cancer cells.
Collapse
Affiliation(s)
- Zhong-Sheng Xia
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Di Wu
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Wa Zhong
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xi-Ji Lu
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Tao Yu
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Qi-Kui Chen
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
17
|
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: 5.3] [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
|
18
|
Tang YL, Liu X, Gao SY, Feng H, Jiang YP, Wang SS, Yang J, Jiang J, Ma XR, Tang YJ, Chen Y, Liang XH. WIP1 stimulates migration and invasion of salivary adenoid cystic carcinoma by inducing MMP-9 and VEGF-C. Oncotarget 2016; 6:9031-44. [PMID: 25797250 PMCID: PMC4496200 DOI: 10.18632/oncotarget.3320] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/08/2015] [Indexed: 02/05/2023] Open
Abstract
The wild-type p53 induced phosphatase 1 (WIP1) is an oncogene overexpressed in a variety of human cancers. Here, we demonstrated that WIP1 silencing reduced MMP-9 and VEGF-C expression as well as migration and invasion of salivary adenoid cystic carcinoma (ACC) cells. Overexpression of MMP-9 or VEGF-C restored migration and invasion in WIP1 knockdown cells, indicating that MMP-9 and VEGF-C are downstream targets of WIP1 signaling. Levels of cyclin D1 and c-Myc, targets of Wnt/β-catenin pathway, were significantly decreased by WIP1 silencing. In addition, WIP1 expression was positively associated with metastasis and prognosis of ACC patients as well as with MMP-9 or VEGF-C in ACC tissues.
Collapse
Affiliation(s)
- Ya-ling Tang
- Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China.,State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Xin Liu
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Shi-yu Gao
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Hao Feng
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Ya-ping Jiang
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Sha-sha Wang
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Jing Yang
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Jian Jiang
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Xiang-rui Ma
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Ya-jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Yu Chen
- Department of Oral Pathology, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| | - Xin-hua Liang
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology (Sichuan University), Chengdu Sichuan 610041, People's Republic of China
| |
Collapse
|
19
|
Xu Z, Cao C, Xia H, Shi S, Hong L, Wei X, Gu D, Bian J, Liu Z, Huang W, Zhang Y, He S, Lee NPY, Chen J. Protein phosphatase magnesium-dependent 1δ is a novel tumor marker and target in hepatocellular carcinoma. Front Med 2016; 10:52-60. [PMID: 26809466 DOI: 10.1007/s11684-016-0433-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 12/10/2015] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is a lethal liver malignancy worldwide. In this study, we reported that protein phosphatase magnesium-dependent 1δ (PPM1D) was highly expressed in the majority of HCC cases (approximately 59%) and significantly associated with high serum α-fetoprotein (AFP) level (P = 0.044). Kaplan- Meier and Cox regression data indicated that PPM1D overexpression was an independent predictor of HCCspecific overall survival (HR, 2.799; 95% CI, 1.346-5.818, P = 0.006). Overexpressing PPM1D promoted cell viability and invasion, whereas RNA interference-mediated knockdown of PPM1D inhibited proliferation, invasion, and migration of cultured HCC cells. In addition, PPM1D suppression by small interfering RNA decreased the tumorigenicity of HCC cells in vivo. Overall, results suggest that PPM1D is a potential prognostic marker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Zhi Xu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Chunxiang Cao
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Haiyan Xia
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Shujing Shi
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Lingzhi Hong
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaowei Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Dongying Gu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Jianmin Bian
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Zijun Liu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wenbin Huang
- Department of Pathology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yixin Zhang
- Department of General Surgery, Nantong Tumor Hospital, Nantong, 226361, China
| | - Song He
- Department of Pathology, Nantong Tumor Hospital, Nantong, 226361, China
| | - Nikki Pui-Yue Lee
- Department of Surgery, Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jinfei Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
20
|
Liu C, Wang S, Zhu S, Wang H, Gu J, Gui Z, Jing J, Hou X, Shao Y. MAP3K1-targeting therapeutic artificial miRNA suppresses the growth and invasion of breast cancer in vivo and in vitro. SPRINGERPLUS 2016; 5:11. [PMID: 26759750 PMCID: PMC4700027 DOI: 10.1186/s40064-015-1597-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 12/07/2015] [Indexed: 01/11/2023]
Abstract
Recent investigations have highlighted that therapeutic artificial microRNAs could be promising candidates for cancer therapy through the modulation of tumor promoter or suppressor. MEK kinase 1 (MEKK1) is expressed by mitogen-activated kinase kinase kinase 1 (MAP3K1), an important kinase that links Ras activation to MAPK signaling. In the present study, we showed that synthetic MAP3K1-targeting artificial miRNA may provide considerable beneficial effects in the prevention of breast cancer growth and metastasis. We showed that MEKK1 was highly expressed in human breast cancer specimens, compared with adjacent normal tissues. Using a miRNA-expressing lentivirus system, we delivered a artificial miRNA (Map3k1 amiRNA) that targets MAP3K1 into 4T1 breast cancer cells and investigated the impact of MAP3K1-targeting miRNA on the growth and invasive behavior of breast cancer in vitro and in vivo. We found that overexpression of Map3k1 amiRNA led to impaired activities of p-ERK and p-p38. In addition, Map3k1 amiRNA induced marked proliferative impairment and invasive attenuation in breast cancer cells. However, Map3k1 amiRNA did not have evident influence on the apoptotic response of 4T1 cells. Moreover, using in vivo nude mice model, we identified that Map3k1 amiRNA attenuated tumor growth and lung metastasis of breast cancer cells. Taken together, our findings explicitly indicated that MEKK1 exerted important oncogenic property in breast cancer development, and MAP3K1-targeting artificial miRNA may provide promising therapeutic effects in the treatment of breast cancer.
Collapse
Affiliation(s)
- Chun Liu
- Laboratory animal center of Nantong University, 19 Qixiu Road, Nantong, Jiangsu People's Republic of China
| | - Shengjie Wang
- Kangda College of Nanjing Medical University, 88 Chunhui Road, Lianyungang, Jiangsu People's Republic of China
| | - Shunxing Zhu
- Laboratory animal center of Nantong University, 19 Qixiu Road, Nantong, Jiangsu People's Republic of China
| | - Haifeng Wang
- Kangda College of Nanjing Medical University, 88 Chunhui Road, Lianyungang, Jiangsu People's Republic of China
| | - Jiayi Gu
- Laboratory animal center of Nantong University, 19 Qixiu Road, Nantong, Jiangsu People's Republic of China
| | - Zeping Gui
- Laboratory animal center of Nantong University, 19 Qixiu Road, Nantong, Jiangsu People's Republic of China
| | - Jin Jing
- Laboratory animal center of Nantong University, 19 Qixiu Road, Nantong, Jiangsu People's Republic of China
| | - Xiaofan Hou
- Laboratory animal center of Nantong University, 19 Qixiu Road, Nantong, Jiangsu People's Republic of China
| | - Yixiang Shao
- Laboratory animal center of Nantong University, 19 Qixiu Road, Nantong, Jiangsu People's Republic of China
| |
Collapse
|
21
|
Leoni A, Locatelli A, Morigi R, Rambaldi M. 2-Indolinone a versatile scaffold for treatment of cancer: a patent review (2008-2014). Expert Opin Ther Pat 2015; 26:149-73. [PMID: 26561198 DOI: 10.1517/13543776.2016.1118059] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION 2-Indolinone is a well-known aromatic heterocyclic organic compound. A lot of work has been done on this bicyclic structure by academic and company researchers to synthesize compounds directed to a plethora of molecular targets in order to discover new drug leads. This review presents up-to-date information in the field of cancer therapy research based on this small building block. AREAS COVERED The present review gives an account of the recent patent literature (2008-2014) describing the discovery of 2-indolinone derivatives with selected therapeutic activities. In this period, a large amount of patents were published on this topic. We have limited the analysis to 37 patents on 2-indolinone derivatives having potential clinical application as chemotherapeutic agents. In this review, the therapeutic applications of 2-indolinone derivatives for the treatment of cancer reported in international patents have been discussed. EXPERT OPINION 2-Indolinone is the scaffold of the compounds considered from a medicinal chemistry perspective. Many of them have been developed and marketed for therapeutic use. In cancer chemotherapy, progress has been made in designing selective 2-indolinone derivatives. Some of them show preclinical efficacy. However, 2-indolinone has not exhausted all of its potential in the development of new compounds for clinical applications and remains a great tool for future research.
Collapse
Affiliation(s)
- Alberto Leoni
- a Dipartimento di Farmacia e Biotecnologie , Università degli Studi di Bologna , Bologna , Italy
| | - Alessandra Locatelli
- a Dipartimento di Farmacia e Biotecnologie , Università degli Studi di Bologna , Bologna , Italy
| | - Rita Morigi
- a Dipartimento di Farmacia e Biotecnologie , Università degli Studi di Bologna , Bologna , Italy
| | - Mirella Rambaldi
- a Dipartimento di Farmacia e Biotecnologie , Università degli Studi di Bologna , Bologna , Italy
| |
Collapse
|
22
|
Mice deficient for wild-type p53-induced phosphatase 1 display elevated anxiety- and depression-like behaviors. Neuroscience 2015; 293:12-22. [DOI: 10.1016/j.neuroscience.2015.02.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/13/2015] [Accepted: 02/19/2015] [Indexed: 02/02/2023]
|
23
|
Yang H, Gao XY, Li P, Jiang TS. PPM1D overexpression predicts poor prognosis in non-small cell lung cancer. Tumour Biol 2014; 36:2179-84. [PMID: 25412952 DOI: 10.1007/s13277-014-2828-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 11/06/2014] [Indexed: 12/30/2022] Open
Abstract
It has been reported that protein phosphatase, Mg(2+)/Mn(2+) dependent, 1D (PPM1D) plays an important role in cancer tumorigenesis. However, the clinical and functional significance of PPM1D expression has not been characterized previously in non-small cell lung cancer (NSCLC). The purpose of this study was to assess PPM1D expression and to explore its contribution to NSCLC. We examined PPM1D messenger RNA (mRNA) expression in 53 NSCLC tissues and matched adjacent noncancerous tissues by quantitative reverse transcription PCR (qRT-PCR). Furthermore, the PPM1D protein expression was analyzed by immunohistochemistry in 157 NSCLC samples. The relationship between PPM1D expression and clinicopathological features was analyzed by appropriate statistics. Kaplan-Meier analysis and Cox proportional hazards regression models were used to investigate the correlation between PPM1D expression and prognosis of NSCLC patients. The relative mRNA expression of PPM1D was significantly elevated in NSCLC tissues as compared with adjacent noncancerous tissues (P < 0.001). The high expression of PPM1D in NSCLC tissues was significantly correlated with tumor grade (P = 0.006), tumor size (P = 0.017), clinical stage (P = 0.001), and lymph node metastases (P = 0.002). Kaplan-Meier survival analysis revealed that high PPM1D expression correlated with poor prognosis of NSCLC patients (P < 0.001). Multivariate analysis showed that PPM1D expression was an independent prognostic marker for overall survival of NSCLC patients. In conclusion, PPM1D plays an important role in the progression of NSCLC. PPM1D may potentially be used as an independent biomarker for the prognostic evaluation of NSCLC.
Collapse
Affiliation(s)
- Hua Yang
- Respiratory Department, Yantai Hospital of Traditional Chinese Medicine, No. 39, Xingfu Road, Zhifu District, Yantai, 264002, China
| | | | | | | |
Collapse
|
24
|
Wip1 phosphatase in breast cancer. Oncogene 2014; 34:4429-38. [PMID: 25381821 DOI: 10.1038/onc.2014.375] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/02/2014] [Accepted: 10/05/2014] [Indexed: 12/13/2022]
Abstract
Understanding the factors contributing to tumor initiation, progression and evolution is of paramount significance. Among them, wild-type p53-induced phosphatase 1 (Wip1) is emerging as an important oncogene by virtue of its negative control on several key tumor suppressor pathways. Originally discovered as a p53-regulated gene, Wip1 has been subsequently found amplified and more recently mutated in a significant fraction of human cancers including breast tumors. Recent development in the field further uncovered the utility of anti-Wip1-directed therapies in delaying tumor onset or in reducing the tumor burden. Furthermore, Wip1 could be an important factor that contributes to tumor heterogeneity, suggesting that its inhibition may decrease the rate of cancer evolution. These effects depend on several signaling pathways modulated by Wip1 phosphatase in a spatial and temporal manner. In this review we discuss the recent development in understanding how Wip1 contributes to tumorigenesis with its relevance to breast cancer.
Collapse
|
25
|
Yang D, Zhang H, Hu X, Xin S, Duan Z. Abnormality of pl6/p38MAPK/p53/Wipl pathway in papillary thyroid cancer. Gland Surg 2014; 1:33-8. [PMID: 25083425 DOI: 10.3978/j.issn.2227-684x.2012.04.01] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 03/31/2012] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To investigate the expression of the pl6/p38MAPK/p53/Wipl pathway in patients with papillary thyroid cancer (PTC) and its clinical significance. METHODS The protein expressions of Wipl, p53, p38MAPK, and p16 in 70 cases of PTC tissues and 20 cases of normal thyroid tissues were detected by immunohistochemical staining. The correlations of Wipl protein high-expression with p53, p38MAPK, and pl6 protein expressions were analyzed. RESULTS The high-expression rate of Wipl protein in the PTC tissue was 64.3% (45/70), which was significantly different than that in normal thyroid tissue (0/20) (P<0.01). There were no significant differences between the paired groups in terms of age, gender, tumor size, and lymph node metastasis (all P>0.05). The Wipl protein high-expression was negatively correlated with the expressions of p38MAPK, p53 and pl6 (r value was -0.620, 0.356 and 0.550, respectively, and all P<0.01). CONCLUSIONS The pl6/p38MAPK/p53/Wipl pathway is abnormal in PTC, and this abnormality may possibly be associated with the aberrantly up-regulated Wipl, which can induce inhibition of p38MAPK, p53 and pi6.
Collapse
Affiliation(s)
- Dehua Yang
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Hao Zhang
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Xinhua Hu
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Shijie Xin
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Zhiquan Duan
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital, China Medical University, Shenyang 110001, China
| |
Collapse
|
26
|
Peng TS, He YH, Nie T, Hu XD, Lu HY, Yi J, Shuai YF, Luo M. PPM1D is a prognostic marker and therapeutic target in colorectal cancer. Exp Ther Med 2014; 8:430-434. [PMID: 25009596 PMCID: PMC4079395 DOI: 10.3892/etm.2014.1762] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 04/29/2014] [Indexed: 12/11/2022] Open
Abstract
Protein phosphatase, Mg2+/Mn2+ dependent, 1D (PPM1D) has been associated with carcinogenesis. The present study investigated PPM1D expression as a potential biomarker in colorectal cancer (CRC). PPM1D expression was assessed using immunohistochemistry in 368 patients with CRC. The correlation between PPM1D expression, clinicopathological features and prognosis was analyzed. PPM1D small interfering (si)RNA-induced PPM1D silencing was performed in CRC cell lines to assess the effect of PPM1D on tumor cell proliferation and invasion in vitro. A total of 68.48% (252/368) of the CRC samples displayed high PPM1D expression. By contrast, only 9.24% (34/368) of the matched non-cancerous tissue samples exhibited high PPM1D expression. High PPM1D expression was correlated with node metastasis (P=0.0024), distant metastasis (P<0.001) and TNM stage (P=0.0016). Kaplan-Meier survival analysis revealed that patients with low PPM1D expression had significantly longer survival than those with high PPM1D expression (P=0.012). Moreover, multivariate analyses demonstrated that high PPM1D expression was an independent prognostic factor for overall survival (hazard ratio = 0.24; 95% confidence interval, 0.13–0.86; P=0.004). Furthermore, PPM1D gene silencing was found to significantly reduce the proliferation and invasion of CRC cells in vitro. These findings suggest a role for PPM1D as a prognostic marker and potential therapeutic target in CRC.
Collapse
Affiliation(s)
- Tian-Shu Peng
- Department of Anorectal Disease, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410005, P.R. China
| | - Yong-Heng He
- Department of Anorectal Disease, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410005, P.R. China
| | - Tian Nie
- Department of Blood and Oncology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Xiang-Dang Hu
- Department of Anorectal Disease, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410005, P.R. China
| | - Hai-Yan Lu
- Department of Anorectal Disease, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410005, P.R. China
| | - Jian Yi
- Department of Blood and Oncology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Yun-Fei Shuai
- Department of Blood and Oncology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Min Luo
- Department of Anorectal Disease, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
27
|
WIP1 regulates the proliferation and invasion of nasopharyngeal carcinoma in vitro. Tumour Biol 2014; 35:7651-7. [PMID: 24801909 DOI: 10.1007/s13277-014-2034-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/28/2014] [Indexed: 01/07/2023] Open
Abstract
Wild-type p53-induced phosphatase (WIP1) is overexpressed and functionally altered in multiple human malignancies. The present study investigated its abnormal expression and dysfunctions in nasopharyngeal carcinoma (NPC) in vitro. Here, analysis of WIP1 mRNA and protein in human NPC tissues revealed that both WIP1 messenger RNA (mRNA) and protein were elevated and were correlated with NPC clinical stage and metastasis in patients. In vitro experiments further showed that WIP1 inhibition led to a decrease in the proliferative ability of NPC CNE-2 and 5-8F cells accompanied by cell cycle arrest and increased apoptosis. In addition, WIP1 knockdown inhibited the invasiveness of CNE-2 and 5-8F cells and was associated with the down-regulation of the expression of matrix metallopeptidase 9 (MMP-9) mRNA and protein. Taken together, our data demonstrate that WIP1 regulates the proliferation and invasiveness of NPC cells in vitro, and this may be correlated with its modulation of MMP-9 expression, cell cycle progression and apoptosis. WIP1 functioned as a potential therapeutic target in NPC management.
Collapse
|
28
|
Sasaki MS, Tachibana A, Takeda S. Cancer risk at low doses of ionizing radiation: artificial neural networks inference from atomic bomb survivors. JOURNAL OF RADIATION RESEARCH 2014; 55:391-406. [PMID: 24366315 PMCID: PMC4014156 DOI: 10.1093/jrr/rrt133] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 10/20/2013] [Accepted: 10/22/2013] [Indexed: 06/03/2023]
Abstract
Cancer risk at low doses of ionizing radiation remains poorly defined because of ambiguity in the quantitative link to doses below 0.2 Sv in atomic bomb survivors in Hiroshima and Nagasaki arising from limitations in the statistical power and information available on overall radiation dose. To deal with these difficulties, a novel nonparametric statistics based on the 'integrate-and-fire' algorithm of artificial neural networks was developed and tested in cancer databases established by the Radiation Effects Research Foundation. The analysis revealed unique features at low doses that could not be accounted for by nominal exposure dose, including (i) the presence of a threshold that varied with organ, gender and age at exposure, and (ii) a small but significant bumping increase in cancer risk at low doses in Nagasaki that probably reflects internal exposure to (239)Pu. The threshold was distinct from the canonical definition of zero effect in that it was manifested as negative excess relative risk, or suppression of background cancer rates. Such a unique tissue response at low doses of radiation exposure has been implicated in the context of the molecular basis of radiation-environment interplay in favor of recently emerging experimental evidence on DNA double-strand break repair pathway choice and its epigenetic memory by histone marking.
Collapse
Affiliation(s)
- Masao S. Sasaki
- Kyoto University, 17-12 Shironosato, Nagaokakyo-shi, Kyoto 617-0835, Japan
| | - Akira Tachibana
- Department of Biology, Faculty of Science, Ibaraki University, Bunkyo 2-1-1, Mito, Ibaraki 310-8512, Japan
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshida-konoecho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
29
|
Ma D, Zhang CJ, Chen ZL, Yang H. Prognostic value of PPM1D in 800 gastric cancer patients. Mol Med Rep 2014; 10:191-4. [PMID: 24788664 DOI: 10.3892/mmr.2014.2165] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 03/19/2014] [Indexed: 11/06/2022] Open
Abstract
Protein phosphatase magnesium‑dependent 1 delta (PPM1D) has recently been associated with tumor biology. However, the expression pattern and clinical significance of PPM1D in gastric cancer (GC) have yet to be elucidated. The present study aimed to investigate the clinical and prognostic significance of PPM1D in GC. PPM1D expression was assessed in 800 patients with GC using immunohistochemistry and tissue samples were divided into a PPM1D‑positive and ‑negative group. The correlation between PPM1D expression and clinicopathological parameters or prognosis was investigated. PPM1D expression was significantly higher in GC tissue than in adjacent normal tissue (48 versus 9.5%; P<0.001). PPM1D positivity was significantly correlated with nodal status, distant metastasis and vascular invasion. Survival analysis indicated that the five‑year survival rate in the PPM1D‑positive group was significantly lower than that in the PPM1D‑negative group (41 versus 72%; p=0.0012). Furthermore, the association between PPM1D positivity and survival rate was still significant following regulation of other prognostic markers in a multivariate analysis [hazard ratio (HR), 6.572; 95% confidence interval (CI), 3.108‑13.471; P=0.0018]. In conclusion, the present study suggested that PPM1D positivity is associated with GC invasion and metastasis, and proposed PPM1D positivity as an indicator of unfavorable prognosis in patients with GC.
Collapse
Affiliation(s)
- Dan Ma
- Department of General Surgery, The Second Affiliated Hospital of The Third Military Medical University, Chongqing 400037, P.R. China
| | - Chao-Jun Zhang
- Department of General Surgery, The Second Affiliated Hospital of The Third Military Medical University, Chongqing 400037, P.R. China
| | - Zu-Lin Chen
- Department of General Surgery, The Second Affiliated Hospital of The Third Military Medical University, Chongqing 400037, P.R. China
| | - Hua Yang
- Department of General Surgery, The Second Affiliated Hospital of The Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
30
|
Fu L, Chen XZ, Zhang HJ, Zhang YY, Wang LF, Xu SB, Zhang YJ, Wang MC. Mechanisms underlying suppressive effect of lutein on cell proliferation in human colon cancer cell line HT29. Shijie Huaren Xiaohua Zazhi 2013; 21:1239-1244. [DOI: 10.11569/wcjd.v21.i13.1239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the anti-proliferative effect of lutein on human colon cancer HT29 cells and to explore the possible mechanisms involved.
METHODS: HT29 cells were treated with different concentrations of lutein (20, 40, 80, 160 mg/L) for 24, 48 or 72 h. After treatment, cell proliferation was detected by SRB assay, cell cycle progression was analyzed by flow cytometry, cell apoptosis was detected by fluorescence microscopy and Hoechst 33342/PI staining, and the levels of p-ERK and p-p38 proteins were determined by Western blot.
RESULTS: Lutein treatment inhibited the proliferation of HT29 cells in a dose- and time-dependent manner. After treatment of HT29 cells with lutein (80 mg/L) for 48 h, the percentage of cells at G0/G1 phase cells increased from 58.67% to 63.23%, and with the increase in drug concentration, the percentage of cells at G0/G1 phase increased significantly. When the lutein concentration was 160 mg/L, the percentage of cells at G0/G1 phase increased to 70.81%, indicating that lutein arrested HT29 cells in G0/G1 phase. Fluorescence microscopy and Hoechst 33342/PI staining suggested that lutein induced HT29 cell apoptosis in a dose-dependent manner. Western blot analysis showed that lutein down-regulated the expression of p-ERK and up-regulated the expression of p-p38 protein in dose-dependent manner (both P < 0.01).
CONCLUSION: Lutein could significantly inhibit the proliferation of HT29 cells, induce apoptosis, and arrest cell cycle in G0/G1 phase. Down-regulation of p-ERK protein expression and up-regulation of p-p38 protein expression may be important mechanisms responsible for the apoptosis-inducing effect of lutein.
Collapse
|
31
|
Li ZT, Zhang L, Gao XZ, Jiang XH, Sun LQ. Expression and Significance of the Wip1 Proto-oncogene in Colorectal Cancer. Asian Pac J Cancer Prev 2013; 14:1975-9. [DOI: 10.7314/apjcp.2013.14.3.1975] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
32
|
Li GB, Zhang XL, Yuan L, Jiao QQ, Liu DJ, Liu J. Protein phosphatase magnesium-dependent 1δ (PPM1D) mRNA expression is a prognosis marker for hepatocellular carcinoma. PLoS One 2013; 8:e60775. [PMID: 23556002 PMCID: PMC3610683 DOI: 10.1371/journal.pone.0060775] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 03/02/2013] [Indexed: 12/28/2022] Open
Abstract
Background Protein phosphatase magnesium-dependent 1δ (PPM1D) is an oncogene, overexpressed in many solid tumors, including ovarian cancer and breast cancer. The current study examined the expression and the prognostic value of PPM1D mRNA in human hepatocellular carcinoma (HCC). Methods Total RNA was extracted from 86 HCC and paired non-cancerous liver tissues. PPM1D mRNA expression was determined by real-time quantitative reverse transcriptase-polymerase chain reaction (qPCR). Immunohistochemistry assay was used to verify the expression of ppm1d protein in the HCC and non-cancerous liver tissues. HCC patients were grouped according to PPM1D mRNA expression with the average PPM1D mRNA level in non-cancerous liver tissue samples as the cut-off. Correlations between clinicopathologic variables, overall survival and PPM1D mRNA expression were analyzed. Findings PPM1D mRNA was significantly higher in HCC than in the paired non-cancerous tissue (p<0.01). This was confirmed by ppm1d staining. 56 patients were classified as high expression group and the other 30 patients were categorized as low expression group. There were significant differences between the two groups in term of alpha-fetoprotein (α-FP) level (p<0.01), tumor size (p<0.01), TNM stage (p<0.01), recurrence incidence (p<0.01) and family history of liver cancer (p<0.01). The current study failed to find significant differences between the two groups in the following clinical characteristics: age, gender, portal vein invasion, lymphnode metastasis, hepatitis B virus (HBV) infection and alcohol intake. Survival time of high expression group was significantly shorter than that of low expression group (median survival, 13 months and 32 months, respectively, p<0.01). Conclusion Up-regulation of PPM1D mRNA was associated with progressive pathological feature and poor prognosis in HCC patients. PPM1D mRNA may serve as a prognostic marker in HCC.
Collapse
Affiliation(s)
- Guang-Bing Li
- Department of Liver Transplantation and Hepatobiliary Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | | | | | | | | | | |
Collapse
|
33
|
Zane L, Yasunaga J, Mitagami Y, Yedavalli V, Tang SW, Chen CY, Ratner L, Lu X, Jeang KT. Wip1 and p53 contribute to HTLV-1 Tax-induced tumorigenesis. Retrovirology 2012; 9:114. [PMID: 23256545 PMCID: PMC3532233 DOI: 10.1186/1742-4690-9-114] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 12/15/2012] [Indexed: 01/07/2023] Open
Abstract
Background Human T-cell Leukemia Virus type 1 (HTLV-1) infects 20 million individuals world-wide and causes Adult T-cell Leukemia/Lymphoma (ATLL), a highly aggressive T-cell cancer. ATLL is refractory to treatment with conventional chemotherapy and fewer than 10% of afflicted individuals survive more than 5 years after diagnosis. HTLV-1 encodes a viral oncoprotein, Tax, that functions in transforming virus-infected T-cells into leukemic cells. All ATLL cases are believed to have reduced p53 activity although only a minority of ATLLs have genetic mutations in their p53 gene. It has been suggested that p53 function is inactivated by the Tax protein. Results Using genetically altered mice, we report here that Tax expression does not achieve a functional equivalence of p53 inactivation as that seen with genetic mutation of p53 (i.e. a p53−/− genotype). Thus, we find statistically significant differences in tumorigenesis between Tax+p53+/+versus Tax+p53−/− mice. We also find a role contributed by the cellular Wip1 phosphatase protein in tumor formation in Tax transgenic mice. Notably, Tax+Wip1−/− mice show statistically significant reduced prevalence of tumorigenesis compared to Tax+Wip1+/+ counterparts. Conclusions Our findings provide new insights into contributions by p53 and Wip1 in the in vivo oncogenesis of Tax-induced tumors in mice.
Collapse
Affiliation(s)
- Linda Zane
- Molecular Virology Section, Laboratory of Molecular Microbiology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-0460, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Liang C, Guo E, Lu S, Wang S, Kang C, Chang L, Liu L, Zhang G, Wu Z, Zhao Z, Ma S, Wang L, Jiao BH. Over-expression of Wild-type p53-induced phosphatase 1 confers poor prognosis of patients with gliomas. Brain Res 2012; 1444:65-75. [DOI: 10.1016/j.brainres.2011.12.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 11/30/2011] [Accepted: 12/27/2011] [Indexed: 01/07/2023]
|
35
|
Park JY, Song JY, Kim HM, Han HS, Seol HS, Jang SJ, Choi J. p53-Independent expression of wild-type p53-induced phosphatase 1 (Wip1) in methylmethane sulfonate-treated cancer cell lines and human tumors. Int J Biochem Cell Biol 2012; 44:896-904. [PMID: 22405851 DOI: 10.1016/j.biocel.2012.02.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 02/08/2012] [Accepted: 02/15/2012] [Indexed: 01/03/2023]
Abstract
Wild-type p53-induced phosphatase 1 (Wip1, PPM1D) is induced by p53 in response to various stressors and dephosphorylates cellular target proteins involved in DNA repair and cell cycle checkpoint pathways. The Wip1 gene is frequently amplified or overexpressed in human cancers, promoting tumor growth by switching off major checkpoint kinases and p53. To explore wild-type p53-independent Wip1 induction, Wip1 promoter activity and its transcript level were evaluated by luciferase assay and real-time PCR, after methylmethane sulfonate (MMS) treatment in breast cancer cell lines and p53-null cell lines. Wip1 promoter activities in response to UV irradiation and various anti-cancer agents were compared between wild-type and a p53-response element (p53RE) mutated construct. Wip1 expression and its effects were examined in primary non-small cell lung cancer (NSCLC) and colon tumor cells by using Wip1-specific siRNA. MMS induced Wip1 promoter activity in Hs578T, MDA-MB-231, and SK-BR-3 cells expressing DNA binding-deficient p53 mutants. A549-E6 and HCT116 (p53(-/-)) cells retained substantial Wip1 induction. Wip1 promoter activity was reduced, but not eliminated, in cells expressing a promoter containing a mutated p53-response element. Wip1 induction was not blocked by SB202190 or SP600125. MMS increased Wip1 expression in primary non-small cell lung cancer cells expressing a p53 R175H mutant. Our data indicate that Wip1 is induced in the absence of functional p53, like p38 MAPK and JNK, as a stress response terminator.
Collapse
Affiliation(s)
- Ji-Young Park
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical center, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
36
|
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: 44] [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
|
37
|
Abstract
PURPOSE Because evasion of tumor suppression is a critical step in cancer development, cancer cells have developed a variety of mechanisms to circumvent the influence of tumor suppressive pathways. Thus, genes that negatively regulate tumor suppressors could be considered novel types of oncogenes such as Bmi-1 repressing p16Ink4a and inhibiting p53 and were found to be frequently up-regulated in a variety of cancers. p38 mitogen-activated protein kinase (MAPK), which reportedly plays a crucial role as a tumor suppressor, is activated in number of lung adenocarcinomas, which is seemingly at odds with its role as a tumor suppressor. METHODS We examined 10 lung adenocarcinomas and corresponding normal tissues and determined the expression levels of a variety of tumor suppressor proteins through real-time polymerase chain reaction and immunohistochemistry and measured p38 MAPK activity by immunoblotting or immunohistochemistry analysis. In the in vitro cellular model, p38 activation by H-Ras and consequent senescence induction was achieved through retro-viral gene transduction. Similarly, the suppression of p16Ink4a by Bmi-1 after the introduction of H-Ras was achieved through transient transfection with cationic liposome. RESULTS We detected several lung adenocarcinomas that were positive for activated p38 MAPK but evidenced reduced levels of p16Ink4a expression. The suppression of p16Ink4a occurred in parallel with an increase in Bmi-1 and/or p16Ink4a promoter hypermethylation. Consistent with these observations, the H-Ras-stimulated induction of p16Ink4a was suppressed significantly through the coexpression of Bmi-1 in vitro. DISCUSSION These results demonstrate that the suppression of p16Ink4a by either the induction of Bmi-1 or the hypermethylation of p16Ink4 may be an important step in avoiding tumor surveillance by p38 MAPK during the development of lung cancer.
Collapse
|
38
|
Wang P, Rao J, Yang H, Zhao H, Yang L. Wip1 over-expression correlated with TP53/p14(ARF) pathway disruption in human astrocytomas. J Surg Oncol 2011; 104:679-84. [PMID: 21695702 DOI: 10.1002/jso.22004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 05/31/2011] [Indexed: 12/16/2022]
Abstract
PURPOSE Wip1 over-expression inhibits p53 function and reduces selection for TP53 mutations during cancer progression. To clarify the correlation of Wip1 with TP53/p14(ARF) pathway disruption in astyrocytomas, the expression of Wip1 and TP53/p14(ARF) pathway alterations have been investigated. METHODS Tumor samples of 52 patients of astrocytomas were examined for TP53 mutations, p14(ARF) expression, and Wip1 expression. Direct sequencing of region from exons 5 to 8 of the TP53 gene was performed on the genomic DNA in each sample. The DNA methylation states of the CpG islands of the p14(ARF) gene were determined by MSP. The expression of Wip1 was analyzed by real-time quantitative PCR, Western blot, and immunohistochemical staining. RESULTS Disruption of the TP53/p14ARF pathway was detected in 57.7% of samples. Among 22 cases without TP53 and p14ARF alterations, 11 (50%) had Wip1 mRNA over-expression. In tumors with wild-type TP53 and p14ARF, Wip1 mRNA was over-expressed only 1 case out of 30 (3.3%). Higher levels of Wip1 were associated with TP53 mutations but not with lower levels of expression of p14(ARF) or aberrant promoter hypermethylation of the p14(ARF) gene. CONCLUSION Wip1 is selectively over-expressed in astyrocytomas without alterations in TP53 or p14(ARF). Wip1 may inhibit the TP53/p14(ARF) pathway.
Collapse
Affiliation(s)
- Peng Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | | | | | | | | |
Collapse
|
39
|
|
40
|
Kim JJ, Lee JS, Moon BH, Lee MO, Song SH, Li H, Fornace AJ, Cha HJ. Wip1-expressing feeder cells retain pluripotency of co-cultured mouse embryonic stem cells under leukemia inhibitory factor-deprivated condition. Arch Pharm Res 2010; 33:1253-60. [PMID: 20803129 DOI: 10.1007/s12272-010-0816-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 06/16/2010] [Accepted: 06/18/2010] [Indexed: 01/07/2023]
Abstract
The optimization of in vitro culture conditions for embryonic stem cells (ESCs) is a matter of critical importance; a prompt supply of a sufficient population of cells that retain their pluripotency capabilities must be secured in order to make possible future cell therapies. Despite a number of reports asserting that a variety of cytokines, signaling ligands, and small molecules can help in maintaining the pluripotency of ESCs, mammalian feeder cells continue to be broadly accepted as the method of choice for ESC cultures. This appears to be because mammalian feeder cells seem to produce some as-yet-unidentified factor that makes them very effective as feeder cells. In this study, we investigated wild-type p53 inducible phosphatase (Wip1), the knockdown of which increases Wnt inhibitory factor-1 expression, in its feeder functions toward mouse embryonic stem cells, lowering the effect of Wnt, one of key signaling in maintaining stemness of ESCs. For this purpose, Wip1 was stably expressed in mouse embryonic fibroblast cell line (STO) using retro-viral gene delivery system and then the function as a feeder cell was monitored either with or without leukemia inhibitory factor (LIF) in culture medium. We demonstrated that mouse embryonic stem cells grown with Wip1 expressing STO showed higher alkaline phosphatase activity and sustained Oct-4 expression level even under LIF deprivation condition compared to both control and Wip1 phosphatase activity dead mutant expressing STO. These results imply that Wip1 phosphatase activity in feeder cells is important to retain pluripotency of mouse embryonic stem cells under LIF deprivation conditions. These results indicate that genetically engineered feeder cells such as Wip1 expressing cell lines, are alternative strategy for the optimization of maintenance and expansion of mouse embryonic stem cells.
Collapse
Affiliation(s)
- Jin-Ju Kim
- Department of Biomedical Science, College of Life Science, CHA University, Pochon, 487-010, Korea.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Cha H, Lowe JM, Li H, Lee JS, Belova GI, Bulavin DV, Fornace AJ. Wip1 directly dephosphorylates gamma-H2AX and attenuates the DNA damage response. Cancer Res 2010; 70:4112-22. [PMID: 20460517 PMCID: PMC2904079 DOI: 10.1158/0008-5472.can-09-4244] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The integrity of DNA is constantly challenged throughout the life of a cell by both endogenous and exogenous stresses. A well-organized rapid damage response and proficient DNA repair, therefore, become critically important for maintaining genomic stability and cell survival. When DNA is damaged, the DNA damage response (DDR) can be initiated by alterations in chromosomal structure and histone modifications, such as the phosphorylation of the histone H2AX (the phosphorylated form is referred to as gamma-H2AX). gamma-H2AX plays a crucial role in recruiting DDR factors to damage sites for accurate DNA repair. On repair completion, gamma-H2AX must then be reverted to H2AX by dephosphorylation for attenuation of the DDR. Here, we report that the wild-type p53-induced phosphatase 1 (Wip1) phosphatase, which is often overexpressed in a variety of tumors, effectively dephosphorylates gamma-H2AX in vitro and in vivo. Ectopic expression of Wip1 significantly reduces the level of gamma-H2AX after ionizing as well as UV radiation. Forced premature dephosphorylation of gamma-H2AX by Wip1 disrupts recruitment of important DNA repair factors to damaged sites and delays DNA damage repair. Additionally, deletion of Wip1 enhances gamma-H2AX levels in cells undergoing constitutive oncogenic stress. Taken together, our studies show that Wip1 is an important mammalian phosphatase for gamma-H2AX and shows an additional mechanism for Wip1 in the tumor surveillance network.
Collapse
Affiliation(s)
- Hyukjin Cha
- Georgetown University, Dept. of Biochemistry and Molecular & Cellular Biology, Dept. of Oncology, Washington DC 20057, USA
- CHA University, Dept. Of Bio-medical Sciences, Cha Stem Cell Institute, Seoul 135-081 Korea
- John B. Little Center for the Radiation Sciences and Environmental Health, Harvard School of Public Health, Boston, MA 02115
| | - Julie M. Lowe
- Georgetown University, Dept. of Biochemistry and Molecular & Cellular Biology, Dept. of Oncology, Washington DC 20057, USA
| | - Henghong Li
- Georgetown University, Dept. of Biochemistry and Molecular & Cellular Biology, Dept. of Oncology, Washington DC 20057, USA
- John B. Little Center for the Radiation Sciences and Environmental Health, Harvard School of Public Health, Boston, MA 02115
| | - Ji-Seon Lee
- CHA University, Dept. Of Bio-medical Sciences, Cha Stem Cell Institute, Seoul 135-081 Korea
| | - Galina I. Belova
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Albert J. Fornace
- Georgetown University, Dept. of Biochemistry and Molecular & Cellular Biology, Dept. of Oncology, Washington DC 20057, USA
- John B. Little Center for the Radiation Sciences and Environmental Health, Harvard School of Public Health, Boston, MA 02115
| |
Collapse
|
42
|
Abstract
The really interesting genes (RING)-finger-containing oncoprotein, Mdm2, is a promising drug target for cancer therapy. A key Mdm2 function is to promote ubiquitylation and proteasomal-dependent degradation of the tumor suppressor protein p53. Recent reports provide novel important insights into Mdm2-mediated regulation of p53 and how the physical and functional interactions between these two proteins are regulated. Moreover, a p53-independent role of Mdm2 has recently been confirmed by genetic data. These advances and their potential implications for the development of new cancer therapeutic strategies form the focus of this review.
Collapse
Affiliation(s)
- J-C Marine
- Laboratory For Molecular Cancer Biology, VIB-UGent, Ghent B-9052, Belgium.
| | | |
Collapse
|
43
|
Song JY, Han HS, Sabapathy K, Lee BM, Yu E, Choi J. Expression of a homeostatic regulator, Wip1 (wild-type p53-induced phosphatase), is temporally induced by c-Jun and p53 in response to UV irradiation. J Biol Chem 2010; 285:9067-76. [PMID: 20093361 DOI: 10.1074/jbc.m109.070003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Wild-type p53-induced phosphatase (Wip1) is induced by p53 in response to stress, which results in the dephosphorylation of proteins (i.e. p38 MAPK, p53, and uracil DNA glycosylase) involved in DNA repair and cell cycle checkpoint pathways. p38 MAPK-p53 signaling is a unique way to induce Wip1 in response to stress. Here, we show that c-Jun directly binds to and activates the Wip1 promoter in response to UV irradiation. The binding of p53 to the promoter occurs earlier than that of c-Jun. In experiments, mutation of the p53 response element (p53RE) or c-Jun consensus sites reduced promoter activity in both non-stressed and stressed A549 cells. Overexpression of p53 significantly decreased Wip1 expression in HCT116 p53(+/+) cells but increased it in HCT116 p53(-/-) cells. Adenovirus-mediated p53 overexpression greatly decreased JNK activity. Up-regulation of Wip1 via the p38 MAPK-p53 and JNK-c-Jun pathways is specific, as demonstrated by our findings that p38 MAPK and JNK inhibitors affected the expression of the Wip1 protein, whereas an ERK inhibitor did not. c-Jun activation occurred much more quickly, and to a greater extent, in A549-E6 cells than in A549 cells, with delayed but fully induced Wip1 expression. These data indicate that Wip1 is activated via both the JNK-c-Jun and p38 MAPK-p53 signaling pathways and that temporal induction of Wip1 depends largely on the balance between c-Jun and p53, which compete for JNK binding. Moreover, our results suggest that JNK-c-Jun-mediated Wip1 induction could serve as a major signaling pathway in human tumors in response to frequent p53 mutation.
Collapse
Affiliation(s)
- Ji-young Song
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-gu, Seoul 138-736, Korea
| | | | | | | | | | | |
Collapse
|
44
|
Lee JS, Lee MO, Moon BH, Shim SH, Fornace AJ, Cha HJ. Senescent growth arrest in mesenchymal stem cells is bypassed by Wip1-mediated downregulation of intrinsic stress signaling pathways. Stem Cells 2010; 27:1963-75. [PMID: 19544416 DOI: 10.1002/stem.121] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Human mesenchymal stem cells (hMSCs) have been widely studied as a source of primary adult stem cells for cell therapy because of their multidifferentiation potential; however, the growth arrest (also known as "premature senescence") often found in hMSCs cultured in vitro has been a major obstacle to the in-depth characterization of these cells. In addition, the inability to maintain constant cell growth hampers the development of additional genetic modifications aimed at achieving desired levels of differentiation to specific tissues; however, the molecular mechanisms that govern this phenomenon remain unclear, with the exception of a few studies demonstrating that induction of p16INK4a is responsible for this senescence-like event. Here, we observed that the premature growth arrest in hMSCs occurs in parallel with the induction of p16INK4a, following abrogation of inhibitory phosphorylation of retinoblastoma protein. These stress responses were concurrent with increased formation of reactive oxygen species (ROSs) from mitochondria and increased p38 mitogen-activated protein kinase (MAPK) activity. The introduction of Wip1 (wild-type p53 inducible phosphatase-1), a well-studied stress modulator, significantly lowered p16INK4a expression and led to p38 MAPK inactivation, although it failed to affect the levels of ROSs. Moreover, the suppression of stress responses by Wip1 apparently extended the life span of hMSCs, compared with control conditions, while maintaining their multilineage differentiation potential. Based on these results, we suggest that senescent growth arrest in hMSCs may result from activation of stress signaling pathways and consequent onset of stress responses, due in part to ROS production during prolonged in vitro culture.
Collapse
Affiliation(s)
- Ji-Seon Lee
- Stem Cell Research Laboratory, CHA Stem Cell Institute, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
45
|
Germline mutations and polymorphisms in the origins of cancers in women. JOURNAL OF ONCOLOGY 2010; 2010:297671. [PMID: 20111735 PMCID: PMC2810468 DOI: 10.1155/2010/297671] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Accepted: 11/14/2009] [Indexed: 12/18/2022]
Abstract
Several female malignancies including breast, ovarian, and endometrial cancers can be characterized based on known somatic and germline mutations. Initiation and propagation of tumors reflect underlying genomic alterations such as mutations, polymorphisms, and copy number variations found in genes of multiple cellular pathways. The contributions of any single genetic variation or mutation in a population depend on its frequency and penetrance as well as tissue-specific functionality. Genome wide association studies, fluorescence in situ hybridization, comparative genomic hybridization, and candidate gene studies have enumerated genetic contributors to cancers in women. These include p53, BRCA1, BRCA2, STK11, PTEN, CHEK2, ATM, BRIP1, PALB2, FGFR2, TGFB1, MDM2, MDM4 as well as several other chromosomal loci. Based on the heterogeneity within a specific tumor type, a combination of genomic alterations defines the cancer subtype, biologic behavior, and in some cases, response to therapeutics. Consideration of tumor heterogeneity is therefore important in the critical analysis of gene associations in cancer.
Collapse
|
46
|
Lowe JM, Cha H, Yang Q, Fornace AJ. Nuclear factor-kappaB (NF-kappaB) is a novel positive transcriptional regulator of the oncogenic Wip1 phosphatase. J Biol Chem 2009; 285:5249-57. [PMID: 20007970 DOI: 10.1074/jbc.m109.034579] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The nuclear factor-kappaB (NF-kappaB) family of transcription factors plays a key role in inflammation and augments the initiation, promotion, and progression of cancer. NF-kappaB activation generally leads to transcriptional enhancement of genes important in cell survival and cell growth, which is exploited in cancer cells. In this study, we identify an additional oncogene, PPM1D, which encodes for Wip1, as a transcriptional target of NF-kappaB in breast cancer cells. Inhibition of NF-kappaB or activation of NF-kappaB resulted in decreased or increased Wip1 expression, respectively, at both the mRNA and protein levels. PPM1D promoter activity was positively regulated by NF-kappaB, and this regulation was dependent on the presence of the conserved kappaB site in the PPM1D promoter region. Chromatin immunoprecipitation analysis showed basal binding of the p65 NF-kappaB subunit to the PPM1D promoter region encompassing the kappaB site, which is enhanced after NF-kappaB activation by tumor necrosis factor-alpha. Finally, we show that Wip1 expression is induced in lipopolysaccharide-stimulated mouse splenic B-cells and is required for maximum proliferation. Taken together, these data suggest an additional mechanism by which NF-kappaB may promote tumorigenesis, support the selective use of NF-kappaB inhibitors as chemotherapeutic agents for the treatment of human cancers, and further define a function for Wip1 in inflammation.
Collapse
Affiliation(s)
- Julie M Lowe
- Department of Biochemistry and Molecular and Cellular Biology, Lombardi Comprehensive Center, Georgetown University, Washington, DC 20057, USA
| | | | | | | |
Collapse
|
47
|
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: 112] [Impact Index Per Article: 7.0] [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
|
48
|
Han HS, Yu E, Song JY, Park JY, Jang SJ, Choi J. The estrogen receptor alpha pathway induces oncogenic Wip1 phosphatase gene expression. Mol Cancer Res 2009; 7:713-23. [PMID: 19435816 DOI: 10.1158/1541-7786.mcr-08-0247] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Wild-type p53-induced phosphatase (Wip1) is a serine/threonine phosphatase induced by DNA-damaging agents. This enzyme dephosphorylates several cell cycle regulating proteins, including p53, p38 mitogen-activated protein kinase, Chk1, and Chk2, resulting in negative feedback regulation of p38-p53 signaling after damage repair. Moreover, the Wip1 gene may be amplified or overexpressed, especially in hormone-regulated organs, and Wip1 gene amplification has been correlated with poor prognosis in hormone-related malignancies, including ovarian cancers. We therefore investigated the link between estrogen signaling and Wip1 expression. We identified seven putative estrogen response elements within 3 kb of the Wip1 promoter. We also found that estradiol (E(2)) treatment produced a 3-fold increase in endogenous Wip1 mRNA and protein expression in MCF7 cells. Direct binding of estrogen receptor (ER)alpha to the Wip1 promoter after E(2) treatment was confirmed by a chromatin immunoprecipitation assay using ERalpha antibody and an electrophoretic mobility shift assay. Wip1 overexpression induced by adenovirus and E(2) facilitated the proliferation of serum-starved ZR-75-1 cells, with cell proliferation induced by overexpressed Wip1 approximately 25% higher than that induced by E(2). Wip1 phosphatase activity was essential for cell cycle progression. Wip1 stimulated the transcriptional activity of its own promoter through E(2)-ERalpha signaling. In addition, Wip1 overexpression induced Rb phosphorylation during cancer cell proliferation. These results indicate that Wip1 up-regulation is important in the pathogenesis of p53(+) and ER(+) breast cancer through the inactivation of p53 by dephosphorylation and the amplification of subsequent estrogenic effects through the E(2)-ERalpha-Wip1 pathway.
Collapse
Affiliation(s)
- Hye-Sook Han
- Department of Pathology, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-gu, Seoul 138-736, Republic of Korea
| | | | | | | | | | | |
Collapse
|
49
|
Natrajan R, Lambros MB, Rodríguez-Pinilla SM, Moreno-Bueno G, Tan DSP, Marchió C, Vatcheva R, Rayter S, Mahler-Araujo B, Fulford LG, Hungermann D, Mackay A, Grigoriadis A, Fenwick K, Tamber N, Hardisson D, Tutt A, Palacios J, Lord CJ, Buerger H, Ashworth A, Reis-Filho JS. Tiling path genomic profiling of grade 3 invasive ductal breast cancers. Clin Cancer Res 2009; 15:2711-22. [PMID: 19318498 DOI: 10.1158/1078-0432.ccr-08-1878] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE To characterize the molecular genetic profiles of grade 3 invasive ductal carcinomas of no special type using high-resolution microarray-based comparative genomic hybridization (aCGH) and to identify recurrent amplicons harboring putative therapeutic targets associated with luminal, HER-2, and basal-like tumor phenotypes. EXPERIMENTAL DESIGN Ninety-five grade 3 invasive ductal carcinomas of no special type were classified into luminal, HER-2, and basal-like subgroups using a previously validated immunohistochemical panel. Tumor samples were microdissected and subjected to aCGH using a tiling path 32K BAC array platform. Selected regions of recurrent amplification were validated by means of in situ hybridization. Expression of genes pertaining to selected amplicons was investigated using quantitative real-time PCR and gene silencing was done using previously validated short hairpin RNA constructs. RESULTS We show that basal-like and HER-2 tumors are characterized by "sawtooth" and "firestorm" genetic patterns, respectively, whereas luminal cancers were more heterogeneous. Apart from confirming known amplifications associated with basal-like (1q21, 10p, and 12p), luminal (8p12, 11q13, and 11q14), and HER-2 (17q12) cancers, we identified previously unreported recurrent amplifications associated with each molecular subgroup: 19q12 in basal-like, 1q32.1 in luminal, and 14q12 in HER-2 cancers. PPM1D gene amplification (17q23.2) was found in 20% and 8% of HER-2 and luminal cancers, respectively. Silencing of PPM1D by short hairpin RNA resulted in selective loss of viability in tumor cell lines harboring the 17q23.2 amplification. CONCLUSIONS Our results show the power of aCGH analysis in unraveling the genetic profiles of specific subgroups of cancer and for the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Rachael Natrajan
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lu X, Nguyen TA, Moon SH, Darlington Y, Sommer M, Donehower LA. The type 2C phosphatase Wip1: an oncogenic regulator of tumor suppressor and DNA damage response pathways. Cancer Metastasis Rev 2008; 27:123-35. [PMID: 18265945 PMCID: PMC2362138 DOI: 10.1007/s10555-008-9127-x] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Wild-type p53-induced phosphatase 1, Wip1 (or PPM1D), is unusual in that it is a serine/threonine phosphatase with oncogenic activity. A member of the type 2C phosphatases (PP2Cδ), Wip1 has been shown to be amplified and overexpressed in multiple human cancer types, including breast and ovarian carcinomas. In rodent primary fibroblast transformation assays, Wip1 cooperates with known oncogenes to induce transformed foci. The recent identification of target proteins that are dephosphorylated by Wip1 has provided mechanistic insights into its oncogenic functions. Wip1 acts as a homeostatic regulator of the DNA damage response by dephosphorylating proteins that are substrates of both ATM and ATR, important DNA damage sensor kinases. Wip1 also suppresses the activity of multiple tumor suppressors, including p53, ATM, p16INK4a and ARF. We present evidence that the suppression of p53, p38 MAP kinase, and ATM/ATR signaling pathways by Wip1 are important components of its oncogenicity when it is amplified and overexpressed in human cancers.
Collapse
Affiliation(s)
- Xiongbin Lu
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|