1
|
Meller A, de Oliveira S, Davtyan A, Abramyan T, Bowman GR, van den Bedem H. Discovery of a cryptic pocket in the AI-predicted structure of PPM1D phosphatase explains the binding site and potency of its allosteric inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533829. [PMID: 36993233 PMCID: PMC10055338 DOI: 10.1101/2023.03.22.533829] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Virtual screening is a widely used tool for drug discovery, but its predictive power can vary dramatically depending on how much structural data is available. In the best case, crystal structures of a ligand-bound protein can help find more potent ligands. However, virtual screens tend to be less predictive when only ligand-free crystal structures are available, and even less predictive if a homology model or other predicted structure must be used. Here, we explore the possibility that this situation can be improved by better accounting for protein dynamics, as simulations started from a single structure have a reasonable chance of sampling nearby structures that are more compatible with ligand binding. As a specific example, we consider the cancer drug target PPM1D/Wip1 phosphatase, a protein that lacks crystal structures. High-throughput screens have led to the discovery of several allosteric inhibitors of PPM1D, but their binding mode remains unknown. To enable further drug discovery efforts, we assessed the predictive power of an AlphaFold-predicted structure of PPM1D and a Markov state model (MSM) built from molecular dynamics simulations initiated from that structure. Our simulations reveal a cryptic pocket at the interface between two important structural elements, the flap and hinge regions. Using deep learning to predict the pose quality of each docked compound for the active site and cryptic pocket suggests that the inhibitors strongly prefer binding to the cryptic pocket, consistent with their allosteric effect. The predicted affinities for the dynamically uncovered cryptic pocket also recapitulate the relative potencies of the compounds (τ b =0.70) better than the predicted affinities for the static AlphaFold-predicted structure (τ b =0.42). Taken together, these results suggest that targeting the cryptic pocket is a good strategy for drugging PPM1D and, more generally, that conformations selected from simulation can improve virtual screening when limited structural data is available.
Collapse
Affiliation(s)
- Artur Meller
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110
- Medical Scientist Training Program, Washington University in St. Louis, 660 S Euclid Ave., St. Louis, MO, 63110
| | - Saulo de Oliveira
- Atomwise, Inc., 717 Market Street, Suite 800, San Francisco, California 94103
| | - Aram Davtyan
- Atomwise, Inc., 717 Market Street, Suite 800, San Francisco, California 94103
| | - Tigran Abramyan
- Atomwise, Inc., 717 Market Street, Suite 800, San Francisco, California 94103
| | - Gregory R. Bowman
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104
| | - Henry van den Bedem
- Atomwise, Inc., 717 Market Street, Suite 800, San Francisco, California 94103
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
2
|
Meller A, De Oliveira S, Davtyan A, Abramyan T, Bowman GR, van den Bedem H. Discovery of a cryptic pocket in the AI-predicted structure of PPM1D phosphatase explains the binding site and potency of its allosteric inhibitors. Front Mol Biosci 2023; 10:1171143. [PMID: 37143823 PMCID: PMC10151774 DOI: 10.3389/fmolb.2023.1171143] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/07/2023] [Indexed: 05/06/2023] Open
Abstract
Virtual screening is a widely used tool for drug discovery, but its predictive power can vary dramatically depending on how much structural data is available. In the best case, crystal structures of a ligand-bound protein can help find more potent ligands. However, virtual screens tend to be less predictive when only ligand-free crystal structures are available, and even less predictive if a homology model or other predicted structure must be used. Here, we explore the possibility that this situation can be improved by better accounting for protein dynamics, as simulations started from a single structure have a reasonable chance of sampling nearby structures that are more compatible with ligand binding. As a specific example, we consider the cancer drug target PPM1D/Wip1 phosphatase, a protein that lacks crystal structures. High-throughput screens have led to the discovery of several allosteric inhibitors of PPM1D, but their binding mode remains unknown. To enable further drug discovery efforts, we assessed the predictive power of an AlphaFold-predicted structure of PPM1D and a Markov state model (MSM) built from molecular dynamics simulations initiated from that structure. Our simulations reveal a cryptic pocket at the interface between two important structural elements, the flap and hinge regions. Using deep learning to predict the pose quality of each docked compound for the active site and cryptic pocket suggests that the inhibitors strongly prefer binding to the cryptic pocket, consistent with their allosteric effect. The predicted affinities for the dynamically uncovered cryptic pocket also recapitulate the relative potencies of the compounds (τb = 0.70) better than the predicted affinities for the static AlphaFold-predicted structure (τb = 0.42). Taken together, these results suggest that targeting the cryptic pocket is a good strategy for drugging PPM1D and, more generally, that conformations selected from simulation can improve virtual screening when limited structural data is available.
Collapse
Affiliation(s)
- Artur Meller
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, United States
- Medical Scientist Training Program, Washington University in St. Louis, St. Louis, MO, United States
| | | | - Aram Davtyan
- Atomwise, Inc., San Francisco, CA, United States
| | | | - Gregory R. Bowman
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Gregory R. Bowman, ; Henry van den Bedem,
| | - Henry van den Bedem
- Atomwise, Inc., San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, United States
- *Correspondence: Gregory R. Bowman, ; Henry van den Bedem,
| |
Collapse
|
3
|
Marabitti V, Giansanti M, De Mitri F, Gatto F, Mastronuzzi A, Nazio F. Pathological implications of metabolic reprogramming and its therapeutic potential in medulloblastoma. Front Cell Dev Biol 2022; 10:1007641. [PMID: 36340043 PMCID: PMC9627342 DOI: 10.3389/fcell.2022.1007641] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 10/05/2022] [Indexed: 07/30/2023] Open
Abstract
Tumor-specific alterations in metabolism have been recognized to sustain the production of ATP and macromolecules needed for cell growth, division and survival in many cancer types. However, metabolic heterogeneity poses a challenge for the establishment of effective anticancer therapies that exploit metabolic vulnerabilities. Medulloblastoma (MB) is one of the most heterogeneous malignant pediatric brain tumors, divided into four molecular subgroups (Wingless, Sonic Hedgehog, Group 3 and Group 4). Recent progresses in genomics, single-cell sequencing, and novel tumor models have updated the classification and stratification of MB, highlighting the complex intratumoral cellular diversity of this cancer. In this review, we emphasize the mechanisms through which MB cells rewire their metabolism and energy production networks to support and empower rapid growth, survival under stressful conditions, invasion, metastasis, and resistance to therapy. Additionally, we discuss the potential clinical benefits of currently available drugs that could target energy metabolism to suppress MB progression and increase the efficacy of the current MB therapies.
Collapse
Affiliation(s)
- Veronica Marabitti
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Manuela Giansanti
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Francesca De Mitri
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Francesca Gatto
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Angela Mastronuzzi
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Francesca Nazio
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
4
|
Clausse V, Fang Y, Tao D, Tagad HD, Sun H, Wang Y, Karavadhi S, Lane K, Shi ZD, Vasalatiy O, LeClair CA, Eells R, Shen M, Patnaik S, Appella E, Coussens NP, Hall MD, Appella DH. Discovery of Novel Small-Molecule Scaffolds for the Inhibition and Activation of WIP1 Phosphatase from a RapidFire Mass Spectrometry High-Throughput Screen. ACS Pharmacol Transl Sci 2022; 5:993-1006. [PMID: 36268125 PMCID: PMC9578142 DOI: 10.1021/acsptsci.2c00147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Indexed: 11/28/2022]
Abstract
Wild-type P53-induced phosphatase 1 (WIP1), also known as PPM1D or PP2Cδ, is a serine/threonine protein phosphatase induced by P53 after genotoxic stress. WIP1 inhibition has been proposed as a therapeutic strategy for P53 wild-type cancers in which it is overexpressed, but this approach would be ineffective in P53-negative cancers. Furthermore, there are several cancers with mutated P53 where WIP1 acts as a tumor suppressor. Therefore, activating WIP1 phosphatase might also be a therapeutic strategy, depending on the P53 status. To date, no specific, potent WIP1 inhibitors with appropriate pharmacokinetic properties have been reported, nor have WIP1-specific activators. Here, we report the discovery of new WIP1 modulators from a high-throughput screen (HTS) using previously described orthogonal biochemical assays suitable for identifying both inhibitors and activators. The primary HTS was performed against a library of 102 277 compounds at a single concentration using a RapidFire mass spectrometry assay. Hits were further evaluated over a range of 11 concentrations with both the RapidFire MS assay and an orthogonal fluorescence-based assay. Further biophysical, biochemical, and cell-based studies of confirmed hits revealed a WIP1 activator and two inhibitors, one competitive and one uncompetitive. These new scaffolds are prime candidates for optimization which might enable inhibitors with improved pharmacokinetics and a first-in-class WIP1 activator.
Collapse
Affiliation(s)
- Victor Clausse
- Synthetic
Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yuhong Fang
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Dingyin Tao
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Harichandra D. Tagad
- Laboratory
of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hongmao Sun
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Yuhong Wang
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Surendra Karavadhi
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Kelly Lane
- Chemistry
and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Zhen-Dan Shi
- Chemistry
and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Olga Vasalatiy
- Chemistry
and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Christopher A. LeClair
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Rebecca Eells
- Reaction
Biology Corporation, 1 Great Valley Parkway, Suite 2, Malvern, Pennsylvania 19355, United States
| | - Min Shen
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Samarjit Patnaik
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Ettore Appella
- Laboratory
of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nathan P. Coussens
- Molecular
Pharmacology Laboratories, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States
| | - Matthew D. Hall
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Daniel H. Appella
- Synthetic
Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
5
|
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: 1.5] [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
|
6
|
McSwain LF, Parwani KK, Shahab SW, Hambardzumyan D, MacDonald TJ, Spangle JM, Kenney AM. Medulloblastoma and the DNA Damage Response. Front Oncol 2022; 12:903830. [PMID: 35747808 PMCID: PMC9209741 DOI: 10.3389/fonc.2022.903830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/10/2022] [Indexed: 12/04/2022] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children with standard of care consisting of surgery, radiation, and chemotherapy. Recent molecular profiling led to the identification of four molecularly distinct MB subgroups – Wingless (WNT), Sonic Hedgehog (SHH), Group 3, and Group 4. Despite genomic MB characterization and subsequent tumor stratification, clinical treatment paradigms are still largely driven by histology, degree of surgical resection, and presence or absence of metastasis rather than molecular profile. Patients usually undergo resection of their tumor followed by craniospinal radiation (CSI) and a 6 month to one-year multi-agent chemotherapeutic regimen. While there is clearly a need for development of targeted agents specific to the molecular alterations of each patient, targeting proteins responsible for DNA damage repair could have a broader impact regardless of molecular subgrouping. DNA damage response (DDR) protein inhibitors have recently emerged as targeted agents with potent activity as monotherapy or in combination in different cancers. Here we discuss the molecular underpinnings of genomic instability in MB and potential avenues for exploitation through DNA damage response inhibition.
Collapse
Affiliation(s)
- Leon F. McSwain
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Kiran K. Parwani
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
- Department of Radiation Oncology, Emory University, Atlanta, GA, United States
| | - Shubin W. Shahab
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Dolores Hambardzumyan
- Departments of Neurosurgery and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tobey J. MacDonald
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Jennifer M. Spangle
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
- Department of Radiation Oncology, Emory University, Atlanta, GA, United States
| | - Anna Marie Kenney
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- *Correspondence: Anna Marie Kenney,
| |
Collapse
|
7
|
Chen M, Wang W, Hu S, Tong Y, Li Y, Wei Q, Yu L, Zhu L, Zhu Y, Liu L, Ju Z, Wang X, Jin H, Feng L. Co-targeting WIP1 and PARP induces synthetic lethality in hepatocellular carcinoma. Cell Commun Signal 2022; 20:39. [PMID: 35346236 PMCID: PMC8962187 DOI: 10.1186/s12964-022-00850-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most fatal cancers. Due to limited strategies for effective treatments, patients with advanced HCC have a very poor prognosis. This study aims to identify new insights in HCC to develop novel strategies for HCC management. Methods The role of WIP1 (wild type p53 induced protein phosphatase1) in HCC was analyzed in HCC cells, xenograft model, DEN (Diethylnitrosamine) induced mice liver cancer model with WIP1 knockout mice, and TCGA database. DNA damage was evaluated by Gene Set Enrichment Analysis, western blotting, comet assay, and Immunofluorescence. Results High expression of WIP1 is associated with the poor prognosis of patients with HCC. Genetically and chemically suppression of WIP1 drastically reduced HCC cell proliferation. Besides, WIP1 knockout retarded DEN induced mice hepato-carcinogenesis. Mechanically, WIP1 inhibition induced DNA damage by increasing H2AX phosphorylation (γH2AX). Therefore, suppression of WIP1 and PARP induced synthetic lethality in HCC in vitro and in vivo by augmenting DNA damage. Conclusion WIP1 plays an oncogenic effect in HCC development, and targeting WIP1-dependent DNA damage repair alone or in combination with PARP inhibition might be a reasonable strategy for HCC management. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00850-2.
Collapse
|
8
|
Milosevic J, Treis D, Fransson S, Gallo-Oller G, Sveinbjörnsson B, Eissler N, Tanino K, Sakaguchi K, Martinsson T, Wickström M, Kogner P, Johnsen JI. PPM1D Is a Therapeutic Target in Childhood Neural Tumors. Cancers (Basel) 2021; 13:cancers13236042. [PMID: 34885154 PMCID: PMC8657050 DOI: 10.3390/cancers13236042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/20/2021] [Accepted: 11/25/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Medulloblastoma and neuroblastoma are childhood tumors of the central nervous system or the peripheral nervous system, respectively. These are the most common and deadly tumors of childhood. A common genetic feature of medulloblastoma and neuroblastoma is frequent segmental gain or amplification of chromosome 17q. Located on chromosome 17q23.2 is PPM1D which encodes WIP1, a phosphatase that acts as a regulator of p53 and DNA repair. Overexpression of WIP1 correlates with poor patient prognosis. We investigated the effects of genetic or pharmacologic inhibition of WIP1 activity and found that medulloblastoma and neuroblastoma cells were strongly dependent on WIP1 expression for survival. We also tested a number of small molecule inhibitors of WIP1 and show that SL-176 was the most effective compound suppressing the growth of medulloblastoma and neuroblastoma in vitro and in vivo. Abstract Childhood medulloblastoma and high-risk neuroblastoma frequently present with segmental gain of chromosome 17q corresponding to aggressive tumors and poor patient prognosis. Located within the 17q-gained chromosomal segments is PPM1D at chromosome 17q23.2. PPM1D encodes a serine/threonine phosphatase, WIP1, that is a negative regulator of p53 activity as well as key proteins involved in cell cycle control, DNA repair and apoptosis. Here, we show that the level of PPM1D expression correlates with chromosome 17q gain in medulloblastoma and neuroblastoma cells, and both medulloblastoma and neuroblastoma cells are highly dependent on PPM1D expression for survival. Comparison of different inhibitors of WIP1 showed that SL-176 was the most potent compound inhibiting medulloblastoma and neuroblastoma growth and had similar or more potent effects on cell survival than the MDM2 inhibitor Nutlin-3 or the p53 activator RITA. SL-176 monotherapy significantly suppressed the growth of established medulloblastoma and neuroblastoma xenografts in nude mice. These results suggest that the development of clinically applicable compounds inhibiting the activity of WIP1 is of importance since PPM1D activating mutations, genetic gain or amplifications and/or overexpression of WIP1 are frequently detected in several different cancers.
Collapse
Affiliation(s)
- Jelena Milosevic
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.T.); (G.G.-O.); (B.S.); (N.E.) (M.W.); (P.K.)
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Correspondence: (J.M.); (J.I.J.)
| | - Diana Treis
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.T.); (G.G.-O.); (B.S.); (N.E.) (M.W.); (P.K.)
| | - Susanne Fransson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, 41345 Gothenburg, Sweden; (S.F.); (T.M.)
| | - Gabriel Gallo-Oller
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.T.); (G.G.-O.); (B.S.); (N.E.) (M.W.); (P.K.)
| | - Baldur Sveinbjörnsson
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.T.); (G.G.-O.); (B.S.); (N.E.) (M.W.); (P.K.)
| | - Nina Eissler
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.T.); (G.G.-O.); (B.S.); (N.E.) (M.W.); (P.K.)
| | - Keiji Tanino
- Laboratory of Organic Chemistry II, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan;
| | - Kazuyasu Sakaguchi
- Laboratory of Biological Chemistry, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan;
| | - Tommy Martinsson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, 41345 Gothenburg, Sweden; (S.F.); (T.M.)
| | - Malin Wickström
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.T.); (G.G.-O.); (B.S.); (N.E.) (M.W.); (P.K.)
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.T.); (G.G.-O.); (B.S.); (N.E.) (M.W.); (P.K.)
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.T.); (G.G.-O.); (B.S.); (N.E.) (M.W.); (P.K.)
- Correspondence: (J.M.); (J.I.J.)
| |
Collapse
|
9
|
High Expression of PPM1D Induces Tumors Phenotypically Similar to TP53 Loss-of-Function Mutations in Mice. Cancers (Basel) 2021; 13:cancers13215493. [PMID: 34771656 PMCID: PMC8582939 DOI: 10.3390/cancers13215493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Aberrant expression of the PPM1D gene which encodes a phosphatase called WIP1 is frequently observed in cancers of different origins. WIP1 is a negative regulator of the tumor suppressor p53. Improper inactivation of p53 results in genomic instability and can induce neoplastic transformation. We show that overexpression of PPM1D induces tumors in mice similar to cancers harboring p53 mutations. Our results suggest that PPM1D can act as an oncogenic driver by inducing genomic instability, impaired growth arrest, and apoptotic escape that can result in neoplastic transformation and malignant tumor development. Abstract PPM1D is a negative regulator of p53 and genomic aberrations resulting in increased activity of PPM1D have been observed in cancers of different origins, indicating that PPM1D has oncogenic properties. We established a transgenic mouse model overexpressing PPM1D and showed that these mice developed a wide variety of cancers. PPM1D-expressing mice developed tumors phenotypically and genetically similar to tumors in mice with dysfunctional p53. T-cell lymphoblastic lymphoma was the most frequent cancer observed in these mice (55%) followed by adenocarcinomas (24%), leukemia (12%) and other solid tumors including neuroblastoma. Characterization of T-cell lymphomas in mice overexpressing PPM1D demonstrates Pten-deletion and p53-accumulation similar to mice with p53 loss-of-function. Also, Notch1 mutations which are recurrently observed in T-cell acute lymphoblastic lymphoma (T-ALL) were frequently detected in PPM1D-transgenic mice. Hence, PPM1D acts as an oncogenic driver in connection with cellular stress, suggesting that the PPM1D gene status and expression levels should be investigated in TP53 wild-type tumors.
Collapse
|
10
|
Inhibition of the DNA damage response phosphatase PPM1D reprograms neutrophils to enhance anti-tumor immune responses. Nat Commun 2021; 12:3622. [PMID: 34131120 PMCID: PMC8206133 DOI: 10.1038/s41467-021-23330-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
PPM1D/Wip1 is a negative regulator of the tumor suppressor p53 and is overexpressed in several human solid tumors. Recent reports associate gain-of-function mutations of PPM1D in immune cells with worse outcomes for several human cancers. Here we show that mice with genetic knockout of Ppm1d or with conditional knockout of Ppm1d in the hematopoietic system, in myeloid cells, or in neutrophils all display significantly reduced growth of syngeneic melanoma or lung carcinoma tumors. Ppm1d knockout neutrophils infiltrate tumors extensively. Chemical inhibition of Wip1 in human or mouse neutrophils increases anti-tumor phenotypes, p53-dependent expression of co-stimulatory ligands, and proliferation of co-cultured cytotoxic T cells. These results suggest that inhibition of Wip1 in neutrophils enhances immune anti-tumor responses.
Collapse
|
11
|
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: 1.3] [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
|
12
|
Wang H, Pan J, Yu L, Meng L, Liu Y, Chen X. MicroRNA-16 Inhibits Glioblastoma Growth in Orthotopic Model by Targeting Cyclin D1 and WIP1. Onco Targets Ther 2020; 13:10807-10816. [PMID: 33122919 PMCID: PMC7591102 DOI: 10.2147/ott.s250369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/11/2020] [Indexed: 01/07/2023] Open
Abstract
Introduction To examine the molecular mechanism by which miRNA-16 (miR-16) suppresses glioblastoma in vitro and in vivo. Methods Gene expression of miR-16 in normal brain tissues and human glioma cell lines was examined. To characterize the functional role of miR-16 in vitro, miR-16 was ectopically expressed in U87 cells by lentiviral transduction. Expression of miR-16 downstream targets cyclin D1 and Bcl-2 in U87 was studied using Western blotting. Cell proliferation and clonogenic property were examined using CCK-8 and clone formation assay, respectively. Migration and invasiveness of U87 was studied using wound-healing assay and transwell assay, respectively. In vivo tumorigenic properties of the miR-16-transduced U87 cells were examined in an orthotopic xenograft model. Immunohistochemistry was performed to examine cyclin D1, WIP1 and CD31 expressions. Results Expression of miR-16 was reduced in glioblastoma cell lines compared to normal human brain tissues. Ectopic miR-16 expression reduced cyclin D1 and Bcl-2 in U87 cells. miR-16 also induced apoptosis, reduced cell proliferation and clone formation. Furthermore, miR-16 suppressed U87 migration in wound-healing assay and invasion across transwell membrane in vitro. In an orthotopic tumor model, overexpression of miR-16 inhibited tumor growth in vivo was accompanied with reduction in cyclin D1 and WIP1 expression in the xenografts. CD31 expression in miR-16-overexpressed xenografts was also decreased. The determined microvessel density of the miR-16 overexpression group was significantly lower than those groups treated with vehicle and empty vector. Discussion MicroRNA-16 exhibits inhibitory effects of glioblastoma. MicroRNA-16 and its downstream targets could be potential therapeutic targets for treatment of glioblastoma.
Collapse
Affiliation(s)
- Heng Wang
- Department of Gastrointestinal Surgery/Pediatric Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, People's Republic of China
| | - Jun Pan
- Department of Traditional Chinese Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, People's Republic of China
| | - Lisheng Yu
- Department of Neurosurgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Linghu Meng
- Department of Neurosurgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, People's Republic of China
| | - Yue Liu
- Department of Neurosurgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, People's Republic of China
| | - Xin Chen
- Department of Neurosurgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, People's Republic of China
| |
Collapse
|
13
|
Hat B, Jaruszewicz-Błońska J, Lipniacki T. Model-based optimization of combination protocols for irradiation-insensitive cancers. Sci Rep 2020; 10:12652. [PMID: 32724100 PMCID: PMC7387345 DOI: 10.1038/s41598-020-69380-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/19/2020] [Indexed: 01/07/2023] Open
Abstract
Alternations in the p53 regulatory network may render cancer cells resistant to the radiation-induced apoptosis. In this theoretical study we search for the best protocols combining targeted therapy with radiation to treat cancers with wild-type p53, but having downregulated expression of PTEN or overexpression of Wip1 resulting in resistance to radiation monotherapy. Instead of using the maximum tolerated dose paradigm, we exploit stochastic computational model of the p53 regulatory network to calculate apoptotic fractions for both normal and cancer cells. We consider combination protocols, with irradiations repeated every 12, 18, 24, or 36 h to find that timing between Mdm2 inhibitor delivery and irradiation significantly influences the apoptotic cell fractions. We assume that uptake of the inhibitor is higher by cancer than by normal cells and that cancer cells receive higher irradiation doses from intersecting beams. These two assumptions were found necessary for the existence of protocols inducing massive apoptosis in cancer cells without killing large fraction of normal cells neighboring tumor. The best found protocols have irradiations repeated every 24 or 36 h with two inhibitor doses per irradiation cycle, and allow to induce apoptosis in more than 95% of cancer cells, killing less than 10% of normal cells.
Collapse
Affiliation(s)
- Beata Hat
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | | | - Tomasz Lipniacki
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
14
|
Metal-dependent Ser/Thr protein phosphatase PPM family: Evolution, structures, diseases and inhibitors. Pharmacol Ther 2020; 215:107622. [PMID: 32650009 DOI: 10.1016/j.pharmthera.2020.107622] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Protein phosphatases and kinases control multiple cellular events including proliferation, differentiation, and stress responses through regulating reversible protein phosphorylation, the most important post-translational modification. Members of metal-dependent protein phosphatase (PPM) family, also known as PP2C phosphatases, are Ser/Thr phosphatases that bind manganese/magnesium ions (Mn2+/Mg2+) in their active center and function as single subunit enzymes. In mammals, there are 20 isoforms of PPM phosphatases: PPM1A, PPM1B, PPM1D, PPM1E, PPM1F, PPM1G, PPM1H, PPM1J, PPM1K, PPM1L, PPM1M, PPM1N, ILKAP, PDP1, PDP2, PHLPP1, PHLPP2, PP2D1, PPTC7, and TAB1, whereas there are only 8 in yeast. Phylogenetic analysis of the DNA sequences of vertebrate PPM isoforms revealed that they can be divided into 12 different classes: PPM1A/PPM1B/PPM1N, PPM1D, PPM1E/PPM1F, PPM1G, PPM1H/PPM1J/PPM1M, PPM1K, PPM1L, ILKAP, PDP1/PDP2, PP2D1/PHLPP1/PHLPP2, TAB1, and PPTC7. PPM-family members have a conserved catalytic core region, which contains the metal-chelating residues. The different isoforms also have isoform specific regions within their catalytic core domain and terminal domains, and these regions may be involved in substrate recognition and/or functional regulation of the phosphatases. The twenty mammalian PPM phosphatases are involved in regulating diverse cellular functions, such as cell cycle control, cell differentiation, immune responses, and cell metabolism. Mutation, overexpression, or deletion of the PPM phosphatase gene results in abnormal cellular responses, which lead to various human diseases. This review focuses on the structures and biological functions of the PPM-phosphatase family and their associated diseases. The development of specific inhibitors against the PPM phosphatase family as a therapeutic strategy will also be discussed.
Collapse
|
15
|
Akamandisa MP, Nie K, Nahta R, Hambardzumyan D, Castellino RC. Inhibition of mutant PPM1D enhances DNA damage response and growth suppressive effects of ionizing radiation in diffuse intrinsic pontine glioma. Neuro Oncol 2020; 21:786-799. [PMID: 30852603 DOI: 10.1093/neuonc/noz053] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Children with diffuse intrinsic pontine glioma (DIPG) succumb to disease within 2 years of diagnosis despite treatment with ionizing radiation (IR) and/or chemotherapy. Our aim was to determine the role of protein phosphatase, magnesium-dependent 1, delta (PPM1D) mutation, present in up to 25% of cases, in DIPG pathogenesis and treatment. METHODS Using genetic and pharmacologic approaches, we assayed effects of PPM1D mutation on DIPG growth and murine survival. We assayed effects of targeting mutated PPM1D alone or with IR on signaling, cell cycle, proliferation, and apoptosis in patient-derived DIPG cells in vitro, in organotypic brain slices, and in vivo. RESULTS PPM1D-mutated DIPG cell lines exhibited increased proliferation in vitro and in vivo, conferring reduced survival in orthotopically xenografted mice, through stabilization of truncated PPM1D protein and inactivation of DNA damage response (DDR) effectors p53 and H2A.X. PPM1D knockdown or treatment with PPM1D inhibitors suppressed growth of PPM1D-mutated DIPGs in vitro. Orthotopic xenografting of PPM1D short hairpin RNA-transduced or PPM1D inhibitor-treated, PPM1D-mutated DIPG cells into immunodeficient mice resulted in reduced tumor proliferation, increased apoptosis, and extended mouse survival. PPM1D inhibition had similar effects to IR alone on DIPG growth inhibition and augmented the anti-proliferative and pro-apoptotic effects of IR in PPM1D-mutated DIPG models. CONCLUSIONS PPM1D mutations inactivate DDR and promote DIPG growth. Treatment with PPM1D inhibitors activated DDR pathways and enhanced the anti-proliferative and pro-apoptotic effects of IR in DIPG models. Our results support continued development of PPM1D inhibitors for phase I/II trials in children with DIPG.
Collapse
Affiliation(s)
- Mwangala Precious Akamandisa
- Cancer Biology Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, Georgia.,Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Kai Nie
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Rita Nahta
- Department of Pharmacology, Emory University, Atlanta, GA.,Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Dolores Hambardzumyan
- Cancer Biology Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, Georgia.,Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Robert Craig Castellino
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.,Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
16
|
Ozen M, Lipniacki T, Levchenko A, Emamian ES, Abdi A. Modeling and measurement of signaling outcomes affecting decision making in noisy intracellular networks using machine learning methods. Integr Biol (Camb) 2020; 12:122-138. [PMID: 32424393 DOI: 10.1093/intbio/zyaa009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/30/2022]
Abstract
Characterization of decision-making in cells in response to received signals is of importance for understanding how cell fate is determined. The problem becomes multi-faceted and complex when we consider cellular heterogeneity and dynamics of biochemical processes. In this paper, we present a unified set of decision-theoretic, machine learning and statistical signal processing methods and metrics to model the precision of signaling decisions, in the presence of uncertainty, using single cell data. First, we introduce erroneous decisions that may result from signaling processes and identify false alarms and miss events associated with such decisions. Then, we present an optimal decision strategy which minimizes the total decision error probability. Additionally, we demonstrate how graphing receiver operating characteristic curves conveniently reveals the trade-off between false alarm and miss probabilities associated with different cell responses. Furthermore, we extend the introduced framework to incorporate the dynamics of biochemical processes and reactions in a cell, using multi-time point measurements and multi-dimensional outcome analysis and decision-making algorithms. The introduced multivariate signaling outcome modeling framework can be used to analyze several molecular species measured at the same or different time instants. We also show how the developed binary outcome analysis and decision-making approach can be extended to more than two possible outcomes. As an example and to show how the introduced methods can be used in practice, we apply them to single cell data of PTEN, an important intracellular regulatory molecule in a p53 system, in wild-type and abnormal cells. The unified signaling outcome modeling framework presented here can be applied to various organisms ranging from viruses, bacteria, yeast and lower metazoans to more complex organisms such as mammalian cells. Ultimately, this signaling outcome modeling approach can be utilized to better understand the transition from physiological to pathological conditions such as inflammation, various cancers and autoimmune diseases.
Collapse
Affiliation(s)
- Mustafa Ozen
- Center for Wireless Information Processing, Department of Electrical and Computer Engineering, New Jersey Institute of Technology, 323 King Blvd, Newark, NJ 07102, USA
| | - Tomasz Lipniacki
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawinskiego 5B, 02-106 Warsaw, Poland
| | - Andre Levchenko
- Yale Systems Biology Institute and Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Effat S Emamian
- Advanced Technologies for Novel Therapeutics, Enterprise Development Center, New Jersey Institute of Technology, 211 Warren St., Newark, NJ 07103, USA
| | - Ali Abdi
- Center for Wireless Information Processing, Department of Electrical and Computer Engineering, New Jersey Institute of Technology, 323 King Blvd, Newark, NJ 07102, USA.,Department of Biological Sciences, New Jersey Institute of Technology, 323 King Blvd, Newark, NJ 07102, USA
| |
Collapse
|
17
|
Burdova K, Storchova R, Palek M, Macurek L. WIP1 Promotes Homologous Recombination and Modulates Sensitivity to PARP Inhibitors. Cells 2019; 8:cells8101258. [PMID: 31619012 PMCID: PMC6830099 DOI: 10.3390/cells8101258] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/30/2019] [Accepted: 10/10/2019] [Indexed: 12/23/2022] Open
Abstract
Genotoxic stress triggers a combined action of DNA repair and cell cycle checkpoint pathways. Protein phosphatase 2C delta (referred to as WIP1) is involved in timely inactivation of DNA damage response by suppressing function of p53 and other targets at chromatin. Here we show that WIP1 promotes DNA repair through homologous recombination. Loss or inhibition of WIP1 delayed disappearance of the ionizing radiation-induced 53BP1 foci in S/G2 cells and promoted cell death. We identify breast cancer associated protein 1 (BRCA1) as interactor and substrate of WIP1 and demonstrate that WIP1 activity is needed for correct dynamics of BRCA1 recruitment to chromatin flanking the DNA lesion. In addition, WIP1 dephosphorylates 53BP1 at Threonine 543 that was previously implicated in mediating interaction with RIF1. Finally, we report that inhibition of WIP1 allowed accumulation of DNA damage in S/G2 cells and increased sensitivity of cancer cells to a poly-(ADP-ribose) polymerase inhibitor olaparib. We propose that inhibition of WIP1 may increase sensitivity of BRCA1-proficient cancer cells to olaparib.
Collapse
Affiliation(s)
- Kamila Burdova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ14220 Prague, Czech Republic.
| | - Radka Storchova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ14220 Prague, Czech Republic.
| | - Matous Palek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ14220 Prague, Czech Republic.
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, CZ14220 Prague, Czech Republic.
| |
Collapse
|
18
|
Clausse V, Tao D, Debnath S, Fang Y, Tagad HD, Wang Y, Sun H, LeClair CA, Mazur SJ, Lane K, Shi ZD, Vasalatiy O, Eells R, Baker LK, Henderson MJ, Webb MR, Shen M, Hall MD, Appella E, Appella DH, Coussens NP. Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens. J Biol Chem 2019; 294:17654-17668. [PMID: 31481464 PMCID: PMC6873202 DOI: 10.1074/jbc.ra119.010201] [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] [Received: 07/16/2019] [Revised: 08/30/2019] [Indexed: 01/07/2023] Open
Abstract
WT P53-Induced Phosphatase 1 (WIP1) is a member of the magnesium-dependent serine/threonine protein phosphatase (PPM) family and is induced by P53 in response to DNA damage. In several human cancers, the WIP1 protein is overexpressed, which is generally associated with a worse prognosis. Although WIP1 is an attractive therapeutic target, no potent, selective, and bioactive small-molecule modulator with favorable pharmacokinetics has been reported. Phosphatase enzymes are among the most challenging targets for small molecules because of the difficulty of achieving both modulator selectivity and bioavailability. Another major obstacle has been the availability of robust and physiologically relevant phosphatase assays that are suitable for high-throughput screening. Here, we describe orthogonal biochemical WIP1 activity assays that utilize phosphopeptides from native WIP1 substrates. We optimized an MS assay to quantify the enzymatically dephosphorylated peptide reaction product in a 384-well format. Additionally, a red-shifted fluorescence assay was optimized in a 1,536-well format to enable real-time WIP1 activity measurements through the detection of the orthogonal reaction product, Pi. We validated these two optimized assays by quantitative high-throughput screening against the National Center for Advancing Translational Sciences (NCATS) Pharmaceutical Collection and used secondary assays to confirm and evaluate inhibitors identified in the primary screen. Five inhibitors were further tested with an orthogonal WIP1 activity assay and surface plasmon resonance binding studies. Our results validate the application of miniaturized physiologically relevant and orthogonal WIP1 activity assays to discover small-molecule modulators from high-throughput screens.
Collapse
Affiliation(s)
- Victor Clausse
- Synthetic Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Subrata Debnath
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Yuhong Fang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Harichandra D Tagad
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Yuhong Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Hongmao Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Christopher A LeClair
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Kelly Lane
- Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Zhen-Dan Shi
- Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Olga Vasalatiy
- Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Rebecca Eells
- Reaction Biology Corporation, 1 Great Valley Parkway, Suite 2, Malvern, Pennsylvania 19355
| | - Lynn K Baker
- Reaction Biology Corporation, 1 Great Valley Parkway, Suite 2, Malvern, Pennsylvania 19355
| | - Mark J Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Martin R Webb
- Francis Crick Institute, 1 Midland Road, London NW1 AT, United Kingdom
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Ettore Appella
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Daniel H Appella
- Synthetic Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Nathan P Coussens
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| |
Collapse
|
19
|
Fons NR, Sundaram RK, Breuer GA, Peng S, McLean RL, Kalathil AN, Schmidt MS, Carvalho DM, Mackay A, Jones C, Carcaboso ÁM, Nazarian J, Berens ME, Brenner C, Bindra RS. PPM1D mutations silence NAPRT gene expression and confer NAMPT inhibitor sensitivity in glioma. Nat Commun 2019; 10:3790. [PMID: 31439867 PMCID: PMC6706443 DOI: 10.1038/s41467-019-11732-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 08/01/2019] [Indexed: 12/11/2022] Open
Abstract
Pediatric high-grade gliomas are among the deadliest of childhood cancers due to limited knowledge of early driving events in their gliomagenesis and the lack of effective therapies available. In this study, we investigate the oncogenic role of PPM1D, a protein phosphatase often found truncated in pediatric gliomas such as DIPG, and uncover a synthetic lethal interaction between PPM1D mutations and nicotinamide phosphoribosyltransferase (NAMPT) inhibition. Specifically, we show that mutant PPM1D drives hypermethylation of CpG islands throughout the genome and promotes epigenetic silencing of nicotinic acid phosphoribosyltransferase (NAPRT), a key gene involved in NAD biosynthesis. Notably, PPM1D mutant cells are shown to be sensitive to NAMPT inhibitors in vitro and in vivo, within both engineered isogenic astrocytes and primary patient-derived model systems, suggesting the possible application of NAMPT inhibitors for the treatment of pediatric gliomas. Overall, our results reveal a promising approach for the targeting of PPM1D mutant tumors, and define a critical link between oncogenic driver mutations and NAD metabolism, which can be exploited for tumor-specific cell killing.
Collapse
Affiliation(s)
- Nathan R Fons
- Department of Pathology, Yale University, New Haven, CT, 06520, USA.,Department of Therapeutic Radiology, Yale University, New Haven, CT, 06520, USA
| | - Ranjini K Sundaram
- Department of Therapeutic Radiology, Yale University, New Haven, CT, 06520, USA
| | - Gregory A Breuer
- Department of Pathology, Yale University, New Haven, CT, 06520, USA.,Department of Therapeutic Radiology, Yale University, New Haven, CT, 06520, USA
| | - Sen Peng
- The Translational Genomics Research Institute (TGen), Phoenix, AZ, 85004, USA
| | - Ryan L McLean
- Department of Therapeutic Radiology, Yale University, New Haven, CT, 06520, USA
| | - Aravind N Kalathil
- Department of Therapeutic Radiology, Yale University, New Haven, CT, 06520, USA
| | - Mark S Schmidt
- Department of Biochemistry, University of Iowa, Iowa City, IA, 52242, USA
| | - Diana M Carvalho
- Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London, UK
| | - Alan Mackay
- Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London, UK
| | - Chris Jones
- Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London, UK
| | | | - Javad Nazarian
- Children's National Health System, Washington, DC, 20010, USA
| | - Michael E Berens
- The Translational Genomics Research Institute (TGen), Phoenix, AZ, 85004, USA.
| | - Charles Brenner
- Department of Biochemistry, University of Iowa, Iowa City, IA, 52242, USA.
| | - Ranjit S Bindra
- Department of Pathology, Yale University, New Haven, CT, 06520, USA. .,Department of Therapeutic Radiology, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
20
|
Kim B, Won D, Lee ST, Choi JR. Somatic mosaic truncating mutations of PPM1D in blood can result from expansion of a mutant clone under selective pressure of chemotherapy. PLoS One 2019; 14:e0217521. [PMID: 31242196 PMCID: PMC6594580 DOI: 10.1371/journal.pone.0217521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/13/2019] [Indexed: 12/30/2022] Open
Abstract
Background PPM1D (Protein phosphatase magnesium-dependent 1δ) is known as a damage response regulator, a part of the p53 negative feedback loop. Truncating mutations of PPM1D, resulting in overexpression, are frequently found in the blood of patients with breast or ovarian cancer. To identify whether the PPM1D mutation predisposes patients to such cancers or if it results from the cancer and therapy, somatic PPM1D mutations in association with previous cancer and chemotherapy need to be explored. Methods We performed next-generation sequencing (NGS) analysis of blood samples from patients suspected to have hereditary cancer. We grouped the patients according to their diagnoses and history of chemotherapy. For the patients with PPM1D mutations in blood, tumor tissue specimens were examined for the PPM1D mutation using conventional sequencing. Results A total of 1,195 patients, including 719 patients with breast cancer and 240 with ovarian cancer, were tested, and four (~0.3%) had the truncating mutation in PPM1D. All truncating mutations were in exon 6, in mosaic form, with a mean allele fraction of 11.15%. While 395 out of the 1,195 patients had undergone chemotherapy, the four with the truncating mutation had a history of cisplatin-based chemotherapy. No corresponding mutations were identified in the tumor tissues. Conclusions We investigated the frequency of the somatic mosaic PPM1D mutation, in patients with breast or ovarian cancer, which is suggested to be low and related to a history of cisplatin-based chemotherapy. It may be a marker of previous exposure to selective pressure for cells with an impaired DNA damage response.
Collapse
Affiliation(s)
- Borahm Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Dongju Won
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
21
|
Ma A, Dai X. Exploring the Influence of Parameters on the p53 Response When Single-Stranded Breaks and Double-Stranded Breaks Coexist. Interdiscip Sci 2019; 11:679-690. [PMID: 31222582 DOI: 10.1007/s12539-019-00332-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023]
Abstract
The p53 response to DNA damage is closely related to cell fate decisions. P53 preferentially responds to single-stranded breaks (SSBs) exhibiting a graded response when single-stranded breaks (SSBs) and double-stranded breaks (DSBs) coexist. However, how p53 natural preferential response is affected by kinetic parameters remains to be elucidated. Here, based on the hybrid model I, we computationally searched all the parameters and parameter combinations in the parameter space to identify those that could alter the natural preferential response of p53 when SSBs and DSBs coexist. Firstly, when a single parameter is changed, the parameters that can alter graded response to produce p53 pulse response are production rate of ATM- and Rad3-related kinase(ATR) (beta2), ATR degradation rate (alf2) and ATR-dependent p53 production rate (beta31). Secondly, when double parameters are changed, the combinations of beta2/alf2/beta31 and any other parameters are capable of altering the p53 natural preferential response, and the combination of ataxia-telangiectasia mutated kinase (ATM)-dependent p53 production rate (beta3) and Wip1-dependent p53 degradation rate (alf35) is also capable of altering the p53 natural preferential response. Thirdly, we analyzed the sensitivity of both pulse amplitude and apoptosis to kinetic parameters. We find that pulse amplitude is most sensitive to ATM-dependent p53 production rate (beta3), and apoptosis is more sensitive to damage-dependent ATM production rate (beta1), wip1-dependent ATM degradation rate (alf15), wip1 production rate (beta5) and wip1 degradation rate (alf5). What is more, the smaller the value of alf15/beta5 or the larger the value of beta1/alf5, the more susceptible the cells are to apoptosis. These results provide clues to design more effective and less toxic targeted treatments for cancer.
Collapse
Affiliation(s)
- Aiqing Ma
- School of Electronics and Information Technology, Sun Yat-Sen University, No. 132 East Outer Ring Road, Guangzhou, 510006, China
| | - Xianhua Dai
- School of Electronics and Information Technology, Sun Yat-Sen University, No. 132 East Outer Ring Road, Guangzhou, 510006, China.
| |
Collapse
|
22
|
Long X, Lin X. P65‐mediated miR‐590 inhibition modulates the chemoresistance of osteosarcoma to doxorubicin through targeting wild‐type p53‐induced phosphatase 1. J Cell Biochem 2018; 120:5652-5665. [PMID: 30387173 DOI: 10.1002/jcb.27849] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/17/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Xiao Long
- Department of Orthopedic Surgery The First Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou China
| | - Xiang‐Jin Lin
- Department of Orthopedic Surgery The First Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou China
| |
Collapse
|
23
|
Hsu JI, Dayaram T, Tovy A, De Braekeleer E, Jeong M, Wang F, Zhang J, Heffernan TP, Gera S, Kovacs JJ, Marszalek JR, Bristow C, Yan Y, Garcia-Manero G, Kantarjian H, Vassiliou G, Futreal PA, Donehower LA, Takahashi K, Goodell MA. PPM1D Mutations Drive Clonal Hematopoiesis in Response to Cytotoxic Chemotherapy. Cell Stem Cell 2018; 23:700-713.e6. [PMID: 30388424 PMCID: PMC6224657 DOI: 10.1016/j.stem.2018.10.004] [Citation(s) in RCA: 263] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/17/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
Clonal hematopoiesis (CH), in which stem cell clones dominate blood production, becomes increasingly common with age and can presage malignancy development. The conditions that promote ascendancy of particular clones are unclear. We found that mutations in PPM1D (protein phosphatase Mn2+/Mg2+-dependent 1D), a DNA damage response regulator that is frequently mutated in CH, were present in one-fifth of patients with therapy-related acute myeloid leukemia or myelodysplastic syndrome and strongly correlated with cisplatin exposure. Cell lines with hyperactive PPM1D mutations expand to outcompete normal cells after exposure to cytotoxic DNA damaging agents including cisplatin, and this effect was predominantly mediated by increased resistance to apoptosis. Moreover, heterozygous mutant Ppm1d hematopoietic cells outcompeted their wild-type counterparts in vivo after exposure to cisplatin and doxorubicin, but not during recovery from bone marrow transplantation. These findings establish the clinical relevance of PPM1D mutations in CH and the importance of studying mutation-treatment interactions. VIDEO ABSTRACT.
Collapse
MESH Headings
- Aged
- Animals
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Cell Proliferation/drug effects
- Cisplatin/chemistry
- Cisplatin/pharmacology
- Clone Cells/drug effects
- Doxorubicin/chemistry
- Doxorubicin/pharmacology
- Drug Screening Assays, Antitumor
- Female
- HEK293 Cells
- Hematopoiesis/drug effects
- Hematopoiesis/genetics
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Middle Aged
- Mutation
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Protein Phosphatase 2C/genetics
- Protein Phosphatase 2C/metabolism
Collapse
Affiliation(s)
- Joanne I Hsu
- Translational Biology and Molecular Medicine Graduate Program and Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tajhal Dayaram
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ayala Tovy
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Etienne De Braekeleer
- Haematological Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, UK; Wellcome-MRC Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0XY, UK
| | - Mira Jeong
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Feng Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Timothy P Heffernan
- Center for Co-Clinical Trials, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sonal Gera
- Center for Co-Clinical Trials, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey J Kovacs
- Center for Co-Clinical Trials, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joseph R Marszalek
- Center for Co-Clinical Trials, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher Bristow
- Center for Co-Clinical Trials, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuanqing Yan
- Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George Vassiliou
- Haematological Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, UK; Wellcome-MRC Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0XY, UK
| | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lawrence A Donehower
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Koichi Takahashi
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Margaret A Goodell
- Department of Pediatrics, Section of Hematology Oncology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
24
|
Demirkıran G, Kalaycı Demir G, Güzeliş C. Two-dimensional polynomial type canonical relaxation oscillator model for p53 dynamics. IET Syst Biol 2018; 12:138-147. [PMID: 33451182 DOI: 10.1049/iet-syb.2017.0077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/15/2018] [Accepted: 02/26/2018] [Indexed: 11/19/2022] Open
Abstract
p53 network, which is responsible for DNA damage response of cells, exhibits three distinct qualitative behaviours; low state, oscillation and high state, which are associated with normal cell cycle progression, cell cycle arrest and apoptosis, respectively. The experimental studies demonstrate that these dynamics of p53 are due to the ATM and Wip1 interaction. This paper proposes a simple two-dimensional canonical relaxation oscillator model based on the identified topological structure of ATM and Wip1 interaction underlying these qualitative behaviours of p53 network. The model includes only polynomial terms that have the interpretability of known ATM and Wip1 interaction. The introduced model is useful for understanding relaxation oscillations in gene regulatory networks. Through mathematical analysis, we investigate the roles of ATM and Wip1 in forming of these three essential behaviours, and show that ATM and Wip1 constitute the core mechanism of p53 dynamics. In agreement with biological findings, we show that Wip1 degradation term is a highly sensitive parameter, possibly related to mutations. By perturbing the corresponding parameters, our model characterizes some mutations such as ATM deficiency and Wip1 overexpression. Finally, we provide intervention strategies considering our observation that Wip1 seems to be an important target to conduct therapies for these mutations.
Collapse
Affiliation(s)
- Gökhan Demirkıran
- Department of Electrical-Electronics Engineering, Yaşar University, Bornova, İzmir, 35100, Turkey.,The Graduate School of Natural and Applied Sciences, Dokuz Eylül University, Buca, İzmir, 35160, Turkey
| | - Güleser Kalaycı Demir
- Department of Electrical and Electronics Engineering, Dokuz Eylül University, Buca, İzmir, 35160, Turkey
| | - Cüneyt Güzeliş
- Department of Electrical-Electronics Engineering, Yaşar University, Bornova, İzmir, 35100, Turkey
| |
Collapse
|
25
|
Stolte B, Iniguez AB, Dharia NV, Robichaud AL, Conway AS, Morgan AM, Alexe G, Schauer NJ, Liu X, Bird GH, Tsherniak A, Vazquez F, Buhrlage SJ, Walensky LD, Stegmaier K. Genome-scale CRISPR-Cas9 screen identifies druggable dependencies in TP53 wild-type Ewing sarcoma. J Exp Med 2018; 215:2137-2155. [PMID: 30045945 PMCID: PMC6080915 DOI: 10.1084/jem.20171066] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 03/16/2018] [Accepted: 06/27/2018] [Indexed: 01/06/2023] Open
Abstract
Stolte et al. use genome-scale CRISPR-Cas9 screening technology to identify druggable targets for TP53 wild-type Ewing sarcoma and discover reactivation of p53 through inhibition of MDM2, MDM4, Wip1, or USP7 as therapeutic strategies for the disease. Ewing sarcoma is a pediatric cancer driven by EWS-ETS transcription factor fusion oncoproteins in an otherwise stable genomic background. The majority of tumors express wild-type TP53, and thus, therapies targeting the p53 pathway would benefit most patients. To discover targets specific for TP53 wild-type Ewing sarcoma, we used a genome-scale CRISPR-Cas9 screening approach and identified and validated MDM2, MDM4, USP7, and PPM1D as druggable dependencies. The stapled peptide inhibitor of MDM2 and MDM4, ATSP-7041, showed anti-tumor efficacy in vitro and in multiple mouse models. The USP7 inhibitor, P5091, and the Wip1/PPM1D inhibitor, GSK2830371, decreased the viability of Ewing sarcoma cells. The combination of ATSP-7041 with P5091, GSK2830371, and chemotherapeutic agents showed synergistic action on the p53 pathway. The effects of the inhibitors, including the specific USP7 inhibitor XL-188, were rescued by concurrent TP53 knockout, highlighting the essentiality of intact p53 for the observed cytotoxic activities.
Collapse
Affiliation(s)
- Björn Stolte
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA.,Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, LMU Munich, Munich, Germany.,The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Amanda Balboni Iniguez
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA.,The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Neekesh V Dharia
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA.,The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Amanda L Robichaud
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Amy Saur Conway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Ann M Morgan
- Department of Pediatric Oncology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Gabriela Alexe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA.,The Broad Institute of MIT and Harvard, Cambridge, MA.,Bioinformatics Graduate Program, Boston University, Boston, MA
| | - Nathan J Schauer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Xiaoxi Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Gregory H Bird
- Department of Pediatric Oncology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - Sara J Buhrlage
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Loren D Walensky
- Department of Pediatric Oncology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA .,The Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
26
|
Zhu D, Osuka S, Zhang Z, Reichert ZR, Yang L, Kanemura Y, Jiang Y, You S, Zhang H, Devi NS, Bhattacharya D, Takano S, Gillespie GY, Macdonald T, Tan C, Nishikawa R, Nelson WG, Olson JJ, Van Meir EG. BAI1 Suppresses Medulloblastoma Formation by Protecting p53 from Mdm2-Mediated Degradation. Cancer Cell 2018; 33:1004-1016.e5. [PMID: 29894688 PMCID: PMC6002773 DOI: 10.1016/j.ccell.2018.05.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/29/2017] [Accepted: 05/11/2018] [Indexed: 01/20/2023]
Abstract
Adhesion G protein-coupled receptors (ADGRs) encompass 33 human transmembrane proteins with long N termini involved in cell-cell and cell-matrix interactions. We show the ADGRB1 gene, which encodes Brain-specific angiogenesis inhibitor 1 (BAI1), is epigenetically silenced in medulloblastomas (MBs) through a methyl-CpG binding protein MBD2-dependent mechanism. Knockout of Adgrb1 in mice augments proliferation of cerebellar granule neuron precursors, and leads to accelerated tumor growth in the Ptch1+/- transgenic MB mouse model. BAI1 prevents Mdm2-mediated p53 polyubiquitination, and its loss substantially reduces p53 levels. Reactivation of BAI1/p53 signaling axis by a brain-permeable MBD2 pathway inhibitor suppresses MB growth in vivo. Altogether, our data define BAI1's physiological role in tumorigenesis and directly couple an ADGR to cancer formation.
Collapse
Affiliation(s)
- Dan Zhu
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Satoru Osuka
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Zhaobin Zhang
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | | | - Liquan Yang
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Yonehiro Kanemura
- Division of Regenerative Medicine, Institute for Clinical Research, Osaka National Hospital, National Hospital Organization, 2-1-14 Hoenzaka, Chuo-ku, Osaka 540-0006, Japan
| | - Ying Jiang
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, GA 30322, USA
| | - Shuo You
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Hanwen Zhang
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Narra S Devi
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Debanjan Bhattacharya
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Shingo Takano
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - G Yancey Gillespie
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tobey Macdonald
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, 1365C Clifton Road N.E, C5078, Atlanta, GA 30322, USA
| | - Chalet Tan
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, GA 30322, USA
| | - Ryo Nishikawa
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Saitama, Japan
| | - William G Nelson
- Johns Hopkins University, 401 North Broadway, Baltimore, MD 21287, USA
| | - Jeffrey J Olson
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, 1365C Clifton Road N.E, C5078, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Erwin G Van Meir
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, 1365C Clifton Road N.E, C5078, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
27
|
Tagad HD, Debnath S, Clausse V, Saha M, Mazur SJ, Appella E, Appella DH. Chemical Features Important for Activity in a Class of Inhibitors Targeting the Wip1 Flap Subdomain. ChemMedChem 2018; 13:894-901. [PMID: 29476592 PMCID: PMC8022280 DOI: 10.1002/cmdc.201700779] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Indexed: 12/12/2022]
Abstract
The wild-type p53 induced phosphatase 1, Wip1 (PP2Cδ), is a protein phosphatase 2C (PP2C) family serine/threonine phosphatase that negatively regulates the function of the tumor suppressor p53 and several of its positive regulators such as ATM, Chk1, Chk2, Mdm2, and p38 MAPK. Wip1 dephosphorylates and inactivates its protein targets, which are critical for cellular stress responses. Additionally, Wip1 is frequently amplified and overexpressed in several human cancer types. Because of its negative role in regulating the function of tumor suppressor proteins, Wip1 has been identified as a potential therapeutic target in various types of cancers. Based on a recently reported Wip1 inhibitor (G-1), we performed an extensive structure-activity relationship (SAR) analysis. This led us to interesting findings in SAR trends and to the discovery of new chemical analogues with good specificity and bioavailability.
Collapse
Affiliation(s)
- Harichandra D Tagad
- Laboratory of Cell Biology, National Cancer Institute, US National Institutes of Health, Bethesda, MD, 20892, USA
| | - Subrata Debnath
- Laboratory of Cell Biology, National Cancer Institute, US National Institutes of Health, Bethesda, MD, 20892, USA
| | - Victor Clausse
- Synthetic Bioactive Molecules Section, LBC, NIDDK, US National Institutes of Health, 8 Center Drive, Room 404, Bethesda, MD, 20892, USA
| | - Mrinmoy Saha
- Synthetic Bioactive Molecules Section, LBC, NIDDK, US National Institutes of Health, 8 Center Drive, Room 404, Bethesda, MD, 20892, USA
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, National Cancer Institute, US National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ettore Appella
- Laboratory of Cell Biology, National Cancer Institute, US National Institutes of Health, Bethesda, MD, 20892, USA
| | - Daniel H Appella
- Synthetic Bioactive Molecules Section, LBC, NIDDK, US National Institutes of Health, 8 Center Drive, Room 404, Bethesda, MD, 20892, USA
| |
Collapse
|
28
|
Bhattacharya D, Hiregange D, Rao BJ. ATR kinase regulates its attenuation via PPM1D phosphatase recruitment to chromatin during recovery from DNA replication stress signalling. J Biosci 2018; 43:25-47. [PMID: 29485113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In eukaryotes, in response to replication stress, DNA damage response kinase, ATR is activated, whose signalling abrogation leads to cell lethality due to aberrant fork remodelling and excessive origin firing. Here we report that inhibition of ATR kinase activity specifically during replication stress recovery results in persistent ATR signalling, evidenced by the presence of ATR-dependent phosphorylation marks (gamma H2AX, pChk1 and pRad17) and delayed cell cycle re-entry. Further, such disruption of ATR signalling attenuation leads to double-strand breaks, fork collapse and thereby 'replication catastrophe'. PPM1D phosphatase, a nucleolar localized protein, relocates to chromatin during replication stress and reverts back to nucleolus following stress recovery, under the control of ATR kinase action. Inhibition of ATR kinase activity, specifically during post replication stress, triggers dislodging of the chromatin-bound PPM1D from nucleus to cytoplasm followed by its degradation, thereby leading to persistence of activated ATR marks in the nuclei. Chemical inhibition of PPM1D activity or SiRNA mediated depletion of the protein during post replication stress recovery 'phenocopies' ATR kinase inhibition by failing to attenuate ATR signalling. Collectively, our observations suggest a novel role of ATR kinase in mediating its own signal attenuation via PPM1D recruitment to chromatin as an essential mechanism for restarting the stalled forks, cell-cycle re-entry and cellular recovery from replication stress.
Collapse
Affiliation(s)
- Debadrita Bhattacharya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400 005, India
| | | | | |
Collapse
|
29
|
Bhattacharya D, Hiregange D, Rao BJ. ATR kinase regulates its attenuation via PPM1D phosphatase recruitment to chromatin during recovery from DNA replication stress signalling. J Biosci 2018. [DOI: 10.1007/s12038-018-9736-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
30
|
Demirkıran G, Kalaycı Demir G, Güzeliş C. Revealing determinants of two-phase dynamics of P53 network under gamma irradiation based on a reduced 2D relaxation oscillator model. IET Syst Biol 2018; 12:26-38. [PMID: 29337287 PMCID: PMC8687238 DOI: 10.1049/iet-syb.2017.0041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/17/2017] [Accepted: 10/25/2017] [Indexed: 01/03/2023] Open
Abstract
This study proposes a two-dimensional (2D) oscillator model of p53 network, which is derived via reducing the multidimensional two-phase dynamics model into a model of ataxia telangiectasia mutated (ATM) and Wip1 variables, and studies the impact of p53-regulators on cell fate decision. First, the authors identify a 6D core oscillator module, then reduce this module into a 2D oscillator model while preserving the qualitative behaviours. The introduced 2D model is shown to be an excitable relaxation oscillator. This oscillator provides a mechanism that leads diverse modes underpinning cell fate, each corresponding to a cell state. To investigate the effects of p53 inhibitors and the intrinsic time delay of Wip1 on the characteristics of oscillations, they introduce also a delay differential equation version of the 2D oscillator. They observe that the suppression of p53 inhibitors decreases the amplitudes of p53 oscillation, though the suppression increases the sustained level of p53. They identify Wip1 and P53DINP1 as possible targets for cancer therapies considering their impact on the oscillator, supported by biological findings. They model some mutations as critical changes of the phase space characteristics. Possible cancer therapeutic strategies are then proposed for preventing these mutations' effects using the phase space approach.
Collapse
Affiliation(s)
- Gökhan Demirkıran
- The Graduate School of Natural and Applied Sciences, Dokuz Eylül University, Buca, İzmir 35160, Turkey.
| | - Güleser Kalaycı Demir
- Department of Electrical and Electronics Engineering, Dokuz Eylül University, Buca, İzmir 35160, Turkey
| | - Cüneyt Güzeliş
- Department of Electrical-Electronics Engineering, Yaşar University, Bornova, İzmir 35100, Turkey
| |
Collapse
|
31
|
Pechackova S, Burdova K, Benada J, Kleiblova P, Jenikova G, Macurek L. Inhibition of WIP1 phosphatase sensitizes breast cancer cells to genotoxic stress and to MDM2 antagonist nutlin-3. Oncotarget 2018; 7:14458-75. [PMID: 26883108 PMCID: PMC4924728 DOI: 10.18632/oncotarget.7363] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 01/29/2016] [Indexed: 02/07/2023] Open
Abstract
PP2C family serine/threonine phosphatase WIP1 acts as a negative regulator of the tumor suppressor p53 and is implicated in silencing of cellular responses to genotoxic stress. Chromosomal locus 17q23 carrying the PPM1D (coding for WIP1) is commonly amplified in breast carcinomas and WIP1 was proposed as potential pharmacological target. Here we employed a cellular model with knocked out PPM1D to validate the specificity and efficiency of GSK2830371, novel small molecule inhibitor of WIP1. We have found that GSK2830371 increased activation of the DNA damage response pathway to a comparable level as the loss of PPM1D. In addition, GSK2830371 did not affect proliferation of cells lacking PPM1D but significantly supressed proliferation of breast cancer cells with amplified PPM1D. Over time cells treated with GSK2830371 accumulated in G1 and G2 phases of the cell cycle in a p21-dependent manner and were prone to induction of senescence by a low dose of MDM2 antagonist nutlin-3. In addition, combined treatment with GSK2830371 and doxorubicin or nutlin-3 potentiated cell death through a strong induction of p53 pathway and activation of caspase 9. We conclude that efficient inhibition of WIP1 by GSK2830371 sensitizes breast cancer cells with amplified PPM1D and wild type p53 to chemotherapy.
Collapse
Affiliation(s)
- Sona Pechackova
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220 Prague, Czech Republic
| | - Kamila Burdova
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220 Prague, Czech Republic
| | - Jan Benada
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220 Prague, Czech Republic
| | - Petra Kleiblova
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220 Prague, Czech Republic.,Institute of Biochemistry and Experimental Oncology, Charles University in Prague, CZ-12853 Prague, Czech Republic
| | - Gabriela Jenikova
- 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
|
32
|
Bartek J, Fornara O, Merchut-Maya JM, Maya-Mendoza A, Rahbar A, Stragliotto G, Broholm H, Svensson M, Sehested A, Söderberg Naucler C, Bartek J, Bartkova J. Replication stress, DNA damage signalling, and cytomegalovirus infection in human medulloblastomas. Mol Oncol 2017; 11:945-964. [PMID: 28383788 PMCID: PMC5537913 DOI: 10.1002/1878-0261.12061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/04/2017] [Accepted: 03/13/2017] [Indexed: 12/22/2022] Open
Abstract
Medulloblastomas are the most common, and often fatal, paediatric brain tumours that feature high genomic instability, frequent infection by human cytomegalovirus (HCMV) and resistance to radiation and chemotherapy. The causes of the pronounced chromosomal instability and its potential links with HCMV infection and/or resistance to genotoxic therapies remain largely unknown. To address these issues, here we have combined immunohistochemical analysis of a series of 25 paediatric medulloblastomas, complemented by medulloblastoma cell culture models including experimental HCMV infection. Using eight established immunohistochemical markers to assess the status of the DDR machinery, we found pronounced endogenous DNA damage signalling (γH2AX marker) and robust constitutive activation of both the ATM‐Chk2 and ATR‐Chk1 DNA damage checkpoint kinase cascades, yet unexpectedly modest p53 tumour suppressor activation, across our medulloblastoma cohort. Most tumours showed high proliferation (Ki67 marker), variable oxidative DNA damage (8‐oxoguanine lesions) and formation of 53BP1 nuclear ‘bodies’, the latter indicating (along with ATR‐Chk1 signalling) endogenous replication stress. The bulk of the clinical specimens also showed expression of HCMV immediate early and late proteins, in comparative analyses using three immunohistochemical protocols. Cell culture experiments validated the chronic endogenous replication stress in medulloblastoma cell lines and showed sharply differential, intriguing responses of normal cells and medulloblastoma cells to HCMV infection, including differential subcellular mislocalization and enhancement of replication stress‐related 53BP1 body formation, the latter in cell‐non‐autonomous manner. Overall, our results strongly indicate that in human medulloblastomas, the DDR checkpoint barrier is widely activated, at least in part due to replication stress. Furthermore, we propose that unorthodox p53 defects other than mutations may allow high proliferation despite the ongoing checkpoint signalling and that the highly prevalent HCMV may impact the medulloblastoma host cell replication stress and DNA repair. Collectively, the scenario we report here likely fuels genomic instability and evolution of medulloblastoma resistance to standard‐of‐care genotoxic treatments.
Collapse
Affiliation(s)
- Jiri Bartek
- Department of Medicine, Unit of Microbial Pathogenesis, Karolinska Institutet, Stockholm, Sweden.,Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden.,Department of Neurosurgery, Copenhagen University Hospital Rigshospitalet, Denmark
| | - Olesja Fornara
- Department of Medicine, Unit of Microbial Pathogenesis, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Afshar Rahbar
- Department of Medicine, Unit of Microbial Pathogenesis, Karolinska Institutet, Stockholm, Sweden
| | - Giuseppe Stragliotto
- Department of Medicine, Unit of Microbial Pathogenesis, Karolinska Institutet, Stockholm, Sweden
| | - Helle Broholm
- Department of Pathology, Copenhagen University Hospital Rigshospitalet, Denmark
| | - Mikael Svensson
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Astrid Sehested
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Denmark
| | | | - Jiri Bartek
- Danish Cancer Society Research Center, Copenhagen, Denmark.,Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Jirina Bartkova
- Danish Cancer Society Research Center, Copenhagen, Denmark.,Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
33
|
Liu KM, Zhang HH, Wang YN, Wang LM, Chen HY, Long CF, Zhang LF, Zhang HB, Yan HB. Wild-type p53-induced Phosphatase 1 Deficiency Exacerbates Myocardial Infarction-induced Ischemic Injury. Chin Med J (Engl) 2017; 130:1333-1341. [PMID: 28524834 PMCID: PMC5455044 DOI: 10.4103/0366-6999.206353] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Myocardial infarction (MI) is a major disease burden. Wild-type p53-induced phosphatase 1 (Wip1) has been studied extensively in the context of cancer and the regulation of different types of stem cells, but the role of Wip1 in cardiac adaptation to MI is unknown. We investigated the significance of Wip1 in a mouse model of MI. METHODS The study began in June 2014 and was completed in July 2016. We compared Wip1-knockout (Wip1-KO) mice and wild-type (WT) mice to determine changes in cardiac function and survival in response to MI. The heart weight/body weight (HW/BW) ratio and cardiac function were measured before MI. Mouse MI was established by ligating the left anterior descending (LAD) coronary artery under 1.5% isoflurane anesthesia. After MI, survival of the mice was observed for 4 weeks. Cardiac function was examined by echocardiography. The HW/BW ratio was analyzed, and cardiac hypertrophy was measured by wheat germ agglutinin staining. Hematoxylin and eosin (H&E) staining was used to determine the infarct size. Gene expression of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) was assessed by quantitative real-time polymerase chain reaction (qPCR), and the levels of signal transducers and activators of transcription 3 (stat3) and phosphor-stat3 (p-stat3) were also analyzed by Western blotting. Kaplan-Meier survival analysis, log-rank test, unpaired t-test, and one-way analysis of variance (ANOVA) were used for statistical analyses. RESULTS Wip1-KO mice had a marginally increased HW/BW ratio and slightly impaired cardiac function before LAD ligation. After MI, Wip1-deficient mice exhibited increased mortality (57.14% vs. 29.17%; n = 24 [WT], n = 35 [Wip1-KO], P< 0.05), increased cardiac hypertrophy (HW/BW ratio: 7 days: 7.25 ± 0.36 vs. 5.84 ± 0.18, n = 10, P< 0.01, and 4 weeks: 6.05 ± 0.17 vs. 5.87 ± 0.24, n = 10, P > 0.05; cross-sectional area: 7 days: 311.80 ± 8.29 vs. 268.90 ± 11.15, n = 6, P< 0.05, and 4 weeks: 308.80 ± 11.26 vs. 317.00 ± 13.55, n = 6, P > 0.05), and reduced cardiac function (ejection fraction: 7 days: 29.37 ± 1.38 vs. 34.72 ± 1.81, P< 0.05, and 4 weeks: 19.06 ± 2.07 vs. 26.37 ± 2.95, P< 0.05; fractional shortening: 7 days: 13.72 ± 0.71 vs. 16.50 ± 0.94, P< 0.05, and 4 weeks: 8.79 ± 1.00 vs. 12.48 ± 1.48, P< 0.05; n = 10 [WT], n = 15 [Wip1-KO]). H&E staining revealed a larger infarct size in Wip1-KO mice than in WT mice (34.79% ± 2.44% vs. 19.55% ± 1.48%, n = 6, P< 0.01). The expression of IL-6 and p-stat3 was downregulated in Wip1-KO mice (IL-6: 1.71 ± 0.27 vs. 4.46 ± 0.79, n = 6, P< 0.01; and p-stat3/stat3: 1.15 ± 0.15 vs. 1.97 ± 0.23, n = 6, P< 0.05). CONCLUSION The results suggest that Wip1 could protect the heart from MI-induced ischemic injury.
Collapse
Affiliation(s)
- Ke-Mei Liu
- Department of Coronary Artery Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Hai-Hong Zhang
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ya-Nan Wang
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lian-Mei Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100029, China
| | - Hong-Yu Chen
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Cai-Feng Long
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lian-Feng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Hong-Bing Zhang
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hong-Bing Yan
- Department of Coronary Artery Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
34
|
Pecháčková S, Burdová K, Macurek L. WIP1 phosphatase as pharmacological target in cancer therapy. J Mol Med (Berl) 2017; 95:589-599. [PMID: 28439615 PMCID: PMC5442293 DOI: 10.1007/s00109-017-1536-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/13/2017] [Accepted: 04/19/2017] [Indexed: 12/31/2022]
Abstract
DNA damage response (DDR) pathway protects cells from genome instability and prevents cancer development. Tumor suppressor p53 is a key molecule that interconnects DDR, cell cycle checkpoints, and cell fate decisions in the presence of genotoxic stress. Inactivating mutations in TP53 and other genes implicated in DDR potentiate cancer development and also influence the sensitivity of cancer cells to treatment. Protein phosphatase 2C delta (referred to as WIP1) is a negative regulator of DDR and has been proposed as potential pharmaceutical target. Until recently, exploitation of WIP1 inhibition for suppression of cancer cell growth was compromised by the lack of selective small-molecule inhibitors effective at cellular and organismal levels. Here, we review recent advances in development of WIP1 inhibitors and discuss their potential use in cancer treatment.
Collapse
Affiliation(s)
- Soňa Pecháčková
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220, Prague, Czech Republic
| | - Kamila Burdová
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220, Prague, Czech Republic
| | - Libor Macurek
- Department of Cancer Cell Biology, Institute of Molecular Genetics of the ASCR, CZ-14220, Prague, Czech Republic.
| |
Collapse
|
35
|
Feng Y, Liu F, Du Z, Zhao D, Cheng J, Guo W. Wip1 regulates SKOV3 cell apoptosis through the p38 MAPK signaling pathway. Mol Med Rep 2017; 15:3651-3657. [PMID: 28440479 PMCID: PMC5436208 DOI: 10.3892/mmr.2017.6469] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022] Open
Abstract
The aim of the present study was to explore the effect of silencing wild-type p53-induced phosphatase 1 (Wip1) on apoptosis of human ovarian cancer SKOV3 cells. SKOV3 cells cultured in vitro were divided into three groups: untreated cells, cells transfected with control small interfering RNA (siRNA) and cells transfected with siRNA targeting Wip1. Flow cytometry analysis was used to detect cell apoptosis. Western blot analysis was performed to determine expression of tumor protein 53 (p53), cleaved caspase-3, caspase-3, BCL2 associated X (Bax), BCL2 apoptosis regulator (Bcl-2), p38 mitogen-activated protein kinase (p38 MAPK) and phosphorylated (p)-p38 MAPK. Reverse transcription-quantitative polymerase chain reaction was used to detect expression of p53, Bax, Bcl-2 and caspase-3 mRNAs. Compared with control, apoptosis of SKOV3 cell was significantly increased following Wip1 siRNA silencing. Wip1 silencing also resulted in a significant increase of p53 and p-p38 MAPK expression, as well as increased cleaved caspase-3/caspase-3 and Bax/Bcl-2 protein ratios. No significant differences were observed in apoptosis and apoptosis-related protein expression in the control siRNA transfected cells. The present study demonstrated that Wip1 silencing promotes apoptosis of human ovarian cancer SKOV3 cells by activation of the p38 MAPK signaling pathways and through subsequent upregulation of p53, and cleaved caspase-3/caspase-3 and Bax/Bcl-2 protein ratios. Overall, the findings of the present study suggest that targeting Wip1 may be a potential therapeutic avenue for the treatment of human ovarian cancer in the future.
Collapse
Affiliation(s)
- Yanping Feng
- Department of Reproductive Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Fang Liu
- Department of Reproductive Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Zhixiang Du
- Department of Reproductive Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Dongjie Zhao
- Department of Surgery, The Third Hospital of Tangshan, Tangshan, Hebei 063100, P.R. China
| | - Jianxin Cheng
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Wei Guo
- Department of Reproductive Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| |
Collapse
|
36
|
Wen J, Lee J, Malhotra A, Nahta R, Arnold AR, Buss MC, Brown BD, Maier C, Kenney AM, Remke M, Ramaswamy V, Taylor MD, Castellino RC. WIP1 modulates responsiveness to Sonic Hedgehog signaling in neuronal precursor cells and medulloblastoma. Oncogene 2016; 35:5552-5564. [PMID: 27086929 PMCID: PMC5069081 DOI: 10.1038/onc.2016.96] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 12/28/2015] [Accepted: 01/22/2016] [Indexed: 12/18/2022]
Abstract
High-level amplification of the protein phosphatase PPM1D (WIP1) is present in a subset of medulloblastomas (MBs) that have an expression profile consistent with active Sonic Hedgehog (SHH) signaling. We found that WIP1 overexpression increased expression of Shh target genes and cell proliferation in response to Shh stimulation in NIH3T3 and cerebellar granule neuron precursor cells in a p53-independent manner. Thus, we developed a mouse in which WIP1 is expressed in the developing brain under control of the Neurod2 promoter (ND2:WIP1). The external granule layer (EGL) in early postnatal ND2:WIP1 mice exhibited increased proliferation and expression of Shh downstream targets. MB incidence increased and survival decreased when ND2:WIP1 mice were crossed with an Shh-activated MB mouse model. Conversely, Wip1 knockout significantly suppressed MB formation in two independent mouse models of Shh-activated MB. Furthermore, Wip1 knockdown or treatment with a WIP1 inhibitor suppressed the effects of Shh stimulation and potentiated the growth inhibitory effects of SHH pathway-inhibiting drugs in Shh-activated MB cells in vitro. This suggests an important cross-talk between SHH and WIP1 pathways that accelerates tumorigenesis and supports WIP1 inhibition as a potential treatment strategy for MB.
Collapse
Affiliation(s)
- Jing Wen
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Atlanta, GA 30322, USA
| | - Juhyun Lee
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Atlanta, GA 30322, USA
| | - Anshu Malhotra
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Atlanta, GA 30322, USA
| | - Rita Nahta
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Pharmacology, Atlanta, GA 30322, USA
| | - Amanda R. Arnold
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Atlanta, GA 30322, USA
| | - Meghan C. Buss
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Atlanta, GA 30322, USA
| | - Briana D. Brown
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Atlanta, GA 30322, USA
| | - Caroline Maier
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Atlanta, GA 30322, USA
| | - Anna M. Kenney
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Marc Remke
- Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumour Research Center, and Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Vijay Ramaswamy
- Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumour Research Center, and Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Michael D. Taylor
- Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumour Research Center, and Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Robert C. Castellino
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
37
|
Akamandisa MP, Nahta R, Castellino RC. PPM1D/WIP1 in medulloblastoma. Oncoscience 2016; 3:186-187. [PMID: 27713908 PMCID: PMC5043068 DOI: 10.18632/oncoscience.318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 08/12/2016] [Indexed: 11/25/2022] Open
Affiliation(s)
- Mwangala P Akamandisa
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Rita Nahta
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Robert C Castellino
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
38
|
Wang HY, Liu ZS, Qiu L, Guo J, Li YF, Zhang J, Wang TJ, Liu XD. Knockdown of Wip1 Enhances Sensitivity to Radiation in HeLa Cells Through Activation of p38 MAPK. Oncol Res 2016; 22:225-233. [PMID: 26351212 PMCID: PMC7838432 DOI: 10.3727/096504015x14386062091479] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The objectives of the study were to investigate the functional role and potential mechanism of wild-type p53-induced phosphatase (Wip1) in cervical cancer cell line HeLa cells, along with the effect of knockdown of Wip1 in combination with γ-irradiation on the HeLa cells. Expression of Wip1 was silenced or overexpressed. After transfection, cell viability was determined. Moreover, γ-irradiation and SB203580 were performed to explore the effect of colony formation and cell apoptosis. Likewise, protein expression levels of p38, p-p38, p53, and p-p53 were assessed in the presence or not of SB203580 and overexpression of Wip1. Both the mRNA and protein levels of Wip1 were significantly decreased by transfection with Wip1-specific small interfering RNA (siRNA) but were significantly increased by transfection with pcDNA3.1-Wip1. Knockdown of Wip1 significantly decreased cell growth and colony formation ability and increased apoptotic rate. Additionally, better results were obtained by knockdown of Wip1 in combination with γ-irradiation. The protein expression levels of p-p38 (p < 0.05), p53 (p < 0.01), and p-p53 (p < 0.05) were all significantly increased by knockdown of Wip1. However, application of SB203580 reversed the effects. Our study confirms the important roles of Wip1 in cervical cancer. Knockdown of Wip1 enhances sensitivity to radiation in HeLa cells by inhibiting cell proliferation and inducing apoptosis through activation of p38 MAPK.
Collapse
Affiliation(s)
- Hong-Yong Wang
- Department of Radiotherapy, 2nd Hospital Affiliated to Jilin University, Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The mechanisms leading to brain tumor formation are poorly understood. Using Ptch1+/- mice as a medulloblastoma model, sequential mutations were found to shape tumor evolution. Initially, medulloblastoma preneoplastic lesions display loss of heterozygosity of the Ptch1 wild-type allele, an event associated with cell senescence in preneoplasia. Subsequently, p53 mutations lead to senescence evasion and progression from preneoplasia to medulloblastoma. These findings are consistent with a model where high levels of Hedgehog signaling caused by the loss of the tumor suppressor Ptch1 lead to oncogene-induced senescence and drive p53 mutations. Thus, cell senescence is an important characteristic of a subset of SHH medulloblastoma and might explain the acquisition of somatic TP53 mutations in human medulloblastoma. This mode of medulloblastoma formation contrasts with the one characterizing Li-Fraumeni patients with medulloblastoma, where TP53 germ-line mutations cause chromothriptic genomic instability and lead to mutations in Hedgehog signaling genes, which drive medulloblastoma growth. Here we discuss in detail these 2 alternative mechanisms leading to medulloblastoma tumorigenesis.
Collapse
Affiliation(s)
- Lukas Tamayo-Orrego
- a Molecular Biology of Neural Development , Institut de Recherches Cliniques de Montréal (IRCM) , Montreal , Quebec , Canada.,b Integrated Program in Neuroscience , McGill University , Montreal , Quebec , Canada
| | - Shannon M Swikert
- a Molecular Biology of Neural Development , Institut de Recherches Cliniques de Montréal (IRCM) , Montreal , Quebec , Canada.,b Integrated Program in Neuroscience , McGill University , Montreal , Quebec , Canada
| | - Frédéric Charron
- a Molecular Biology of Neural Development , Institut de Recherches Cliniques de Montréal (IRCM) , Montreal , Quebec , Canada.,b Integrated Program in Neuroscience , McGill University , Montreal , Quebec , Canada.,c Department of Medicine , University of Montreal , Montreal , Quebec , Canada.,d Division of Experimental Medicine , Department of Medicine, Department of Anatomy and Cell Biology, Department of Biology , McGill University , Quebec , Canada
| |
Collapse
|
40
|
Wang B, Li D, Sidler C, Rodriguez-Juarez R, Singh N, Heyns M, Ilnytskyy Y, Bronson RT, Kovalchuk O. A suppressive role of ionizing radiation-responsive miR-29c in the development of liver carcinoma via targeting WIP1. Oncotarget 2016; 6:9937-50. [PMID: 25888625 PMCID: PMC4496408 DOI: 10.18632/oncotarget.3157] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/15/2015] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related deaths worldwide, and it has been linked to radiation exposure. As a well-defined oncogene, wild-type p53-induced phosphatase 1 (WIP1) plays an inhibitory role in several tumor suppressor pathways, including p53. WIP1 is amplified and overexpressed in many malignancies, including HCC. However, the underlying mechanisms remain largely unknown. Here, we show that low-dose ionizing radiation (IR) induces miR-29c expression in female mouse liver, while inhibiting its expression in HepG2, a human hepatocellular carcinoma cell line which is used as a model system in this study. miR-29c expression is downregulated in human hepatocellular carcinoma cells, which is inversely correlated with WIP1 expression. miR-29c attenuates luciferase activity of a reporter harboring the 3'UTR binding motif of WIP1 mRNA. Ectopic expression of miR-29c significantly represses cell proliferation and induces apoptosis and G1 arrest in HepG2. In contrast, the knockdown of miR-29c greatly enhances HepG2 cell proliferation and suppresses apoptosis. The biological effects of miR-29c may be mediated by its target WIP1 which regulates p53 activity via dephosphorylation at Ser-15. Finally, fluorescence in situ hybridization (FISH) and immunohistochemical analyses indicate that miR-29c is downregulated in 50.6% of liver carcinoma tissues examined, whereas WIP1 is upregulated in 45.4% of these tissues. The expression of miR-29c inversely correlates with that of WIP1 in HCC. Our results suggest that the IR-responsive miR-29c may function as a tumor suppressor that plays a crucial role in the development of liver carcinoma via targeting WIP1, therefore possibly representing a target molecule for therapeutic intervention for this disease.
Collapse
Affiliation(s)
- Bo Wang
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Canada.,Department of Biochemistry, Qiqihar Medical University, Qiqihar, P.R. China
| | - Dongping Li
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Canada.,Department of Biochemistry, Qiqihar Medical University, Qiqihar, P.R. China
| | - Corinne Sidler
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Canada
| | | | - Natasha Singh
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Canada
| | - Mieke Heyns
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Canada
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Canada
| | - Roderick T Bronson
- The Dana Farber/Harvard Comprehensive Cancer Center, Boston, Massachusetts, USA
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Canada
| |
Collapse
|
41
|
Evasion of Cell Senescence Leads to Medulloblastoma Progression. Cell Rep 2016; 14:2925-37. [PMID: 26997276 DOI: 10.1016/j.celrep.2016.02.061] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 12/28/2015] [Accepted: 02/11/2016] [Indexed: 11/23/2022] Open
Abstract
How brain tumors progress from precancerous lesions to advanced cancers is not well understood. Using Ptch1(+/-) mice to study medulloblastoma progression, we found that Ptch1 loss of heterozygosity (LOH) is an early event that is associated with high levels of cell senescence in preneoplasia. In contrast, advanced tumors have evaded senescence. Remarkably, we discovered that the majority of advanced medulloblastomas display either spontaneous, somatic p53 mutations or Cdkn2a locus inactivation. Consistent with senescence evasion, these p53 mutations are always subsequent to Ptch1 LOH. Introduction of a p53 mutation prevents senescence, accelerates tumor formation, and increases medulloblastoma incidence. Altogether, our results show that evasion of senescence associated with Ptch1 LOH allows progression to advanced tumors.
Collapse
|
42
|
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: 2.0] [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
|
43
|
Hat B, Kochańczyk M, Bogdał MN, Lipniacki T. Feedbacks, Bifurcations, and Cell Fate Decision-Making in the p53 System. PLoS Comput Biol 2016; 12:e1004787. [PMID: 26928575 PMCID: PMC4771203 DOI: 10.1371/journal.pcbi.1004787] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/03/2016] [Indexed: 01/07/2023] Open
Abstract
The p53 transcription factor is a regulator of key cellular processes including DNA repair, cell cycle arrest, and apoptosis. In this theoretical study, we investigate how the complex circuitry of the p53 network allows for stochastic yet unambiguous cell fate decision-making. The proposed Markov chain model consists of the regulatory core and two subordinated bistable modules responsible for cell cycle arrest and apoptosis. The regulatory core is controlled by two negative feedback loops (regulated by Mdm2 and Wip1) responsible for oscillations, and two antagonistic positive feedback loops (regulated by phosphatases Wip1 and PTEN) responsible for bistability. By means of bifurcation analysis of the deterministic approximation we capture the recurrent solutions (i.e., steady states and limit cycles) that delineate temporal responses of the stochastic system. Direct switching from the limit-cycle oscillations to the “apoptotic” steady state is enabled by the existence of a subcritical Neimark—Sacker bifurcation in which the limit cycle loses its stability by merging with an unstable invariant torus. Our analysis provides an explanation why cancer cell lines known to have vastly diverse expression levels of Wip1 and PTEN exhibit a broad spectrum of responses to DNA damage: from a fast transition to a high level of p53 killer (a p53 phosphoform which promotes commitment to apoptosis) in cells characterized by high PTEN and low Wip1 levels to long-lasting p53 level oscillations in cells having PTEN promoter methylated (as in, e.g., MCF-7 cell line). Cancers are diseases of signaling networks. Transcription factor p53 is a pivotal node of a network that integrates a variety of stress signals and governs critical processes of DNA repair, cell cycle arrest, and apoptosis. Somewhat paradoxically, despite the fact that carcinogenesis is prevalently caused by p53 network malfunction, most of our knowledge about p53 signaling is based on cancer or immortalized cell lines. In this paper, we construct a mathematical model of intact p53 network to understand dynamics of non-cancerous cells and then dynamics of cancerous cells by introducing perturbations to the regulatory system. Cell fate decisions are enabled by the presence of interlinked feedback loops which give rise to a rich repertoire of behaviors. We explain and analyze by means of numerical simulations how the dynamical structure of the regulatory system allows for generating unambiguous single-cell fate decisions, also in the case when the cell population splits into an apoptotic and a surviving subpopulation. Perturbation analysis provides an explanation why cancer cell lines known to have vastly diverse expression levels of p53 regulators can exhibit a broad spectrum of responses to DNA damage.
Collapse
Affiliation(s)
- Beata Hat
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Marek Kochańczyk
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Marta N. Bogdał
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Tomasz Lipniacki
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
- Department of Statistics, Rice University, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
44
|
Zhao M, Zhang H, Zhu G, Liang J, Chen N, Yang Y, Liang X, Cai H, Liu W. Association between overexpression of Wip1 and prognosis of patients with non-small cell lung cancer. Oncol Lett 2016; 11:2365-2370. [PMID: 27073481 PMCID: PMC4812323 DOI: 10.3892/ol.2016.4245] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 01/28/2016] [Indexed: 01/07/2023] Open
Abstract
Wild-type p53-induced phosphatase 1 (Wip1), also termed PPM1D, is a member of the protein phosphatase 2C family, which is characterized by distinctive oncogenic properties. Overexpression of Wip1 is observed in certain types of human tumors that are associated with significantly poor prognosis. The present study aimed to detect the expression of Wip1 in non-small cell lung cancer (NSCLC) and to analyze its prognostic value in such patients. The protein expression level of Wip1 was compared between NSCLC and normal lung tissue specimens using by immunohistochemistry, and it was found that Wip1 was highly expressed in NSCLCs but was absent or weakly expressed in normal lung tissues. Detailed clinical and demographic information of patients were retrospectively collected pre- and postoperatively, and Kaplan-Meier survival and Cox's regression analyses were performed to evaluate the prognosis of patients. Survival analysis revealed that the overall survival rate for patients in the Wip1-positive expression group was significantly lower than that of the Wip1-negative group, and Cox multivariate analysis indicated that positive Wip1 expression, pN classification and pathological stage were significant prognostic predictors. The results of the current study suggest that Wip1 may be associated with pathological diagnosis and prognostic evaluation of NSCLC.
Collapse
Affiliation(s)
- Min Zhao
- Department of Oncology, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China; Department of Lung Oncology, Hebei Chest Hospital, Research Center of Hebei Lung Cancer Prevention and Treatment, Shijiazhuang, Hebei 050041, P.R. China
| | - Hongbin Zhang
- Department of Lung Oncology, Hebei Chest Hospital, Research Center of Hebei Lung Cancer Prevention and Treatment, Shijiazhuang, Hebei 050041, P.R. China
| | - Guiyun Zhu
- Department of Pathology, Hebei Chest Hospital, Research Center of Hebei Lung Cancer Prevention and Treatment, Shijiazhuang, Hebei 050041, P.R. China
| | - Jian Liang
- Department of Lung Oncology, Hebei Chest Hospital, Research Center of Hebei Lung Cancer Prevention and Treatment, Shijiazhuang, Hebei 050041, P.R. China
| | - Ning Chen
- Department of Pathology, Hebei Chest Hospital, Research Center of Hebei Lung Cancer Prevention and Treatment, Shijiazhuang, Hebei 050041, P.R. China
| | - Yonghui Yang
- Department of Pathology, Hebei Chest Hospital, Research Center of Hebei Lung Cancer Prevention and Treatment, Shijiazhuang, Hebei 050041, P.R. China
| | - Xiangcun Liang
- Department of Lung Oncology, Hebei Chest Hospital, Research Center of Hebei Lung Cancer Prevention and Treatment, Shijiazhuang, Hebei 050041, P.R. China
| | - Hongmei Cai
- Department of Lung Oncology, Hebei Chest Hospital, Research Center of Hebei Lung Cancer Prevention and Treatment, Shijiazhuang, Hebei 050041, P.R. China
| | - Wei Liu
- Department of Oncology, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China; Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050012, P.R. China
| |
Collapse
|
45
|
Esfandiari A, Hawthorne TA, Nakjang S, Lunec J. Chemical Inhibition of Wild-Type p53-Induced Phosphatase 1 (WIP1/PPM1D) by GSK2830371 Potentiates the Sensitivity to MDM2 Inhibitors in a p53-Dependent Manner. Mol Cancer Ther 2016; 15:379-91. [PMID: 26832796 DOI: 10.1158/1535-7163.mct-15-0651] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 12/23/2015] [Indexed: 01/10/2023]
Abstract
Sensitivity to MDM2 inhibitors is widely different among responsive TP53 wild-type cell lines and tumors. Understanding the determinants of MDM2 inhibitor sensitivity is pertinent for their optimal clinical application. Wild-type p53-inducible phosphatase-1 (WIP1) encoded by PPM1D, is activated, gained/amplified in a range of TP53 wild-type malignancies, and is involved in p53 stress response homeostasis. We investigated cellular growth/proliferation of TP53 wild-type and matched mutant/null cell line pairs, differing in PPM1D genetic status, in response to Nutlin-3/RG7388 ± a highly selective WIP1 inhibitor, GSK2830371. We also assessed the effects of GSK2830371 on MDM2 inhibitor-induced p53(Ser15) phosphorylation, p53-mediated global transcriptional activity, and apoptosis. The investigated cell line pairs were relatively insensitive to single-agent GSK2830371. However, a non-growth-inhibitory dose of GSK2830371 markedly potentiated the response to MDM2 inhibitors in TP53 wild-type cell lines, most notably in those harboring PPM1D-activating mutations or copy number gain (up to 5.8-fold decrease in GI50). Potentiation also correlated with significant increase in MDM2 inhibitor-induced cell death endpoints that were preceded by a marked increase in a WIP1 negatively regulated substrate, phosphorylated p53(Ser15), known to increase p53 transcriptional activity. Microarray-based gene expression analysis showed that the combination treatment increases the subset of early RG7388-induced p53 transcriptional target genes. These findings demonstrate that potent and selective WIP1 inhibition potentiates the response to MDM2 inhibitors in TP53 wild-type cells, particularly those with PPM1D activation or gain, while highlighting the mechanistic importance of p53(Ser15) and its potential use as a biomarker for response to this combination regimen.
Collapse
Affiliation(s)
- Arman Esfandiari
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Thomas A Hawthorne
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Sirintra Nakjang
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom. Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John Lunec
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
46
|
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: 0.5] [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
|
47
|
Wu B, Guo BM, Kang J, Deng XZ, Fan YB, Zhang XP, Ai KX. PPM1D exerts its oncogenic properties in human pancreatic cancer through multiple mechanisms. Apoptosis 2015; 21:365-78. [DOI: 10.1007/s10495-015-1211-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
48
|
Abstract
Our understanding of medulloblastoma biology has increased dramatically over the past decade, in part a result of the recognition that there exists tremendous intertumoral heterogeneity not apparent by morphology alone. A particular area that significantly changed our approach to medulloblastoma has been an increased understanding of the role of p53. A role for p53 in medulloblastoma has been established over the past 20 years, however, not until recently has its significance been identified. Recent developments in the understanding of intertumor heterogeneity has clarified the role of TP53 mutations, as the importance of TP53 mutations is highly dependent on the molecular subgroup of medulloblastoma, with TP53 mutant Sonic Hedgehog medulloblastomas forming an extremely high-risk group of patients. As such, there is now a tremendous push to understand the role that p53 plays in treatment resistance of medulloblastoma. In this review, we will summarize the current understanding of p53 in medulloblastoma drawn primarily from recent advances in integrated genomics.
Collapse
Affiliation(s)
- Vijay Ramaswamy
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A1, Canada Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Carolina Nör
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada Division of Neurosurgery, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Michael D Taylor
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A1, Canada Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada Division of Neurosurgery, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|
49
|
Wang H, Zhang X, Teng L, Legerski RJ. DNA damage checkpoint recovery and cancer development. Exp Cell Res 2015; 334:350-8. [PMID: 25842165 DOI: 10.1016/j.yexcr.2015.03.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/12/2015] [Accepted: 03/14/2015] [Indexed: 12/21/2022]
Abstract
Cell cycle checkpoints were initially presumed to function as a regulator of cell cycle machinery in response to different genotoxic stresses, and later found to play an important role in the process of tumorigenesis by acting as a guard against DNA over-replication. As a counterpart of checkpoint activation, the checkpoint recovery machinery is working in opposition, aiming to reverse the checkpoint activation and resume the normal cell cycle. The DNA damage response (DDR) and oncogene induced senescence (OIS) are frequently found in precancerous lesions, and believed to constitute a barrier to tumorigenesis, however, the DDR and OIS have been observed to be diminished in advanced cancers of most tissue origins. These findings suggest that when progressing from pre-neoplastic lesions to cancer, DNA damage checkpoint barriers are overridden. How the DDR checkpoint is bypassed in this process remains largely unknown. Activated cytokine and growth factor-signaling pathways were very recently shown to suppress the DDR and to promote uncontrolled cell proliferation in the context of oncovirus infection. In recent decades, data from cell line and tumor models showed that a group of checkpoint recovery proteins function in promoting tumor progression; data from patient samples also showed overexpression of checkpoint recovery proteins in human cancer tissues and a correlation with patients׳ poor prognosis. In this review, the known cell cycle checkpoint recovery proteins and their roles in DNA damage checkpoint recovery are reviewed, as well as their implications in cancer development. This review also provides insight into the mechanism by which the DDR suppresses oncogene-driven tumorigenesis and tumor progression.
Collapse
Affiliation(s)
- Haiyong Wang
- First affiliated hospital, Zhejiang University, School of medicine, Cancer Center, 79 Qingchun Road, Hangzhou 310003, China
| | - Xiaoshan Zhang
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Department of Genetics Unit 1010, 1515 Holcombe Blvd. Houston, TX 77030 USA
| | - Lisong Teng
- First affiliated hospital, Zhejiang University, School of medicine, Cancer Center, 79 Qingchun Road, Hangzhou 310003, China.
| | - Randy J Legerski
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Department of Genetics Unit 1010, 1515 Holcombe Blvd. Houston, TX 77030 USA.
| |
Collapse
|
50
|
Cooperative integration between HEDGEHOG-GLI signalling and other oncogenic pathways: implications for cancer therapy. Expert Rev Mol Med 2015; 17:e5. [PMID: 25660620 PMCID: PMC4836208 DOI: 10.1017/erm.2015.3] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The HEDGEHOG-GLI (HH-GLI) signalling is a key pathway critical in embryonic development, stem cell biology and tissue homeostasis. In recent years, aberrant activation of HH-GLI signalling has been linked to several types of cancer, including those of the skin, brain, lungs, prostate, gastrointestinal tract and blood. HH-GLI signalling is initiated by binding of HH ligands to the transmembrane receptor PATCHED and is mediated by transcriptional effectors that belong to the GLI family, whose activity is finely tuned by a number of molecular interactions and post-translation modifications. Several reports suggest that the activity of the GLI proteins is regulated by several proliferative and oncogenic inputs, in addition or independent of upstream HH signalling. The identification of this complex crosstalk and the understanding of how the major oncogenic signalling pathways interact in cancer is a crucial step towards the establishment of efficient targeted combinatorial treatments. Here we review recent findings on the cooperative integration of HH-GLI signalling with the major oncogenic inputs and we discuss how these cues modulate the activity of the GLI proteins in cancer. We then summarise the latest advances on SMO and GLI inhibitors and alternative approaches to attenuate HH signalling through rational combinatorial therapies.
Collapse
|