151
|
The MDM2-p53 pathway is involved in preconditioning-induced neuronal tolerance to ischemia. Sci Rep 2018; 8:1610. [PMID: 29371613 PMCID: PMC5785500 DOI: 10.1038/s41598-018-19921-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 01/10/2018] [Indexed: 01/01/2023] Open
Abstract
Brain preconditioning (PC) refers to a state of transient tolerance against a lethal insult that can be evoked by a prior mild event. It is thought that PC may induce different pathways responsible for neuroprotection, which may involve the attenuation of cell damage pathways, including the apoptotic cell death. In this context, p53 is a stress sensor that accumulates during brain ischemia leading to neuronal death. The murine double minute 2 gene (MDM2), a p53-specific E3 ubiquitin ligase, is the main cellular antagonist of p53, mediating its degradation by the proteasome. Here, we study the role of MDM2-p53 pathway on PC-induced neuroprotection both in cultured neurons (in vitro) and rat brain (in vivo). Our results show that PC increased neuronal MDM2 protein levels, which prevented ischemia-induced p53 stabilization and neuronal death. Indeed, PC attenuated ischemia-induced activation of the p53/PUMA/caspase-3 signaling pathway. Pharmacological inhibition of MDM2-p53 interaction in neurons abrogated PC-induced neuroprotection against ischemia. Finally, the relevance of the MDM2-p53 pathway was confirmed in rat brain using a PC model in vivo. These findings demonstrate the key role of the MDM2-p53 pathway in PC-induced neuroprotection against a subsequent ischemic insult and poses MDM2 as an essential target in ischemic tolerance.
Collapse
|
152
|
Affiliation(s)
- Francesca Pentimalli
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori – IRCCS -Fondazione G, Pascale, Naples, Italy
| |
Collapse
|
153
|
Chopra H, Khan Z, Contreras J, Wang H, Sedrak A, Zhu Y. Activation of p53 and destabilization of androgen receptor by combinatorial inhibition of MDM2 and MDMX in prostate cancer cells. Oncotarget 2017; 9:6270-6281. [PMID: 29464071 PMCID: PMC5814211 DOI: 10.18632/oncotarget.23569] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 10/13/2017] [Indexed: 01/22/2023] Open
Abstract
Castration-resistant prostate cancer (CRPC) frequently develops after initial standard radiation and androgen deprivation therapy, leaving patients with limited further treatment options. Androgen receptor (AR) is a transcription factor that plays a key role in the initiation and progression of prostate cancer. p53, a major tumor suppressor that is rarely mutated in early-stages of prostate cancer, is often deregulated during prostate cancer progression. Here, we report an unusual co-amplification of MDM2 and MDMX, two crucial negative regulators of p53, in CRPC datasets. We demonstrate that combinatorial inhibition of MDM2 and MDMX, with nutlin-3 and NSC207895 respectively, has a profound inhibitory effect on cell proliferation of androgen-responsive, wild-type TP53 gene carrying prostate cancer cells LNCaP and 22Rv1. We further show that the combinatorial inhibition of MDM2 and MDMX not only activates p53, but also decreases cellular levels of AR and represses its function. Additionally, co-expression of MDM2 and MDMX stabilizes AR. Together, our results indicate that combinatorial inhibition of MDM2 and MDMX may offer a novel compelling strategy for prostate cancer therapy.
Collapse
Affiliation(s)
- Harman Chopra
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Zara Khan
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Jamie Contreras
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Herui Wang
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Abanob Sedrak
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Yan Zhu
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| |
Collapse
|
154
|
Gansmo LB, Vatten L, Romundstad P, Hveem K, Ryan BM, Harris CC, Knappskog S, Lønning PE. Associations between the MDM2 promoter P1 polymorphism del1518 (rs3730485) and incidence of cancer of the breast, lung, colon and prostate. Oncotarget 2017; 7:28637-46. [PMID: 27081698 PMCID: PMC5053751 DOI: 10.18632/oncotarget.8705] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/28/2016] [Indexed: 12/11/2022] Open
Abstract
The MDM2 promoter region contains several polymorphisms, some of which have been associated with MDM2 expression, cancer risk and age at cancer onset. del1518 (rs3730485) is an indel polymorphism residing in the MDM2 promoter P1 and is in almost complete linkage disequilibrium with the MDM2 promoter P2 polymorphism SNP309T>G (rs2279744). Cancer risk assessments of del1518 have previously been conducted in relatively small Chinese populations only. In this study we assessed the genotype distribution of del1518 among healthy Caucasians, African Americans and Chinese, and we estimated the Odds Ratios (OR) for incident cancer of the breast, colon, lung and prostate (n=7,081) as compared to controls (n=3,749) in a large Caucasian (Norwegian) cohort.We found the genotypes of the del1518 to vary significantly between healthy Caucasians, African-Americans and Chinese (p< 1×10-5). Further, we found a positive association of the del1518 del-allele with risk of colon cancer (dominant model: OR = 1.15; 95 % CI = 1.01 - 1.31). Stratifying according to SNP309 status, this association remained among carriers of the SNP309TG genotype (OR = 1.21; 95 % CI = 1.01 - 1.46), but with no clear association among carriers of the SNP309TT genotype. In conclusion, our findings suggest del1518 to be associated with increased risk of colon cancer.
Collapse
Affiliation(s)
- Liv B Gansmo
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Lars Vatten
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Pål Romundstad
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristian Hveem
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bríd M Ryan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Stian Knappskog
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Per E Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
155
|
Garcia-Alonso L, Iorio F, Matchan A, Fonseca N, Jaaks P, Peat G, Pignatelli M, Falcone F, Benes CH, Dunham I, Bignell G, McDade SS, Garnett MJ, Saez-Rodriguez J. Transcription Factor Activities Enhance Markers of Drug Sensitivity in Cancer. Cancer Res 2017; 78:769-780. [PMID: 29229604 DOI: 10.1158/0008-5472.can-17-1679] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/16/2017] [Accepted: 12/04/2017] [Indexed: 12/12/2022]
Abstract
Transcriptional dysregulation induced by aberrant transcription factors (TF) is a key feature of cancer, but its global influence on drug sensitivity has not been examined. Here, we infer the transcriptional activity of 127 TFs through analysis of RNA-seq gene expression data newly generated for 448 cancer cell lines, combined with publicly available datasets to survey a total of 1,056 cancer cell lines and 9,250 primary tumors. Predicted TF activities are supported by their agreement with independent shRNA essentiality profiles and homozygous gene deletions, and recapitulate mutant-specific mechanisms of transcriptional dysregulation in cancer. By analyzing cell line responses to 265 compounds, we uncovered numerous TFs whose activity interacts with anticancer drugs. Importantly, combining existing pharmacogenomic markers with TF activities often improves the stratification of cell lines in response to drug treatment. Our results, which can be queried freely at dorothea.opentargets.io, offer a broad foundation for discovering opportunities to refine personalized cancer therapies.Significance: Systematic analysis of transcriptional dysregulation in cancer cell lines and patient tumor specimens offers a publicly searchable foundation to discover new opportunities to refine personalized cancer therapies. Cancer Res; 78(3); 769-80. ©2017 AACR.
Collapse
Affiliation(s)
- Luz Garcia-Alonso
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.,OpenTargets, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Francesco Iorio
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.,OpenTargets, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Angela Matchan
- OpenTargets, Wellcome Genome Campus, Cambridge, United Kingdom.,Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Nuno Fonseca
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Patricia Jaaks
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Gareth Peat
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.,OpenTargets, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Miguel Pignatelli
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.,OpenTargets, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Fiammetta Falcone
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Cyril H Benes
- Massachusetts General Hospital, Boston, Massachusetts
| | - Ian Dunham
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom.,OpenTargets, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Graham Bignell
- OpenTargets, Wellcome Genome Campus, Cambridge, United Kingdom.,Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Simon S McDade
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Mathew J Garnett
- OpenTargets, Wellcome Genome Campus, Cambridge, United Kingdom.,Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom. .,OpenTargets, Wellcome Genome Campus, Cambridge, United Kingdom.,Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| |
Collapse
|
156
|
Giono LE, Resnick-Silverman L, Carvajal LA, St Clair S, Manfredi JJ. Mdm2 promotes Cdc25C protein degradation and delays cell cycle progression through the G2/M phase. Oncogene 2017; 36:6762-6773. [PMID: 28806397 PMCID: PMC6002854 DOI: 10.1038/onc.2017.254] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 06/15/2017] [Accepted: 06/27/2017] [Indexed: 12/16/2022]
Abstract
Upon different types of stress, the gene encoding the mitosis-promoting phosphatase Cdc25C is transcriptionally repressed by p53, contributing to p53's enforcement of a G2 cell cycle arrest. In addition, Cdc25C protein stability is also decreased following DNA damage. Mdm2, another p53 target gene, encodes a ubiquitin ligase that negatively regulates p53 levels by ubiquitination. Ablation of Mdm2 by siRNA led to an increase in p53 protein and repression of Cdc25C gene expression. However, Cdc25C protein levels were actually increased following Mdm2 depletion. Mdm2 is shown to negatively regulate Cdc25C protein levels by reducing its half-life independently of the presence of p53. Further, Mdm2 physically interacts with Cdc25C and promotes its degradation through the proteasome in a ubiquitin-independent manner. Either Mdm2 overexpression or Cdc25C downregulation delays cell cycle progression through the G2/M phase. Thus, the repression of the Cdc25C promoter by p53, together with p53-dependent induction of Mdm2 and subsequent degradation of Cdc25C, could provide a dual mechanism by which p53 can enforce and maintain a G2/M cell cycle arrest.
Collapse
Affiliation(s)
- L E Giono
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L Resnick-Silverman
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L A Carvajal
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - S St Clair
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J J Manfredi
- Department of Oncological Sciences and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
157
|
Kwon DH, Kim YK, Kook H. New Aspects of Vascular Calcification: Histone Deacetylases and Beyond. J Korean Med Sci 2017; 32:1738-1748. [PMID: 28960024 PMCID: PMC5639052 DOI: 10.3346/jkms.2017.32.11.1738] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/19/2017] [Indexed: 11/20/2022] Open
Abstract
Vascular calcification is a pathologic phenomenon in which calcium phosphate is ectopically deposited in the arteries. Previously, calcification was considered to be a passive process in response to metabolic diseases, vascular or valvular diseases, or even aging. However, now calcification is recognized as a highly-regulated consequence, like bone formation, and many clinical trials have been carried out to elucidate the correlation between vascular calcification and cardiovascular events and mortality. As a result, vascular calcification has been implicated as an independent risk factor in cardiovascular diseases. Many molecules are now known to be actively associated with this process. Recently, our laboratory found that posttranslational modification of histone deacetylase (HDAC) 1 is actively involved in the development of vascular calcification. In addition, we found that modulation of the activity of HDAC as well as its protein stability by MDM2, an HDAC1-E3 ligase, may be a therapeutic target in vascular calcification. In the present review, we overview the pathomechanism of vascular calcification and the involvement of posttranslational modification of epigenetic regulators.
Collapse
Affiliation(s)
- Duk Hwa Kwon
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
- Basic Research Laboratory for Cardiac Remodeling, Chonnam National University Medical School, Gwangju, Korea
| | - Young Kook Kim
- Basic Research Laboratory for Cardiac Remodeling, Chonnam National University Medical School, Gwangju, Korea
- Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
- Basic Research Laboratory for Cardiac Remodeling, Chonnam National University Medical School, Gwangju, Korea.
| |
Collapse
|
158
|
Zhou G, Duan Y, Ma G, Wu W, Hu Z, Chen N, Chee Y, Cui J, Samad A, Matsubara JA, Mukai S, D'Amore PA, Lei H. Introduction of the MDM2 T309G Mutation in Primary Human Retinal Epithelial Cells Enhances Experimental Proliferative Vitreoretinopathy. Invest Ophthalmol Vis Sci 2017; 58:5361-5367. [PMID: 29049737 PMCID: PMC5649510 DOI: 10.1167/iovs.17-22045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Purpose The murine double minute (MDM)2 is a critical negative regulator of the p53 tumor suppressor, and MDM2 SNP309G is associated with a higher risk of proliferative vitreoretinopathy (PVR); in addition, the MDM2 T309G created using clustered regularly interspaced short palindromic repeats (CRISPR)/associated endonuclease (Cas)9 enhances normal rabbit vitreous-induced expression of MDM2 and survival of primary human retinal pigment epithelial (hRPE) cells in vitro. The goal of this study was to determine whether this MDM2 T309G contributes to the development of experimental PVR. Methods hRPE cells expressing MDM2 T309G or T309T only were treated with vitreous from human PVR donors (HV). The expression of MDM2 and p53 in the treated cells was examined by Western blot. The in vitro vitreous-induced cellular responses, such as contraction were assessed, and PVR was induced by intravitreal injection of the hRPE cells with MDM2 T309G or T309T only into rabbit eyes. Results Western blot analyses indicated that treatment of hRPE cells with HV led to a significant increase (1.7 ± 0.2-fold) in the expression of MDM2 and a significant decrease in p53 in the cells expressing MDM2 T309G compared with those with MDM2 T309T. In addition, HV promoted contraction of the hRPE cells expressing MDM2 T309G significantly more than those with MDM2 T309T only. Furthermore, MDM2 T309G in the hRPE cells enhanced the development of PVR in a rabbit model. Conclusions The MDM2 SNP309 in RPE cells enhances their potential of PVR pathogenesis.
Collapse
Affiliation(s)
- Guohong Zhou
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States.,Shanxi Eye Hospital, Taiyuan City, Shanxi Province, China
| | - Yajiang Duan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States.,Shanxi Eye Hospital, Taiyuan City, Shanxi Province, China
| | - Gaoen Ma
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States.,Department of Ophthalmology, the Third Affiliated Hospital of Xinxiang Medical University, Eye Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Wenyi Wu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Na Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Yewlin Chee
- Massachusetts Eye and Ear Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Jing Cui
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Arif Samad
- Dalhousie University, Halifax, Nova Scotia, Canada
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shizuo Mukai
- Massachusetts Eye and Ear Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Hetian Lei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
159
|
Saadatzadeh MR, Elmi AN, Pandya PH, Bijangi-Vishehsaraei K, Ding J, Stamatkin CW, Cohen-Gadol AA, Pollok KE. The Role of MDM2 in Promoting Genome Stability versus Instability. Int J Mol Sci 2017; 18:ijms18102216. [PMID: 29065514 PMCID: PMC5666895 DOI: 10.3390/ijms18102216] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/06/2017] [Accepted: 10/11/2017] [Indexed: 02/07/2023] Open
Abstract
In cancer, the mouse double minute 2 (MDM2) is an oncoprotein that contributes to the promotion of cell growth, survival, invasion, and therapeutic resistance. The impact of MDM2 on cell survival versus cell death is complex and dependent on levels of MDM2 isoforms, p53 status, and cellular context. Extensive investigations have demonstrated that MDM2 protein–protein interactions with p53 and other p53 family members (p63 and p73) block their ability to function as transcription factors that regulate cell growth and survival. Upon genotoxic insults, a dynamic and intricately regulated DNA damage response circuitry is activated leading to release of p53 from MDM2 and activation of cell cycle arrest. What ensues following DNA damage, depends on the extent of DNA damage and if the cell has sufficient DNA repair capacity. The well-known auto-regulatory loop between p53-MDM2 provides an additional layer of control as the cell either repairs DNA damage and survives (i.e., MDM2 re-engages with p53), or undergoes cell death (i.e., MDM2 does not re-engage p53). Furthermore, the decision to live or die is also influenced by chromatin-localized MDM2 which directly interacts with the Mre11-Rad50-Nbs1 complex and inhibits DNA damage-sensing giving rise to the potential for increased genome instability and cellular transformation.
Collapse
Affiliation(s)
- M Reza Saadatzadeh
- Department of Pediatrics (Division of Hematology/Oncology), Indianapolis, IN 46202, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University Simon Cancer Center, 1044 West Walnut Street R4 302, Indianapolis, IN 46202-5525, USA.
| | - Adily N Elmi
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Pankita H Pandya
- Department of Pediatrics (Division of Hematology/Oncology), Indianapolis, IN 46202, USA.
| | | | - Jixin Ding
- Department of Pediatrics (Division of Hematology/Oncology), Indianapolis, IN 46202, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University Simon Cancer Center, 1044 West Walnut Street R4 302, Indianapolis, IN 46202-5525, USA.
| | - Christopher W Stamatkin
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University Simon Cancer Center, 1044 West Walnut Street R4 302, Indianapolis, IN 46202-5525, USA.
| | | | - Karen E Pollok
- Department of Pediatrics (Division of Hematology/Oncology), Indianapolis, IN 46202, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University Simon Cancer Center, 1044 West Walnut Street R4 302, Indianapolis, IN 46202-5525, USA.
| |
Collapse
|
160
|
Therapeutics targeting Bcl-2 in hematological malignancies. Biochem J 2017; 474:3643-3657. [PMID: 29061914 DOI: 10.1042/bcj20170080] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/29/2017] [Accepted: 09/19/2017] [Indexed: 12/25/2022]
Abstract
Members of the B-cell lymphoma 2 (BCL-2) gene family are attractive targets for cancer therapy as they play a key role in promoting cell survival, a long-since established hallmark of cancer. Clinical utility for selective inhibition of specific anti-apoptotic Bcl-2 family proteins has recently been realized with the Food and Drug Administration (FDA) approval of venetoclax (formerly ABT-199/GDC-0199) in relapsed chronic lymphocytic leukemia (CLL) with 17p deletion. Despite the impressive monotherapy activity in CLL, such responses have rarely been observed in other B-cell malignancies, and preclinical data suggest that combination therapies will be needed in other indications. Additional selective antagonists of Bcl-2 family members, including Bcl-XL and Mcl-1, are in various stages of preclinical and clinical development and hold the promise of extending clinical utility beyond CLL and overcoming resistance to venetoclax. In addition to direct targeting of Bcl-2 family proteins with BH3 mimetics, combination therapies that aim at down-regulating expression of anti-apoptotic BCL-2 family members or restoring expression of pro-apoptotic BH3 family proteins may provide a means to deepen responses to venetoclax and extend the utility to additional indications. Here, we review recent progress in direct and selective targeting of Bcl-2 family proteins for cancer therapy and the search for rationale combinations.
Collapse
|
161
|
Singh AK, Chauhan SS, Singh SK, Verma VV, Singh A, Arya RK, Maheshwari S, Akhtar MS, Sarkar J, Rangnekar VM, Chauhan PMS, Datta D. Dual targeting of MDM2 with a novel small-molecule inhibitor overcomes TRAIL resistance in cancer. Carcinogenesis 2017; 37:1027-1040. [PMID: 27543608 DOI: 10.1093/carcin/bgw088] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/16/2016] [Indexed: 01/25/2023] Open
Abstract
Mouse double minute 2 (MDM2) protein functionally inactivates the tumor suppressor p53 in human cancer. Conventional MDM2 inhibitors provide limited clinical application as they interfere only with the MDM2-p53 interaction to release p53 from MDM2 sequestration but do not prevent activated p53 from transcriptionally inducing MDM2 expression. Here, we report a rationally synthesized chalcone-based pyrido[ b ]indole, CPI-7c, as a unique small-molecule inhibitor of MDM2, which not only inhibited MDM2-p53 interaction but also promoted MDM2 degradation. CPI-7c bound to both RING and N-terminal domains of MDM2 to promote its ubiquitin-mediated degradation and p53 stabilization. CPI-7c-induced p53 directly recruited to the promoters of DR4 and DR5 genes and enhanced their expression, resulting in sensitization of TNF-related apoptosis-inducing ligand (TRAIL)-resistant cancer cells toward TRAIL-induced apoptosis. Collectively, we identified CPI-7c as a novel small-molecule inhibitor of MDM2 with a unique two-prong mechanism of action that sensitized TRAIL-resistant cancer cells to apoptosis by modulating the MDM2-p53-DR4/DR5 pathway.
Collapse
Affiliation(s)
| | - Shikha S Chauhan
- Medicinal and Process Chemistry Division and.,Present address: Pennsylvania State University, University Park, PA 16801, USA
| | - Sudhir Kumar Singh
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute , Lucknow, Uttar Pradesh 226031 , India
| | - Ved Vrat Verma
- Department of Biophysics, Delhi University , South Campus, New Delhi 110021 , India
| | | | | | - Shrankhla Maheshwari
- Biochemistry Division.,Academy of Scientific and Innovative Research, New Delhi 110025, India and
| | - Md Sohail Akhtar
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute , Lucknow, Uttar Pradesh 226031 , India
| | | | - Vivek M Rangnekar
- Department of Radiation Medicine and Markey Cancer Center, University of Kentucky , Lexington, KY 40536 , USA and
| | | | - Dipak Datta
- Biochemistry Division.,Academy of Scientific and Innovative Research, New Delhi 110025, India and
| |
Collapse
|
162
|
Leroy B, Ballinger ML, Baran-Marszak F, Bond GL, Braithwaite A, Concin N, Donehower LA, El-Deiry WS, Fenaux P, Gaidano G, Langerød A, Hellstrom-Lindberg E, Iggo R, Lehmann-Che J, Mai PL, Malkin D, Moll UM, Myers JN, Nichols KE, Pospisilova S, Ashton-Prolla P, Rossi D, Savage SA, Strong LC, Tonin PN, Zeillinger R, Zenz T, Fraumeni JF, Taschner PEM, Hainaut P, Soussi T. Recommended Guidelines for Validation, Quality Control, and Reporting of TP53 Variants in Clinical Practice. Cancer Res 2017; 77:1250-1260. [PMID: 28254861 DOI: 10.1158/0008-5472.can-16-2179] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/12/2016] [Accepted: 11/16/2016] [Indexed: 12/21/2022]
Abstract
Accurate assessment of TP53 gene status in sporadic tumors and in the germline of individuals at high risk of cancer due to Li-Fraumeni Syndrome (LFS) has important clinical implications for diagnosis, surveillance, and therapy. Genomic data from more than 20,000 cancer genomes provide a wealth of information on cancer gene alterations and have confirmed TP53 as the most commonly mutated gene in human cancer. Analysis of a database of 70,000 TP53 variants reveals that the two newly discovered exons of the gene, exons 9β and 9γ, generated by alternative splicing, are the targets of inactivating mutation events in breast, liver, and head and neck tumors. Furthermore, germline rearrange-ments in intron 1 of TP53 are associated with LFS and are frequently observed in sporadic osteosarcoma. In this context of constantly growing genomic data, we discuss how screening strategies must be improved when assessing TP53 status in clinical samples. Finally, we discuss how TP53 alterations should be described by using accurate nomenclature to avoid confusion in scientific and clinical reports. Cancer Res; 77(6); 1250-60. ©2017 AACR.
Collapse
Affiliation(s)
- Bernard Leroy
- Sorbonne Université, UPMC Univ Paris 06, Paris, France
| | - Mandy L Ballinger
- Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Fanny Baran-Marszak
- Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'H ematologie Biologique, France
| | - Gareth L Bond
- Ludwig Institute for Cancer Research, University of Oxford, Nuffield Department of Clinical Medicine, Old Road Campus Research Building, Oxford, United Kingdom
| | - Antony Braithwaite
- Dept of Pathology, School of Medicine, University of Otago, Dunedin, New Zealand.,Children's Medical Research Institute, University of Sydney, Westmead NSW, Australia
| | - Nicole Concin
- Department of Gynecology and Obstetrics, Innsbruck Medical University, Innsbruck, Austria
| | | | - Wafik S El-Deiry
- Department of Hematology/Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Pierre Fenaux
- Service d'hématologie séniors, Hôpital St Louis/Université Paris 7, 1 avenue Claude Vellefaux, Paris, France
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Anita Langerød
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Eva Hellstrom-Lindberg
- Karolinska Institute, Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Richard Iggo
- Bergonié Cancer Institute University of Bordeaux 229 cours de l'Argonne 33076 Bordeaux, France
| | | | - Phuong L Mai
- Cancer Genetics Program, Magee Womens Hospital, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - David Malkin
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, New York
| | - Jeffrey N Myers
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kim E Nichols
- Department of Oncology, Division of Cancer Predisposition, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sarka Pospisilova
- Masaryk University, CEITEC - Molecular Medicine and University Hospital Brno, Department of Internal Medicine - Hematology and Oncology, Brno, Czech Republic
| | - Patricia Ashton-Prolla
- Universidade Federal do Rio Grande do Sul (UFRGS) e Serviço deGenética Médica-HCPA, Rua Ramiro Barcelos, Porto Alegre, Brasil
| | - Davide Rossi
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Louise C Strong
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia N Tonin
- Departments of Medicine and Human Genetics, McGill University and Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Robert Zeillinger
- Molecular Oncology Group, Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Thorsten Zenz
- University of Heidelberg, Department of Medicine V, Heidelberg, Germany; Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Joseph F Fraumeni
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Peter E M Taschner
- Generade Centre of Expertise Genomics and University of Applied Sciences Leiden, Leiden, the Netherlands
| | - Pierre Hainaut
- Institut Albert Bonniot, Inserm 823, Université Grenoble Alpes, Rond Point de la Chantourne, La Tronche, France
| | - Thierry Soussi
- Sorbonne Université, UPMC Univ Paris 06, Paris, France. .,Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden.,INSERM, U1138, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
163
|
Co-operative intra-protein structural response due to protein–protein complexation revealed through thermodynamic quantification: study of MDM2-p53 binding. J Comput Aided Mol Des 2017; 31:891-903. [DOI: 10.1007/s10822-017-0057-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/30/2017] [Indexed: 12/29/2022]
|
164
|
Yang Z, Maciejowski J, de Lange T. Nuclear Envelope Rupture Is Enhanced by Loss of p53 or Rb. Mol Cancer Res 2017; 15:1579-1586. [PMID: 28811362 DOI: 10.1158/1541-7786.mcr-17-0084] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 07/05/2017] [Accepted: 08/11/2017] [Indexed: 01/08/2023]
Abstract
The mammalian nuclear envelope (NE) forms a stable physical barrier between the nucleus and the cytoplasm, normally breaking down only during mitosis. However, spontaneous transient NE rupture in interphase can occur when NE integrity is compromised, such as when the nucleus experiences mechanical stress. For instance, deficiencies in the nuclear lamins and their associated proteins can cause NE rupture that is promoted by forces exerted by actin filaments. NE rupture can allow cytoplasmic nucleases to access chromatin, potentially compromising genome integrity. Importantly, spontaneous NE rupture was noted in several human cancer cell lines, but the cause of this defect is not known. Here, we investigated the mechanistic contributions of two major tumor suppressors, p53 (TP53) and Rb (RB1), to the repression of NE rupture. NE rupture was induced in normal human epithelial RPE-1 cells upon impairment of either Rb or p53 achieved by shRNA knockdown and CRISPR/Cas9 gene editing. NE rupture did not involve diminished expression of NE components or greater cell motility. However, cells that underwent NE rupture displayed a larger nuclear projection area. In conclusion, the data indicate that NE rupture in cancer cells is likely due to loss of either the Rb or the p53 pathway.Implications: These findings imply that tumor suppression by Rb and p53 includes the ability to prevent NE rupture, thereby protecting against genome alterations. Mol Cancer Res; 15(11); 1579-86. ©2017 AACR.
Collapse
Affiliation(s)
- Zhe Yang
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, New York
| | - John Maciejowski
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, New York
| | - Titia de Lange
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, New York.
| |
Collapse
|
165
|
Synergistic activity and heterogeneous acquired resistance of combined MDM2 and MEK inhibition in KRAS mutant cancers. Oncogene 2017; 36:6581-6591. [PMID: 28783173 PMCID: PMC5700857 DOI: 10.1038/onc.2017.258] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 05/03/2017] [Accepted: 06/23/2017] [Indexed: 01/10/2023]
Abstract
There are currently no effective targeted therapies for KRAS mutant cancers. Therapeutic strategies that combine MEK inhibitors with agents that target apoptotic pathways may be a promising therapeutic approach. We investigated combining MEK and MDM2 inhibitors as a potential treatment strategy for KRAS mutant non-small cell lung cancers and colorectal carcinomas that harbor wild-type TP53. The combination of pimasertib (MEK inhibitor) + SAR405838 (MDM2 inhibitor) was synergistic and induced the expression of PUMA and BIM, led to apoptosis and growth inhibition in vitro, and tumor regression in vivo. Acquired resistance to the combination commonly resulted from the acquisition of TP53 mutations, conferring complete resistance to MDM2 inhibition. In contrast, resistant clones exhibited marked variability in sensitivity to MEK inhibition, which significantly impacted sensitivity to subsequent treatment with alternative MEK inhibitor-based combination therapies. These results highlight both the potential promise and limitations of combining MEK and MDM2 inhibitors for treatment of KRAS mutant NSCLC and CRC.
Collapse
|
166
|
Antitumoral effects of γCdcPLI, a PLA 2 inhibitor from Crotalus durissus collilineatus via PI3K/Akt pathway on MDA-MB-231 breast cancer cell. Sci Rep 2017; 7:7077. [PMID: 28765552 PMCID: PMC5539153 DOI: 10.1038/s41598-017-07082-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/22/2017] [Indexed: 12/23/2022] Open
Abstract
Phospholipases A2(PLA2s) overexpression is closely associated with the malignant potential of breast cancers. Here, we showed for the first the antitumoral effects of γCdcPLI, a PLA2 inhibitor from Crotalus durissus collilineatus via PI3K/Akt pathway on MDA-MB-231 cell. Firstly, γCdcPLI was more cytotoxic to MDA-MB-231 breast cancer cells than other cell lines (MCF-7, HeLa, PC3 and A549) and did not affect the viability of non-tumorigenic breast cell (MCF 10A). In addition, γCdcPLI induced modulation of important mediators of apoptosis pathways such as p53, MAPK-ERK, BIRC5 and MDM2. γCdcPLI decreased MDA-MB-231 adhesion, migration and invasion. Interestingly, the γCdcPLI also inhibited the adhesion and migration of endothelial cells and blocked angiogenesis by inhibiting tube formation by HUVECs in vitro and sprouting elongation on aortic ring assay ex vivo. Furthermore, γCdcPLI reduced the production of vascular endothelial growth factor (VEGF). γCdcPLI was also able to decrease PGE2 levels in MDA-MB-231 and inhibited gene and protein expression of the PI3K/Akt pathway. In conclusion, γCdcPLI showed in vitro antitumoral, antimestatatic and anti-angiogenic potential effects and could be an attractive approach for futures studies in cancer therapy.
Collapse
|
167
|
Genome-wide reconstruction of complex structural variants using read clouds. Nat Methods 2017; 14:915-920. [PMID: 28714986 PMCID: PMC5578891 DOI: 10.1038/nmeth.4366] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 06/15/2017] [Indexed: 12/16/2022]
Abstract
In read cloud approaches, microfluidic partitioning of long genomic DNA fragments and barcoding of shorter fragments derived from these fragments retains long-range information in short sequencing reads. This combination of short reads with long-range information represents a powerful alternative to single-molecule long-read sequencing. We develop Genome-wide Reconstruction of Complex Structural Variants (GROC-SVs) for SV detection and assembly from read cloud data and apply this method to Illumina-sequenced 10x Genomics sarcoma and breast cancer data sets. Compared with short-fragment sequencing, GROC-SVs substantially improves the specificity of breakpoint detection at comparable sensitivity. This approach also performs sequence assembly across multiple breakpoints simultaneously, enabling the reconstruction of events exhibiting remarkable complexity. We show that chromothriptic rearrangements occurred before copy number amplifications, and that rates of single-nucleotide variants and SVs are not correlated. Our results support the use of read cloud approaches to advance the characterization of large and complex structural variation.
Collapse
|
168
|
Ma G, Duan Y, Huang X, Qian CX, Chee Y, Mukai S, Cui J, Samad A, Matsubara JA, Kazlauskas A, D'Amore PA, Gu S, Lei H. Prevention of Proliferative Vitreoretinopathy by Suppression of Phosphatidylinositol 5-Phosphate 4-Kinases. Invest Ophthalmol Vis Sci 2017; 57:3935-43. [PMID: 27472081 PMCID: PMC4974024 DOI: 10.1167/iovs.16-19405] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Previous studies have shown that vitreous stimulates degradation of the tumor suppressor protein p53 and that knockdown of phosphatidylinositol 5-phosphate 4-kinases (PI5P4Kα and -β) abrogates proliferation of p53-deficient cells. The purpose of this study was to determine whether vitreous stimulated expression of PI5P4Kα and -β and whether suppression of PI5P4Kα and -β would inhibit vitreous-induced cellular responses and experimental proliferative vitreoretinopathy (PVR). Methods PI5P4Kα and -β encoded by PIP4K2A and 2B, respectively, in human ARPE-19 cells were knocked down by stably expressing short hairpin (sh)RNA directed at human PIP4K2A and -2B. In addition, we rescued expression of PI5P4Kα and -β by re-expressing mouse PIP4K2A and -2B in the PI5P4Kα and -β knocked-down ARPE-19 cells. Expression of PI5P4Kα and -β was determined by Western blot and immunofluorescence. The following cellular responses were monitored: cell proliferation, survival, migration, and contraction. Moreover, the cell potential of inducing PVR was examined in a rabbit model of PVR effected by intravitreal cell injection. Results We found that vitreous enhanced expression of PI5P4Kα and -β in RPE cells and that knocking down PI5P4Kα and -β abrogated vitreous-stimulated cell proliferation, survival, migration, and contraction. Re-expression of mouse PIP4Kα and -β in the human PI5P4Kα and -β knocked-down cells recovered the loss of vitreous-induced cell contraction. Importantly, suppression of PI5P4Kα and -β abrogated the pathogenesis of PVR induced by intravitreal cell injection in rabbits. Moreover, we revealed that expression of PI5P4Kα and -β was abundant in epiretinal membranes from PVR grade C patients. Conclusions The findings from this study indicate that PI5P4Kα and -β could be novel therapeutic targets for the treatment of PVR.
Collapse
Affiliation(s)
- Gaoen Ma
- Schepens Eye Research Institute, Boston, Massachusetts, United States 2Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States 4Aier School of Ophthalmology
| | - Yajian Duan
- Schepens Eye Research Institute, Boston, Massachusetts, United States 2Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Xionggao Huang
- Schepens Eye Research Institute, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Cynthia X Qian
- Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Yewlin Chee
- Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Shizuo Mukai
- Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Jing Cui
- The University of British Columbia, Vancouver, British Columbia, Canada
| | - Arif Samad
- Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Andrius Kazlauskas
- Schepens Eye Research Institute, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Patricia A D'Amore
- Schepens Eye Research Institute, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Shuyan Gu
- Aier School of Ophthalmology, Central South University, Changsha, Changsha Province, China
| | - Hetian Lei
- Schepens Eye Research Institute, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
169
|
Wang S, Zhao Y, Aguilar A, Bernard D, Yang CY. Targeting the MDM2-p53 Protein-Protein Interaction for New Cancer Therapy: Progress and Challenges. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026245. [PMID: 28270530 DOI: 10.1101/cshperspect.a026245] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
MDM2 is a primary cellular inhibitor of p53. It inhibits p53 function by multiple mechanisms, each of which, however, is mediated by their direct interaction. It has been proposed that small-molecule inhibitors designed to block the MDM2-p53 interaction may be effective in the treatment of human cancer retaining wild-type p53 by reactivating the p53 tumor suppressor function. Through nearly two decades of intense efforts, a number of structurally distinct, highly potent, nonpeptide, small-molecule inhibitors of the MDM2-p53 interaction (MDM2 inhibitors) have been successfully designed and developed, and at least seven such compounds have now been advanced into human clinical trials as new anticancer drugs. This review offers a perspective on the design and development of MDM2 small-molecule inhibitors and discusses early clinical data for some of the MDM2 small-molecule inhibitors and future challenges for the successful clinical development of MDM2 inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Shaomeng Wang
- University of Michigan Comprehensive Cancer Center and Departments of Internal Medicine, Pharmacology, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Yujun Zhao
- University of Michigan Comprehensive Cancer Center and Departments of Internal Medicine, Pharmacology, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Angelo Aguilar
- University of Michigan Comprehensive Cancer Center and Departments of Internal Medicine, Pharmacology, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Denzil Bernard
- University of Michigan Comprehensive Cancer Center and Departments of Internal Medicine, Pharmacology, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Chao-Yie Yang
- University of Michigan Comprehensive Cancer Center and Departments of Internal Medicine, Pharmacology, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
170
|
Turgut S, Ilhan M, Turan S, Karaman O, Yaylim I, Kucukhuseyin O, Tasan E. The Role of p16 and MDM2 Gene Polymorphisms in Prolactinoma: MDM2 Gene Polymorphisms May Be Associated with Tumor Shrinkage. ACTA ACUST UNITED AC 2017; 31:357-363. [PMID: 28438863 DOI: 10.21873/invivo.11067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 01/05/2023]
Abstract
AIM Prolactinomas are thought to arise from clonal expansion of a single mutated cell which is subjected to growth stimuli of several permissive factors, although the pathogenetic mechanisms underlying tumorigenesis remain unclear. The present study aimed to investigate the role of p16 (540C→G and 580C→T) and mouse double minute 2 (MDM2) (SNP309T→G) gene polymorphisms in tumorigenesis and characteristics of prolactinoma. PATIENTS AND METHODS A total of 74 patients with prolactinoma and 100 age- and gender-matched healthy individuals were enrolled in the study. Serum prolactin levels were measured by enzyme-linked immunosorbent assay (ELISA). p16 and MDM2 polymorphisms were determined by polymerase chain reaction-restriction fragment polymorphism and agarose gel electrophoresis. RESULTS p16 540C→G genotype distribution was found to be: CC: 66.2%, CG: 28.4%, GG: 5.4%; p16 580C→T genotype distribution was found to be: CC: 82.4%, CT: 17.6%, TT: 0% and MDM2 genotype distribution was found to be: TT: 31.1%, TG: 47.3%, GG: 21.6% in patients with prolactinoma. Tumor diameter before treatment was correlated with prolactin levels before treatment and percentage of prolactin decrease with treatment (r=0.719, p<0.001, p=0.034 r=0.256, respectively). The number of patients with tumor size decrease of more than 50% in those with homozygous genotype (TT+GG) of MDM2 SNP309T→G was significantly higher than in heterozygous genotype (TG) carriers (odds ratio(OR)=0.18, 95% confidence interval(CI)=0.06-0.58; p=0.003). CONCLUSION This study showed that p16 and MDM2 polymorphisms do not play a decisive role in tumorigenesis, but some genotypes of these polymorphisms might be associated with follow-up characteristics of prolactinoma.
Collapse
Affiliation(s)
- Seda Turgut
- Department of Internal Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Muzaffer Ilhan
- Department of Endocrinology and Metabolism, Bezmialem Vakif University, Istanbul, Turkey
| | - Saime Turan
- Department of Molecular Medicine, The Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Ozcan Karaman
- Department of Endocrinology and Metabolism, Bezmialem Vakif University, Istanbul, Turkey
| | - Ilhan Yaylim
- Department of Molecular Medicine, The Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Ozlem Kucukhuseyin
- Department of Molecular Medicine, The Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Ertugrul Tasan
- Department of Endocrinology and Metabolism, Bezmialem Vakif University, Istanbul, Turkey
| |
Collapse
|
171
|
Korf BR, Mikhail FM. Overview of Genetic Diagnosis in Cancer. ACTA ACUST UNITED AC 2017; 93:10.1.1-10.1.9. [DOI: 10.1002/cphg.36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Bruce R. Korf
- Department of Genetics University of Alabama at Birmingham Birmingham Alabama
| | - Fady M. Mikhail
- Department of Genetics University of Alabama at Birmingham Birmingham Alabama
| |
Collapse
|
172
|
Wang A, Meng M, Zhao X, Kong L. Long non-coding RNA ENST00462717 suppresses the proliferation, survival, and migration by inhibiting MDM2/MAPK pathway in glioma. Biochem Biophys Res Commun 2017; 485:513-521. [PMID: 28189682 DOI: 10.1016/j.bbrc.2017.02.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 02/05/2017] [Indexed: 01/16/2023]
Abstract
Gliomas are the most common and aggressive primary malignant tumor in the central nervous system, and requires new biomarkers and therapeutic methods. Long noncoding RNAs (lncRNAs) are important factors in numerous human diseases, including cancer. But studies on lncRNAs and gliomas are limited. In this study, we investigated the expression patterns of lncRNAs in 3 pairs of glioma samples and adjacent non-tumor tissues via microarray and selected the most down-regulated lnc00462717 to further verify its roles in glioma. We observed that decreased lnc00462717 expression was associated with the malignant status in glioma. In vitro experiment demonstrated that lnc00462717 overexpression suppressed glioma cell proliferation, survival and migration while knockdown of lnc00462717 had an opposite result. Moreover, we identified MDM2 as a direct target of lnc00462717 and lnc00462717 played a role by partially regulating the MDM2/MAPK pathway. In conclusion, lnc00462717 may function in suppressing glioma cell proliferation, survival, migration and may potentially serve as a novel biomarker and therapeutic target for glioma.
Collapse
Affiliation(s)
- Aiqin Wang
- EEG Room, Department of Neurology, Qilu Hospital of Shandong University, Shandong 250012, China.
| | - Mingzhu Meng
- EEG Room, Department of Neurology, Qilu Hospital of Shandong University, Shandong 250012, China
| | - Xiuhe Zhao
- Department of Neurology, Qilu Hospital of Shandong University, Shandong 250012, China
| | - Lina Kong
- Department of Medical Insurance Office, Qilu Hospital of Shandong University, Shandong 250012, China
| |
Collapse
|
173
|
Gansmo LB, Bjørnslett M, Halle MK, Salvesen HB, Romundstad P, Hveem K, Vatten L, Dørum A, Lønning PE, Knappskog S. MDM2 promoter polymorphism del1518 (rs3730485) and its impact on endometrial and ovarian cancer risk. BMC Cancer 2017; 17:97. [PMID: 28158999 PMCID: PMC5291962 DOI: 10.1186/s12885-017-3094-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/28/2017] [Indexed: 02/07/2023] Open
Abstract
Background The del1518 (rs3730485) polymorphism is an in/del variant in the MDM2 promoter P1. The variant is in complete linkage disequilibrium with MDM2 SNP309 (rs2279744) and has previously been found associated with an increased risk of colon cancer. In this study we assessed the impact of MDM2 del1518 on risk of ovarian and endometrial cancer. Methods Here, we genotyped del1518 in two large hospital-based series of patients diagnosed with ovarian (n = 1,385) or endometrial (n = 1,404) cancer and performed risk estimations as compared to the genotype distribution among 1,872 healthy female controls. Results In overall analysis we observed no association between del1518 and risk of either ovarian or endometrial cancer. However, stratifying according to SNP309 status, we found the del1518 variant to be associated with a reduced risk of endometrial cancer among individuals carrying the SNP309TT genotype both in the dominant (OR = 0.64; 95% CI = 0.45 – 0.90) and the recessive model (OR = 0.80; 95% CI = 0.65 – 1.00). No such association was observed for ovarian cancer risk. Conclusion We found the MDM2 del1518 del variant to be associated with reduced risk of endometrial cancer among individuals carrying the MDM2 SNP309TT genotype. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3094-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liv B Gansmo
- Section of Oncology, Department of Clinical Science, University of Bergen, Haukeland University Hospiltal, Jonas Lies veg 87, 5021, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Merete Bjørnslett
- Department of Molecular Oncology, Oslo University Hospital Radium Hospitalet, Oslo, Norway.,Institute for Cancer Research, University of Oslo, Oslo, Norway
| | - Mari Kyllesø Halle
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway.,Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Helga B Salvesen
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway.,Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pål Romundstad
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristian Hveem
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lars Vatten
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Dørum
- Department of Gynecologic Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | - Per E Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Haukeland University Hospiltal, Jonas Lies veg 87, 5021, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Stian Knappskog
- Section of Oncology, Department of Clinical Science, University of Bergen, Haukeland University Hospiltal, Jonas Lies veg 87, 5021, Bergen, Norway. .,Department of Oncology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
174
|
Jeon YJ, Park JH, Chung CH. Interferon-Stimulated Gene 15 in the Control of Cellular Responses to Genotoxic Stress. Mol Cells 2017; 40:83-89. [PMID: 28241406 PMCID: PMC5339507 DOI: 10.14348/molcells.2017.0027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 12/15/2022] Open
Abstract
Error-free replication and repair of DNA are pivotal to organisms for faithful transmission of their genetic information. Cells orchestrate complex signaling networks that sense and resolve DNA damage. Post-translational protein modifications by ubiquitin and ubiquitin-like proteins, including SUMO and NEDD8, are critically involved in DNA damage response (DDR) and DNA damage tolerance (DDT). The expression of interferon-stimulated gene 15 (ISG15), the first identified ubiquitin-like protein, has recently been shown to be induced under various DNA damage conditions, such as exposure to UV, camptothecin, and doxorubicin. Here we overview the recent findings on the role of ISG15 and its conjugation to target proteins (e.g., p53, ΔNp63α, and PCNA) in the control of cellular responses to genotoxic stress, such as the inhibition of cell growth and tumorigenesis.
Collapse
Affiliation(s)
- Young Joo Jeon
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 35015,
Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015,
Korea
| | - Jong Ho Park
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826,
Korea
| | - Chin Ha Chung
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826,
Korea
| |
Collapse
|
175
|
Zhou X, Cao B, Lu H. Negative auto-regulators trap p53 in their web. J Mol Cell Biol 2017; 9:62-68. [PMID: 28069666 PMCID: PMC5907828 DOI: 10.1093/jmcb/mjx001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/14/2016] [Accepted: 01/06/2017] [Indexed: 01/07/2023] Open
Abstract
The transcriptional factor p53 activates the expression of a myriad of target genes involving a complicated signalling network, resulting in various cellular outcomes, such as growth arrest, senescence, apoptosis, and metabolic changes, and leading to consequent suppression of tumour growth and progression. Because of the profoundly adverse effect of p53 on growth and proliferation of cancer cells, several feedback mechanisms have been employed by the cells to constrain p53 activity. Two major antagonists MDM2 and MDMX (the long forms) are transcriptionally induced by p53, but in return block p53 activity, forming a negative feedback circuit and rendering chemoresistance of several cancer cells. However, they are not alone, as cancer cells also employ other proteins encoded by p53 target genes to inhibit p53 activity at transcriptional, translational, and posttranslational levels. This essay is thus composed to review a recent progress in understanding the mechanisms for how cancer cells hijack the p53 autoregulation by these proteins for their growth advantage and to discuss the clinical implications of these autoregulatory loops.
Collapse
Affiliation(s)
- Xiang Zhou
- Fudan University Shanghai Cancer Center and the Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Bo Cao
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hua Lu
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
176
|
Borzi C, Calzolari L, Centonze G, Milione M, Sozzi G, Fortunato O. mir-660-p53-mir-486 Network: A New Key Regulatory Pathway in Lung Tumorigenesis. Int J Mol Sci 2017; 18:ijms18010222. [PMID: 28124991 PMCID: PMC5297851 DOI: 10.3390/ijms18010222] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/13/2017] [Accepted: 01/17/2017] [Indexed: 01/09/2023] Open
Abstract
Lung cancer is the most frequent cause of cancer-related death worldwide, with limited therapeutic options and rapid development of drug resistance. MicroRNAs, a class of small non-coding RNAs that control different physiological processes, have been associated with cancer development, as either oncomiRNAs or tumor-suppressor miRNAs. In the present study we investigated the interaction between mir-486-5p and mir-660-5p, two independent tumor-suppressor miRNAs, to assess their possible role and synergistic effect in lung cancer treatment. Our data show that mir-660-5p over-expression in A549 lung cancer cells induced a remarkable increase in mir-486-5p expression level and activity, detected as a reduction of its target gene, p85. mir-486-5p expression was confirmed by microRNA in situ hybridization. mir-660-5p modulated mir-486-5p through the silencing of Mouse Double Minute 2 (MDM2), one of its direct target, and then through p53 stimulation. This regulatory pathway was effective in A549, but not in H1299; therefore, only in the context of a functional p53 protein. Our findings support the conclusion that mir-486-5p is positively regulated by mir-660-5p in lung cancer cell lines, through the mir-660-MDM2-p53 pathway, making mir-660-5p even more interesting for its potential successful use in lung cancer therapy.
Collapse
Affiliation(s)
- Cristina Borzi
- Department of Experimental Oncology and Molecular Medicine, Unit of Tumor Genomics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| | - Linda Calzolari
- Department of Experimental Oncology and Molecular Medicine, Unit of Tumor Genomics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| | - Giovanni Centonze
- Department of Experimental Oncology and Molecular Medicine, Unit of Tumor Genomics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| | - Massimo Milione
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| | - Gabriella Sozzi
- Department of Experimental Oncology and Molecular Medicine, Unit of Tumor Genomics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| | - Orazio Fortunato
- Department of Experimental Oncology and Molecular Medicine, Unit of Tumor Genomics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| |
Collapse
|
177
|
Peterson LF, Lo MC, Liu Y, Giannola D, Mitrikeska E, Donato NJ, Johnson CN, Wang S, Mercer J, Talpaz M. Induction of p53 suppresses chronic myeloid leukemia. Leuk Lymphoma 2017; 58:1-14. [PMID: 28084835 DOI: 10.1080/10428194.2016.1272682] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chronic myeloid leukemia (CML) is characterized by the chromosomal translocation 9;22, known as the Philadelphia chromosome (Ph), which produces the BCR-ABL fusion tyrosine kinase. Although well-managed by BCR-ABL tyrosine kinase inhibitors (TKIs), treatment fails to eliminate Ph + primitive progenitors, and cessation of therapy frequently results in relapse. The p53 protein is an important regulator of cell cycle and apoptosis. The small molecules MI-219 target the interaction between p53 and its negative regulator HDM2, leading to its stabilization and activation. We show that treatment with MI-219 reduced the number of CML cells in both in vitro and in vivo settings but not that of normal primitive progenitors, and activated different gene signatures in CML potentially explaining the differential impact of this agent on each population. Our data suggest that a p53-activating agent may be an effective approach in the management and potential operational cure of CML.
Collapse
Affiliation(s)
- Luke F Peterson
- a Comprehensive Cancer Center , University of Michigan , Ann Arbor , MI , USA.,b Department of Internal Medicine, Division of Hematology and Oncology , University of Michigan , Ann Arbor , MI , USA
| | - Miao-Chia Lo
- c College of Pharmacy, University of Michigan , Ann Arbor , MI , USA
| | - Yihong Liu
- a Comprehensive Cancer Center , University of Michigan , Ann Arbor , MI , USA.,b Department of Internal Medicine, Division of Hematology and Oncology , University of Michigan , Ann Arbor , MI , USA
| | - Diane Giannola
- a Comprehensive Cancer Center , University of Michigan , Ann Arbor , MI , USA
| | - Emilija Mitrikeska
- a Comprehensive Cancer Center , University of Michigan , Ann Arbor , MI , USA.,b Department of Internal Medicine, Division of Hematology and Oncology , University of Michigan , Ann Arbor , MI , USA
| | - Nicholas J Donato
- a Comprehensive Cancer Center , University of Michigan , Ann Arbor , MI , USA.,b Department of Internal Medicine, Division of Hematology and Oncology , University of Michigan , Ann Arbor , MI , USA
| | - Craig N Johnson
- d DNA Sequencing and Microarray Core , University of Michigan , Ann Arbor , MI , USA
| | - Shaomeng Wang
- e Department of Pharmacology and Medicinal Chemistry , University of Michigan , Ann Arbor , MI , USA
| | - Jessica Mercer
- a Comprehensive Cancer Center , University of Michigan , Ann Arbor , MI , USA.,b Department of Internal Medicine, Division of Hematology and Oncology , University of Michigan , Ann Arbor , MI , USA
| | - Moshe Talpaz
- a Comprehensive Cancer Center , University of Michigan , Ann Arbor , MI , USA.,b Department of Internal Medicine, Division of Hematology and Oncology , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
178
|
Tang HM, Talbot CC, Fung MC, Tang HL. Molecular signature of anastasis for reversal of apoptosis. F1000Res 2017; 6:43. [PMID: 28299189 DOI: 10.12688/f1000research.10568.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2017] [Indexed: 12/22/2022] Open
Abstract
Anastasis (Greek for "rising to life") is a cell recovery phenomenon that rescues dying cells from the brink of cell death. We recently discovered anastasis to occur after the execution-stage of apoptosis in vitro and in vivo. Promoting anastasis could in principle preserve injured cells that are difficult to replace, such as cardiomyocytes and neurons. Conversely, arresting anastasis in dying cancer cells after cancer therapies could improve treatment efficacy. To develop new therapies that promote or inhibit anastasis, it is essential to identify the key regulators and mediators of anastasis - the therapeutic targets. Therefore, we performed time-course microarray analysis to explore the molecular mechanisms of anastasis during reversal of ethanol-induced apoptosis in mouse primary liver cells. We found striking changes in transcription of genes involved in multiple pathways, including early activation of pro-cell survival, anti-oxidation, cell cycle arrest, histone modification, DNA-damage and stress-inducible responses, and at delayed times, angiogenesis and cell migration. Validation with RT-PCR confirmed similar changes in the human liver cancer cell line, HepG2, during anastasis. Here, we present the time-course whole-genome gene expression dataset revealing gene expression profiles during the reversal of apoptosis. This dataset provides important insights into the physiological, pathological, and therapeutic implications of anastasis.
Collapse
Affiliation(s)
- Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - C Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ming Chiu Fung
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho Lam Tang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA
| |
Collapse
|
179
|
Abstract
Anastasis (Greek for "rising to life") is a cell recovery phenomenon that rescues dying cells from the brink of cell death. We recently discovered anastasis to occur after the execution-stage of apoptosis
in vitro and
in vivo. Promoting anastasis could in principle preserve injured cells that are difficult to replace, such as cardiomyocytes and neurons. Conversely, arresting anastasis in dying cancer cells after cancer therapies could improve treatment efficacy. To develop new therapies that promote or inhibit anastasis, it is essential to identify the key regulators and mediators of anastasis – the therapeutic targets. Therefore, we performed time-course microarray analysis to explore the molecular mechanisms of anastasis during reversal of ethanol-induced apoptosis in mouse primary liver cells. We found striking changes in transcription of genes involved in multiple pathways, including early activation of pro-cell survival, anti-oxidation, cell cycle arrest, histone modification, DNA-damage and stress-inducible responses, and at delayed times, angiogenesis and cell migration. Validation with RT-PCR confirmed similar changes in the human liver cancer cell line, HepG2, during anastasis. Here, we present the time-course whole-genome gene expression dataset revealing gene expression profiles during the reversal of apoptosis. This dataset provides important insights into the physiological, pathological, and therapeutic implications of anastasis.
Collapse
Affiliation(s)
- Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - C Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ming Chiu Fung
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho Lam Tang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA
| |
Collapse
|
180
|
microRNA-1827 represses MDM2 to positively regulate tumor suppressor p53 and suppress tumorigenesis. Oncotarget 2017; 7:8783-96. [PMID: 26840028 PMCID: PMC4891004 DOI: 10.18632/oncotarget.7088] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/15/2016] [Indexed: 12/14/2022] Open
Abstract
The tumor suppressor p53 plays a central role in tumor prevention. The E3 ubiquitin ligase MDM2 is the most critical negative regulator of p53, which binds to p53 and degrades p53 through ubiquitation. MDM2 itself is a transcriptional target of p53, and therefore, MDM2 forms a negative feedback loop with p53 to tightly regulate p53 levels and function. microRNAs (miRNAs) play a key role in regulation of gene expression. miRNA dysregulation plays an important role in tumorigenesis. In this study, we found that miRNA miR-1827 is a novel miRNA that targets MDM2 through binding to the 3′-UTR of MDM2 mRNA. miR-1827 negatively regulates MDM2, which in turn increases p53 protein levels to increase transcriptional activity of p53 and enhance p53-mediated stress responses, including apoptosis and senescence. Overexpression of miR-1827 suppresses the growth of xenograft colorectal tumors, whereas the miR-1827 inhibitor promotes tumor growth in mice in a largely p53-dependent manner. miR-1827 is frequently down-regulated in human colorectal cancer. Decreased miR-1827 expression is associated with high MDM2 expression and poor prognosis in colorectal cancer. In summary, our results reveal that miR-1827 is a novel miRNA that regulates p53 through targeting MDM2, and highlight an important role and the underlying mechanism of miR-1827 in tumor suppression.
Collapse
|
181
|
Raj N, Attardi LD. The Transactivation Domains of the p53 Protein. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026047. [PMID: 27864306 DOI: 10.1101/cshperspect.a026047] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The p53 tumor suppressor is a transcriptional activator, with discrete domains that participate in sequence-specific DNA binding, tetramerization, and transcriptional activation. Mutagenesis and reporter studies have delineated two distinct activation domains (TADs) and specific hydrophobic residues within these TADs that are critical for their function. Knockin mice expressing p53 mutants with alterations in either or both of the two TADs have revealed that TAD1 is critical for responses to acute DNA damage, whereas both TAD1 and TAD2 participate in tumor suppression. Biochemical and structural studies have identified factors that bind either or both TADs, including general transcription factors (GTFs), chromatin modifiers, and negative regulators, helping to elaborate a model through which p53 activates transcription. Posttranslational modifications (PTMs) of the p53 TADs through phosphorylation also regulate TAD activity. Together, these studies on p53 TADs provide great insight into how p53 serves as a tumor suppressor.
Collapse
Affiliation(s)
- Nitin Raj
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305.,Department of Genetics, Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
182
|
Zhang L, Ren W, Wang X, Zhang J, Liu J, Zhao L, Zhang X. Discovery of novel polycyclic spiro-fused carbocyclicoxindole-based anticancer agents. Eur J Med Chem 2017; 126:1071-1082. [DOI: 10.1016/j.ejmech.2016.12.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/30/2016] [Accepted: 12/09/2016] [Indexed: 10/20/2022]
|
183
|
Wang XD, Su GY, Zhao C, Qu FZ, Wang P, Zhao YQ. Anticancer activity and potential mechanisms of 1C, a ginseng saponin derivative, on prostate cancer cells. J Ginseng Res 2016; 42:133-143. [PMID: 29719459 PMCID: PMC5925623 DOI: 10.1016/j.jgr.2016.12.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/27/2016] [Accepted: 12/26/2016] [Indexed: 01/01/2023] Open
Abstract
Background AD-2 (20(R)-dammarane-3b, 12b, 20, 25-tetrol; 25-OH-PPD) is a ginsenoside and isolated from Panax ginseng, showing anticancer activity against extensive human cancer cell lines. In this study, effects and mechanisms of 1C ((20R)-3b-O-(L-alanyl)-dammarane-12b, 20, 25-triol), a modified version of AD-2, were evaluated for its development as a novel anticancer drug. Methods MTT assay was performed to evaluate cell cytotoxic activity. Cell cycle and levels of reactive oxygen species (ROS) were determined using flow cytometry analysis. Western blotting was employed to analyze signaling pathways. Results 1C concentration-dependently reduces prostate cancer cell viability without affecting normal human gastric epithelial cell line-1 viability. In LNCaP prostate cancer cells, 1C triggered apoptosis via Bcl-2 family-mediated mitochondria pathway, downregulated expression of mouse double minute 2, upregulated expression of p53 and stimulated ROS production. ROS scavenger, N-acetylcysteine, can attenuate 1C-induced apoptosis. 1C also inhibited the proliferation of LNCaP cells through inhibition on Wnt/β-catenin signaling pathway. Conclusion 1C shows obvious anticancer activity based on inducing cell apoptosis by Bcl-2 family-mediated mitochondria pathway and ROS production, inhibiting Wnt/β-catenin signaling pathway. These findings demonstrate that 1C may provide leads as a potential agent for cancer therapy. Ginseng saponin derivative 1C was obtained by structural modification. Anticancer activity of 1C is much better than that of the original compound AD-2 on cancer cells. 1C induces cell apoptosis by Bcl-2 family-mediated mitochondria pathway and ROS production. 1C inhibits Wnt/β-catenin signaling pathway
Collapse
Affiliation(s)
- Xu De Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-based Drug Design and Discovery of Education, Shenyang Pharmaceurical University, Shenyang, China
| | - Guang Yue Su
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-based Drug Design and Discovery of Education, Shenyang Pharmaceurical University, Shenyang, China
| | - Chen Zhao
- Key Laboratory of Structure-based Drug Design and Discovery of Education, Shenyang Pharmaceurical University, Shenyang, China.,College of Life Science and Biological Pharmaceutical, Shenyang Pharmaceutical University, Shenyang, China
| | - Fan Zhi Qu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-based Drug Design and Discovery of Education, Shenyang Pharmaceurical University, Shenyang, China
| | - Peng Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-based Drug Design and Discovery of Education, Shenyang Pharmaceurical University, Shenyang, China
| | - Yu Qing Zhao
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China.,Key Laboratory of Structure-based Drug Design and Discovery of Education, Shenyang Pharmaceurical University, Shenyang, China
| |
Collapse
|
184
|
Carr MI, Jones SN. Regulation of the Mdm2-p53 signaling axis in the DNA damage response and tumorigenesis. Transl Cancer Res 2016; 5:707-724. [PMID: 28690977 PMCID: PMC5501481 DOI: 10.21037/tcr.2016.11.75] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The p53 tumor suppressor acts as a guardian of the genome in mammalian cells undergoing DNA double strand breaks induced by a various forms of cell stress, including inappropriate growth signals or ionizing radiation. Following damage, p53 protein levels become greatly elevated in cells and p53 functions primarily as a transcription factor to regulate the expression a wide variety of genes that coordinate this DNA damage response. In cells undergoing high amounts of DNA damage, p53 can promote apoptosis, whereas in cells undergoing less damage, p53 promotes senescence or transient cell growth arrest and the expression of genes involved in DNA repair, depending upon the cell type and level of damage. Failure of the damaged cell to undergo growth arrest or apoptosis, or to respond to the DNA damage by other p53-coordinated mechanisms, can lead to inappropriate cell growth and tumorigenesis. In cells that have successfully responded to genetic damage, the amount of p53 present in the cell must return to basal levels in order for the cell to resume normal growth and function. Although regulation of p53 levels and function is coordinated by many proteins, it is now widely accepted that the master regulator of p53 is Mdm2. In this review, we discuss the role(s) of p53 in the DNA damage response and in tumor suppression, and how post-translational modification of Mdm2 regulates the Mdm2-p53 signaling axis to govern p53 activities in the cell.
Collapse
Affiliation(s)
- Michael I Carr
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Stephen N Jones
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
185
|
Abstract
Since its discovery more than three decades ago, tumor suppressor p53 has been shown to play pivotal roles in both maintaining genomic integrity and tumor suppression. p53 functions as a transcription factor responding to a multitude of cellular stressors, regulating the transcription of many genes involved in cell-cycle arrest, senescence, autophagy, and apoptosis. Extensive work has revealed that p53 is one of the most commonly mutated tumor suppressor genes. The last three decades have demonstrated that p53 activity is controlled through transcriptional regulation and posttranslational modifications. However, evolving work is now uncovering that p53, and other p53 family members, are post-transcriptionally regulated by multiple RNA-binding proteins (RBPs). Understanding the regulation of p53 by RBPs may potentially open up the possibility for cancer therapeutic intervention. This review focuses on the posttranscriptional regulation of p53, and p53 family members, by RNA binding proteins and the reciprocal feedback pathways between several RNA-biding proteins modulating p53, and p53 family members.
Collapse
Affiliation(s)
- Chris Lucchesi
- Comparative Oncology Laboratory, School of Veterinary Medicine, School of Medicine, University of California at Davis, Davis, California 95616, USA
| | - Jin Zhang
- Comparative Oncology Laboratory, School of Veterinary Medicine, School of Medicine, University of California at Davis, Davis, California 95616, USA
| | - Xinbin Chen
- Comparative Oncology Laboratory, School of Veterinary Medicine, School of Medicine, University of California at Davis, Davis, California 95616, USA
| |
Collapse
|
186
|
Olivos DJ, Mayo LD. Emerging Non-Canonical Functions and Regulation by p53: p53 and Stemness. Int J Mol Sci 2016; 17:ijms17121982. [PMID: 27898034 PMCID: PMC5187782 DOI: 10.3390/ijms17121982] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 01/15/2023] Open
Abstract
Since its discovery nearly 40 years ago, p53 has ascended to the forefront of investigated genes and proteins across diverse research disciplines and is recognized most exclusively for its role in cancer as a tumor suppressor. Levine and Oren (2009) reviewed the evolution of p53 detailing the significant discoveries of each decade since its first report in 1979. In this review, we will highlight the emerging non-canonical functions and regulation of p53 in stem cells. We will focus on general themes shared among p53's functions in non-malignant stem cells and cancer stem-like cells (CSCs) and the influence of p53 on the microenvironment and CSC niche. We will also examine p53 gain of function (GOF) roles in stemness. Mutant p53 (mutp53) GOFs that lead to survival, drug resistance and colonization are reviewed in the context of the acquisition of advantageous transformation processes, such as differentiation and dedifferentiation, epithelial-to-mesenchymal transition (EMT) and stem cell senescence and quiescence. Finally, we will conclude with therapeutic strategies that restore wild-type p53 (wtp53) function in cancer and CSCs, including RING finger E3 ligases and CSC maintenance. The mechanisms by which wtp53 and mutp53 influence stemness in non-malignant stem cells and CSCs or tumor-initiating cells (TICs) are poorly understood thus far. Further elucidation of p53's effects on stemness could lead to novel therapeutic strategies in cancer research.
Collapse
Affiliation(s)
- David J Olivos
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Lindsey D Mayo
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
187
|
p53 pathway dysfunction is highly prevalent in acute myeloid leukemia independent of TP53 mutational status. Leukemia 2016; 31:1296-1305. [DOI: 10.1038/leu.2016.350] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 12/17/2022]
|
188
|
Wang P, Wang M, Li S, Ma L, Xi S, He J. Association of the MDM2 SNP285 Polymorphism with Cancer Susceptibility: A Meta-Analysis. DISEASE MARKERS 2016; 2016:4585484. [PMID: 27890964 PMCID: PMC5116512 DOI: 10.1155/2016/4585484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/28/2016] [Accepted: 10/05/2016] [Indexed: 02/07/2023]
Abstract
The mouse double minute 2 (MDM2) gene encodes a negative regulator for p53, and the polymorphism SNP285 in the promoter region of MDM2 gene has been implicated in cancer risk, but individual published studies had inconclusive results. Therefore, we performed this meta-analysis to obtain a more precise estimation between MDM2 SNP285 polymorphism and risk of cancer. A systematic literature search was performed using the PubMed, Embase, and Chinese Biomedical (CBM) databases. Ultimately, 16 published studies comprising 14,573 cases and 9,115 controls were included. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to assess the strength of associations. Overall, MDM2 SNP285 polymorphism was significantly associated with a decreased overall cancer risk with the heterozygous model (OR = 0.89, 95% CI = 0.79-0.99), and reduced ORs were observed with other genetic models (dominant: OR = 0.90, 95% CI = 0.79-1.01 and allele comparison: OR = 0.91, 95% CI = 0.80-1.03) but not reaching statistical significance. Stratification analysis indicated a decreased risk for ovarian cancer, Caucasians, and studies with relatively large sample size. Despite some limitations, this meta-analysis indicated that the MDM2 SNP285 polymorphism was associated with a decreased cancer risk, which warrants further validation in large and well-designed studies.
Collapse
Affiliation(s)
- Ping Wang
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, China
| | - Meilin Wang
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, China
| | - Sanqiang Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang 471023, China
| | - Lingjun Ma
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, China
| | - Shoumin Xi
- The Key Laboratory of Pharmacology and Medical Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang 471023, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
189
|
Simerzin A, Zorde-Khvalevsky E, Rivkin M, Adar R, Zucman-Rossi J, Couchy G, Roskams T, Govaere O, Oren M, Giladi H, Galun E. The liver-specific microRNA-122*, the complementary strand of microRNA-122, acts as a tumor suppressor by modulating the p53/mouse double minute 2 homolog circuitry. Hepatology 2016; 64:1623-1636. [PMID: 27302319 DOI: 10.1002/hep.28679] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 05/10/2016] [Accepted: 05/27/2016] [Indexed: 12/12/2022]
Abstract
UNLABELLED The tumor suppressor p53 is a central regulator of signaling pathways that controls the cell cycle and maintains the integrity of the human genome. p53 level is regulated by mouse double minute 2 homolog (Mdm2), which marks p53 for proteasomal degradation. The p53-Mdm2 circuitry is subjected to complex regulation by a variety of mechanisms, including microRNAs (miRNAs). We found a novel effector of this regulatory circuit, namely, miR-122*, the passenger strand of the abundantly expressed liver-specific miR-122. Here, we demonstrate that miR-122* levels are reduced in human hepatocellular carcinoma (HCC). We found that miR-122* targets Mdm2, thus participating as an important player in the p53-Mdm2 circuitry. Moreover, we observed significant negative correlation between levels of miR-122* and Mdm2 in a large set of human HCC samples. In vivo tumorigenicity assays demonstrate that miR-122* is capable of inhibiting tumor growth, emphasizing the tumor-suppressor characteristics of this miRNA. Furthermore, we show that blocking miR-122 in murine livers with an antagomiR-122 (miRNA inhibitor) results in miR-122* accumulation, leading to Mdm2 repression followed by elevated p53 protein levels. CONCLUSION miR-122*, the passenger strand of miR-122, regulates the activity of p53 by targeting Mdm2. Importantly, similarly to miR-122, miR-122* is significantly down-regulated in human HCC. We therefore propose that miR-122* is an important contributor to the tumor suppression activity previously attributed solely to miR-122. (Hepatology 2016;64:1623-1636).
Collapse
Affiliation(s)
- Alina Simerzin
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem, Israel
| | - Elina Zorde-Khvalevsky
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem, Israel
| | - Mila Rivkin
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem, Israel
| | - Revital Adar
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem, Israel
| | - Jessica Zucman-Rossi
- Inserm, UMR-1162, Functional Genomic of Solid Tumors, Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Bobigny, France.,Université Paris Diderot, IUH, Paris, France
| | - Gabrielle Couchy
- Inserm, UMR-1162, Functional Genomic of Solid Tumors, Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Bobigny, France.,Université Paris Diderot, IUH, Paris, France
| | - Tania Roskams
- Department of Pathology and the Laboratory of Morphology and Molecular Pathology, University Hospitals, University of Leuven, Leuven, Belgium
| | - Olivier Govaere
- Department of Pathology and the Laboratory of Morphology and Molecular Pathology, University Hospitals, University of Leuven, Leuven, Belgium
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hilla Giladi
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem, Israel
| | - Eithan Galun
- The Goldyne Savad Institute of Gene and Cell Therapy, Hadassah Hebrew University Hospital, Ein Karem, Jerusalem, Israel.
| |
Collapse
|
190
|
Slabáková E, Kharaishvili G, Smějová M, Pernicová Z, Suchánková T, Remšík J, Lerch S, Straková N, Bouchal J, Král M, Culig Z, Kozubík A, Souček K. Opposite regulation of MDM2 and MDMX expression in acquisition of mesenchymal phenotype in benign and cancer cells. Oncotarget 2016; 6:36156-71. [PMID: 26416355 PMCID: PMC4742168 DOI: 10.18632/oncotarget.5392] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/15/2015] [Indexed: 01/14/2023] Open
Abstract
Plasticity of cancer cells, manifested by transitions between epithelial and mesenchymal phenotypes, represents a challenging issue in the treatment of neoplasias. Both epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are implicated in the processes of metastasis formation and acquisition of stem cell-like properties. Mouse double minute (MDM) 2 and MDMX are important players in cancer progression, as they act as regulators of p53, but their function in EMT and metastasis may be contradictory. Here, we show that the EMT phenotype in multiple cellular models and in clinical prostate and breast cancer samples is associated with a decrease in MDM2 and increase in MDMX expression. Modulation of EMT-accompanying changes in MDM2 expression in benign and transformed prostate epithelial cells influences their migration capacity and sensitivity to docetaxel. Analysis of putative mechanisms of MDM2 expression control demonstrates that in the context of defective p53 function, MDM2 expression is regulated by EMT-inducing transcription factors Slug and Twist. These results provide an alternative context-specific role of MDM2 in EMT, cell migration, metastasis, and therapy resistance.
Collapse
Affiliation(s)
- Eva Slabáková
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of The Czech Republic, v.v.i., Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Gvantsa Kharaishvili
- Department of Clinical and Molecular Pathology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Monika Smějová
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of The Czech Republic, v.v.i., Brno, Czech Republic.,Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Zuzana Pernicová
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of The Czech Republic, v.v.i., Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Tereza Suchánková
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of The Czech Republic, v.v.i., Brno, Czech Republic
| | - Ján Remšík
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of The Czech Republic, v.v.i., Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Stanislav Lerch
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of The Czech Republic, v.v.i., Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Nicol Straková
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of The Czech Republic, v.v.i., Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Milan Král
- Department of Urology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Zoran Culig
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Alois Kozubík
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of The Czech Republic, v.v.i., Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of The Czech Republic, v.v.i., Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
191
|
Lee YY, Wang CT, Huang SKH, Wu WJ, Huang CN, Li CC, Chan TC, Liang PI, Hsing CH, Li CF. Downregulation of RNF128 Predicts Progression and Poor Prognosis in Patients with Urothelial Carcinoma of the Upper Tract and Urinary Bladder. J Cancer 2016; 7:2187-2196. [PMID: 27994654 PMCID: PMC5166527 DOI: 10.7150/jca.16798] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/04/2016] [Indexed: 01/10/2023] Open
Abstract
Background: The TP53 tumor suppressor gene plays a crucial role in the carcinogenesis of many malignancies, including urothelial carcinoma (UC). Overexpression of p53 is associated with poor prognosis in UC. Recently, RING finger protein 128 (RNF128) was shown to be involved in p53-induced apoptosis, forming a negative feedback loop. However, the significance of RNF128 in patients with UC remains unknown. In this study, our aim was to evaluate the expression of RNF128 in UC and to assess its predictive and prognostic value in a well-established cohort. Methods: Through data mining from a published transcriptome (GSE31684), RNF128 was identified as the most differentially expressed gene in UC among those associated with negative regulation of the cytokine biosynthetic process (GO:0042036). Its immunoexpression was further evaluated using the H-scores of 340 patients with upper urinary tract UC (UTUC) and 295 with urinary bladder UC (UBUC). The scores were correlated with clinicopathological features, disease-specific survival (DSS) and metastasis-free survival (MeFS). We also used Western blot analysis to evaluate RNF128 protein expression in human urothelial cell (HUC) lines. Results: Downregulation of RNF128 expression was significantly associated with advanced pT stage (p<0.001), high histological grade (UTUC, p<0.001; UBUC, p=0.035), nodal metastasis (UTUC, p<0.001; UBUC, p=0.001), vascular invasion (UTUC, p<0.001; UBUC, p=0.008) and high mitotic rate (UTUC, p=0.003; UBUC, p=0.023). Low expression of RNF128 was an adverse prognosticator for DSS (UTUC, p<0.0001; UBUC, p<0.0001) and MeFS (UTUC, p<0.0001; UBUC, p=0.0002). Moreover, low expression was predictive of poor DSS (UTUC, p=0.006; UBUC, p=0.003) and MeFS (UTUC, p=0.009; UBUC, p=0.036) in multivariate comparisons. Western blot analysis showed that the RNF128 protein was downregulated in invasive urothelial cancer cell lines. Conclusion: Our findings showed that downregulation of RNF128 was correlated with cancer invasiveness and metastasis as well as reduced survival in patients with UTUC and UBUC, identifying RNF128 as a prognostic factor in UC.
Collapse
Affiliation(s)
- Yi-Ying Lee
- Department of Pathology, Chi Mei Medical Center, Liouying, Taiwan
- Institute of Biomedical Science National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chieh-Tien Wang
- Department of Pathology, Chi Mei Medical Center, Liouying, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology
| | | | - Wen-Jeng Wu
- Department of Urology, Faculty of Medicine, Kaohsiung Medical University
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital
| | - Chun-Nung Huang
- Department of Urology, Faculty of Medicine, Kaohsiung Medical University
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University
| | - Ching-Chia Li
- Department of Urology, Faculty of Medicine, Kaohsiung Medical University
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital
| | - Ti-Chun Chan
- Institute of Biomedical Science National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Peir-In Liang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
| | - Chien-Feng Li
- Division of Clinical Pathology, Chi Mei Medical Center, Tainan, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
192
|
Hernández-Monge J, Rousset-Roman AB, Medina-Medina I, Olivares-Illana V. Dual function of MDM2 and MDMX toward the tumor suppressors p53 and RB. Genes Cancer 2016; 7:278-287. [PMID: 28050229 PMCID: PMC5115168 DOI: 10.18632/genesandcancer.120] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The orchestrated crosstalk between the retinoblastoma (RB) and p53 pathways contributes to preserving proper homeostasis within the cell. The deregulation of one or both pathways is a common factor in the development of most types of human cancer. The proto-oncoproteins MDMX and MDM2 are the main regulators of the well- known tumor suppressor p53 protein. Under normal conditions, MDM2 and MDMX inhibit p53, either via repression of its transcriptional activity by protein-protein interaction, or via polyubiquitination as a result of MDM2-E3 ubiquitin ligase activity, for which MDM2 needs to dimerize with MDMX. Under genotoxic stress conditions, both become positive regulators of p53. The ATM-dependent phosphorylation of MDM2 and MDMX allow them to bind p53 mRNA, these interactions promote p53 translation. MDM2 and MDMX are also being revealed as effective regulators of the RB protein. MDM2 is able to degrade RB by two different mechanisms, that is, by ubiquitin dependent and independent pathways. MDMX enhances the ability of MDM2 to bind and degrade RB protein. However, MDMX also seems to stabilize RB through interaction and competition with MDM2. Here, we will contextualize the findings that suggest that the MDM2 and MDMX proteins have a dual function on both p53 and RB.
Collapse
Affiliation(s)
- Jesús Hernández-Monge
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Av Manuel Nava No 6 Zona Universitaria CP 78290. SLP, México
| | - Adriana Berenice Rousset-Roman
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Av Manuel Nava No 6 Zona Universitaria CP 78290. SLP, México
| | - Ixaura Medina-Medina
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Av Manuel Nava No 6 Zona Universitaria CP 78290. SLP, México
| | - Vanesa Olivares-Illana
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Av Manuel Nava No 6 Zona Universitaria CP 78290. SLP, México
| |
Collapse
|
193
|
Akhtar S, Khan MKA, Arif JM. Evaluation and Elucidation Studies of Natural Aglycones for Anticancer Potential using Apoptosis-Related Markers: An In silico Study. Interdiscip Sci 2016; 10:297-310. [DOI: 10.1007/s12539-016-0191-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 09/20/2016] [Accepted: 09/23/2016] [Indexed: 12/17/2022]
|
194
|
Matijasevic Z, Krzywicka-Racka A, Sluder G, Gallant J, Jones SN. The Zn-finger domain of MdmX suppresses cancer progression by promoting genome stability in p53-mutant cells. Oncogenesis 2016; 5:e262. [PMID: 27694836 PMCID: PMC5117848 DOI: 10.1038/oncsis.2016.62] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/09/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023] Open
Abstract
The MDMX (MDM4) oncogene is amplified or overexpressed in a significant percentage of human tumors. MDMX is thought to function as an oncoprotein by binding p53 tumor suppressor protein to inhibit p53-mediated transcription, and by complexing with MDM2 oncoprotein to promote MDM2-mediated degradation of p53. However, down-regulation or loss of functional MDMX has also been observed in a variety of human tumors that are mutated for p53, often correlating with more aggressive cancers and a worse patient prognosis. We have previously reported that endogenous levels of MdmX can suppress proliferation and promote pseudo-bipolar mitosis in primary and tumor cells derived from p53-deficient mice, and that MdmX-p53 double deficient mice succumb to spontaneously formed tumors more rapidly than p53-deficient mice. These results suggest that the MdmX oncoprotein may act as a tumor-suppressor in cancers with compromised p53 function. By using orthotopic transplantation and lung colonization assays in mice we now establish a p53-independent anti-oncogenic role for MdmX in tumor progression. We also demonstrate that the roles of MdmX in genome stability and in proliferation are two distinct functions encoded by the separate MdmX protein domains. The central Zn-finger domain suppresses multipolar mitosis and chromosome loss, whereas the carboxy-terminal RING domain suppresses proliferation of p53-deficient cells. Furthermore, we determine that it is the maintenance of genome stability that underlies MdmX role in suppression of tumorigenesis in hyperploid p53 mutant tumors. Our results offer a rationale for the increased metastatic potential of p53 mutant human cancers with aberrant MdmX function and provide a caveat for the application of anti-MdmX treatment of tumors with compromised p53 activity.
Collapse
Affiliation(s)
- Z Matijasevic
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - A Krzywicka-Racka
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - G Sluder
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - J Gallant
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - S N Jones
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
195
|
Kang MH, Reynolds CP, Kolb EA, Gorlick R, Carol H, Lock R, Keir ST, Maris JM, Wu J, Lyalin D, Kurmasheva RT, Houghton PJ, Smith MA. Initial Testing (Stage 1) of MK-8242-A Novel MDM2 Inhibitor-by the Pediatric Preclinical Testing Program. Pediatr Blood Cancer 2016; 63:1744-52. [PMID: 27238606 PMCID: PMC5657425 DOI: 10.1002/pbc.26064] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 03/29/2016] [Accepted: 04/12/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND MK-8242 is an inhibitor of MDM2 that stabilizes the tumor suppressor TP53 and induces growth arrest or apoptosis downstream of TP53 induction. PROCEDURES MK-8242 was tested against the Pediatric Preclinical Testing Program (PPTP) in vitro cell line panel at concentrations from 1.0 nM to 10.0 μM and against the PPTP in vivo xenograft panels using oral gavage on Days 1-5 and Day 15-19 at a dose of 125 mg/kg (solid tumors) or 75 mg/kg (acute lymphoblastic leukemia [ALL] models). RESULTS The median IC50 for MK-8242 was 0.07 μM for TP53 wild-type cell lines versus >10 μM for TP53 mutant cell lines. MK-8242 induced a twofold or greater delay in time to event in 10 of 17 (59%) of TP53 wild-type solid tumor xenografts, excluding osteosarcoma xenografts that have very low TP53 expression. Objective responses were observed in seven solid tumor xenografts representing multiple histotypes. For the systemic-disease ALL panel, among eight xenografts there were two complete responses (CRs) and six partial responses (PRs). Two additional MLL-rearranged xenografts (MV4;11 and RS4;11) grown subcutaneously showed maintained CR and PR, respectively. The expected pharmacodynamic responses to TP53 activation were observed in TP53 wild-type models treated with MK-8242. Pharmacokinetic analysis showed that MK-8242 drug exposure in SCID mice appears to exceed that was observed in adult phase 1 trials. CONCLUSIONS MK-8242-induced tumor regressions across multiple solid tumor histotypes and induced CRs or PRs for most ALL xenografts. This activity was observed at MK-8242 drug exposures that appear to exceed those observed in human phase 1 trials.
Collapse
Affiliation(s)
- Min H. Kang
- Texas Tech University Health Sciences Center, Lubbock, TX
| | | | | | | | - Hernan Carol
- Children’s Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | - Richard Lock
- Children’s Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | | | - John M. Maris
- Children’s Hospital of Philadelphia, University of Pennsylvania School of Medicine and Abramson Family Cancer Research Institute, Philadelphia, PA
| | - Jianwrong Wu
- St. Jude Children's Research Hospital, Memphis, TN
| | | | | | | | | |
Collapse
|
196
|
El Atat O, Antonios D, Hilal G, Hokayem N, Abou-Ghoch J, Hashim H, Serhal R, Hebbo C, Moussa M, Alaaeddine N. An Evaluation of the Stemness, Paracrine, and Tumorigenic Characteristics of Highly Expanded, Minimally Passaged Adipose-Derived Stem Cells. PLoS One 2016; 11:e0162332. [PMID: 27632538 PMCID: PMC5024991 DOI: 10.1371/journal.pone.0162332] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/22/2016] [Indexed: 12/13/2022] Open
Abstract
The use of adipose-derived stem cells (ADSC) in regenerative medicine is rising due to their plasticity, capacity of differentiation and paracrine and trophic effects. Despite the large number of cells obtained from adipose tissue, it is usually not enough for therapeutic purposes for many diseases or cosmetic procedures. Thus, there is the need for culturing and expanding cells in-vitro for several weeks remain. Our aim is to investigate if long- term proliferation with minimal passaging will affect the stemness, paracrine secretions and carcinogenesis markers of ADSC. The immunophenotypic properties and aldehyde dehydrogenase (ALDH) activity of the initial stromal vascular fraction (SVF) and serially passaged ADSC were observed by flow cytometry. In parallel, the telomerase activity and the relative expression of oncogenes and tumor suppressor genes were assessed by q-PCR. We also assessed the cytokine secretion profile of passaged ADSC by an ELISA. The expanded ADSC retain their morphological and phenotypical characteristics. These cells maintained in culture for up to 12 weeks until P4, possessed stable telomerase and ALDH activity, without having a TP53 mutation. Furthermore, the relative expression levels of TP53, RB, and MDM2 were not affected while the relative expression of c-Myc decreased significantly. Finally, the levels of the secretions of PGE2, STC1, and TIMP2 were not affected but the levels of IL-6, VEGF, and TIMP 1 significantly decreased at P2. Our results suggest that the expansion of passaged ADSC does not affect the differentiation capacity of stem cells and does not confer a cancerous state or capacity in vitro to the cells.
Collapse
Affiliation(s)
- Oula El Atat
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Diane Antonios
- Toxicology Laboratory, Faculty of Pharmacy, St. Joseph University, Beirut, Lebanon
| | - George Hilal
- Cancer and Metabolism Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Nabil Hokayem
- Department of Plastic& Reconstructive Surgery, Hotel Dieu de France, and Faculty of Medicine St Joseph University, Beirut, Lebanon
| | - Joelle Abou-Ghoch
- Medical Genetics Unit, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Hussein Hashim
- Department of Plastic& Reconstructive Surgery, Fuad Khoury Hospital, Beirut, Lebanon
| | - Rim Serhal
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Clara Hebbo
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Mayssam Moussa
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Nada Alaaeddine
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
- * E-mail:
| |
Collapse
|
197
|
MDM2 promoter SNP55 (rs2870820) affects risk of colon cancer but not breast-, lung-, or prostate cancer. Sci Rep 2016; 6:33153. [PMID: 27624283 PMCID: PMC5022009 DOI: 10.1038/srep33153] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/22/2016] [Indexed: 12/11/2022] Open
Abstract
Two functional SNPs (SNP285G > C; rs117039649 and SNP309T > G; rs2279744) have previously been reported to modulate Sp1 transcription factor binding to the promoter of the proto-oncogene MDM2, and to influence cancer risk. Recently, a third SNP (SNP55C > T; rs2870820) was also reported to affect Sp1 binding and MDM2 transcription. In this large population based case-control study, we genotyped MDM2 SNP55 in 10,779 Caucasian individuals, previously genotyped for SNP309 and SNP285, including cases of colon (n = 1,524), lung (n = 1,323), breast (n = 1,709) and prostate cancer (n = 2,488) and 3,735 non-cancer controls, as well as 299 healthy African-Americans. Applying the dominant model, we found an elevated risk of colon cancer among individuals harbouring SNP55TT/CT genotypes compared to the SNP55CC genotype (OR = 1.15; 95% CI = 1.01-1.30). The risk was found to be highest for left-sided colon cancer (OR = 1.21; 95% CI = 1.00-1.45) and among females (OR = 1.32; 95% CI = 1.01-1.74). Assessing combined genotypes, we found the highest risk of colon cancer among individuals harbouring the SNP55TT or CT together with the SNP309TG genotype (OR = 1.21; 95% CI = 1.00-1.46). Supporting the conclusions from the risk estimates, we found colon cancer cases carrying the SNP55TT/CT genotypes to be diagnosed at younger age as compared to SNP55CC (p = 0.053), in particular among patients carrying the SNP309TG/TT genotypes (p = 0.009).
Collapse
|
198
|
STIP is a critical nuclear scaffolding protein linking USP7 to p53-Mdm2 pathway regulation. Oncotarget 2016; 6:34718-31. [PMID: 26460617 PMCID: PMC4741485 DOI: 10.18632/oncotarget.5303] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/28/2015] [Indexed: 11/25/2022] Open
Abstract
The ubiquitin-specific protease USP7 stabilizes both Mdm2 and p53 by removing ubiquitins, hence playing an important enzymatic role in the p53-Mdm2 pathway. However, it is poorly understood how USP7 executes its dual-stabilization effect on Mdm2 and p53 in cellular context. Here, we report that STIP is a novel macromolecular scaffold that links USP7 to the p53-Mdm2 pathway. STIP and a fraction of USP7 interact and constitutively colocalize in nucleoplasma. Overexpression of STIP stabilizes Mdm2 and p53, whereas downregulation of STIP decreases Mdm2 and p53 levels. The effect of STIP on Mdm2 and p53 depends on USP7 function as a deubiquitinating enzyme. Furthermore, we demonstrate that STIP mediates the assembly of two separate ternary protein complexes in vivo as STIP-USP7-Mdm2 and STIP-USP7-p53, which facilitates USP7-mediated stabilization of Mdm2 and p53. Collectively, these results pinpoint a new molecular function of STIP and reveal a novel mechanism whereby USP7 executes its dual-stabilization effect on Mdm2 and p53 via STIP scaffolding.
Collapse
|
199
|
Positive feedback regulation of p53 transactivity by DNA damage-induced ISG15 modification. Nat Commun 2016; 7:12513. [PMID: 27545325 PMCID: PMC4996943 DOI: 10.1038/ncomms12513] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/11/2016] [Indexed: 12/20/2022] Open
Abstract
p53 plays a pivotal role in tumour suppression under stresses, such as DNA damage. ISG15 has been implicated in the control of tumorigenesis. Intriguingly, the expression of ISG15, UBE1L and UBCH8 is induced by DNA-damaging agents, such as ultraviolet and doxorubicin, which are known to induce p53. Here, we show that the genes encoding ISG15, UBE1L, UBCH8 and EFP, have the p53-responsive elements and their expression is induced in a p53-dependent fashion under DNA damage conditions. Furthermore, DNA damage induces ISG15 conjugation to p53 and this modification markedly enhances the binding of p53 to the promoters of its target genes (for example, CDKN1 and BAX) as well as of its own gene by promoting phosphorylation and acetylation, leading to suppression of cell growth and tumorigenesis. These findings establish a novel feedback circuit between p53 and ISG15-conjugating system for positive regulation of the tumour suppressive function of p53 under DNA damage conditions. The ‘genome guardian' p53 has a well-established role in suppressing tumour development after DNA damage. Here the authors show that expression of the ubiquitin-like protein ISG15 is regulated by p53 which in turn is modified by ISG15 to enhance binding to target gene promoters.
Collapse
|
200
|
Lysine methylation represses p53 activity in teratocarcinoma cancer cells. Proc Natl Acad Sci U S A 2016; 113:9822-7. [PMID: 27535933 DOI: 10.1073/pnas.1610387113] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
TP53 (which encodes the p53 protein) is the most frequently mutated gene among all human cancers, whereas tumors that retain the wild-type TP53 gene often use alternative mechanisms to repress the p53 tumor-suppressive function. Testicular teratocarcinoma cells rarely contain mutations in TP53, yet the transcriptional activity of wild-type p53 is compromised, despite its high expression level. Here we report that in the teratocarcinoma cell line NTera2, p53 is subject to lysine methylation at its carboxyl terminus, which has been shown to repress p53's transcriptional activity. We show that reduction of the cognate methyltransferases reactivates p53 and promotes differentiation of the NTera2 cells. Furthermore, reconstitution of methylation-deficient p53 mutants into p53-depleted NTera2 cells results in elevated expression of p53 downstream targets and precocious loss of pluripotent gene expression compared with re-expression of wild-type p53. Our results provide evidence that lysine methylation of endogenous wild-type p53 represses its activity in cancer cells and suggest new therapeutic possibilities of targeting testicular teratocarcinoma.
Collapse
|