1
|
Liu L, Gong D, Sun H, Feng F, Xu J, Sun X, Gong L, Yu Z, Fang T, Xu Y, Lyu R, Wang T, Wang W, Tian W, Qiu L, An G, Hao M. DNp73 enhances tumor progression and immune evasion in multiple myeloma by targeting the MYC and MYCN pathways. Front Immunol 2024; 15:1470328. [PMID: 39380995 PMCID: PMC11459316 DOI: 10.3389/fimmu.2024.1470328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction Multiple myeloma (MM) is an incurable hematological malignancy with high chromosome instability and heavy dependence on the immunosuppressive bone marrow microenvironment. P53 mutations are adverse prognostic factors in MM; however, clinically, some patients without P53 mutations also exhibit aggressive disease progression. DNp73, an inhibitor of TP53 tumor suppressor family members, drives drug resistance and cancer progression in several solid malignancies. Nevertheless, the biological functions of DNp73 and the molecular mechanisms in myelomagenesis remain unclear. Methods The effects of DNp73 on proliferation and drug sensitivity were assessed using flow cytometry and xenograft models. To investigate the mechanisms of drug resistance, RNA-seq and ChIP-seq analyses were performed in MM cell lines, with validation by Western blot and RT-qPCR. Immunofluorescence and transwell assays were used to assess DNA damage and cell invasion in MM cells. Additionally, in vitro phagocytosis assays were conducted to confirm the role of DNp73 in immune evasion. Results Our study found that activation of NF-κB-p65 in multiple myeloma cells with different p53 mutation statuses upregulates DNp73 expression at the transcriptional level. Forced expression of DNp73 promoted aggressive proliferation and multidrug resistance in MM cells. Bulk RNA-seq analysis was conducted to assess the levels of MYCN, MYC, and CDK7. A ChIP-qPCR assay was used to reveal that DNp73 acts as a transcription factor regulating MYCN gene expression. Bulk RNA-seq analysis demonstrated increased levels of MYCN, MYC, and CDK7 with forced DNp73 expression in MM cells. A ChIP-qPCR assay revealed that DNp73 upregulates MYCN gene expression as a transcription factor. Additionally, DNp73 promoted immune evasion of MM cells by upregulating MYC target genes CD47 and PD-L1. Blockade of the CD47/SIRPα and PD-1/PD-L1 signaling pathways by the SIRPα-Fc fusion protein IMM01 and monoclonal antibody atezolizumab significantly restored the anti-MM activity of macrophages and T cells in the microenvironment, respectively. Discussion In summary, our study demonstrated for the first time that the p53 family member DNp73 remarkably induces proliferation, drug resistance, and immune escape of myeloma cells by directly targeting MYCN and regulating the MYC pathway. The oncogenic function of DNp73 is independent of p53 status in MM cells. These data contribute to a better understanding of the function of TP53 and its family members in tumorigenesis. Moreover, our study clarified that DNp73 overexpression not only promotes aggressive growth of tumor cells but, more importantly, promotes immune escape of MM cells through upregulation of immune checkpoints. DNp73 could serve as a biomarker for immunotherapy targeting PD-L1 and CD47 blockade in MM patients.
Collapse
Affiliation(s)
- Lanting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dasen Gong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin, China
| | - Hao Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Fangshuo Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jie Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xiyue Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lixin Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zhen Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Teng Fang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Rui Lyu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tingyu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wentian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenzhi Tian
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Gobroad Healthcare Group, Beijing, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
2
|
Li D, Kok CYL, Wang C, Ray D, Osterburg S, Dötsch V, Ghosh S, Sabapathy K. Dichotomous transactivation domains contribute to growth inhibitory and promotion functions of TAp73. Proc Natl Acad Sci U S A 2024; 121:e2318591121. [PMID: 38739802 PMCID: PMC11127001 DOI: 10.1073/pnas.2318591121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/22/2024] [Indexed: 05/16/2024] Open
Abstract
The transcription factor p73, a member of the p53 tumor-suppressor family, regulates cell death and also supports tumorigenesis, although the mechanistic basis for the dichotomous functions is poorly understood. We report here the identification of an alternate transactivation domain (TAD) located at the extreme carboxyl (C) terminus of TAp73β, a commonly expressed p73 isoform. Mutational disruption of this TAD significantly reduced TAp73β's transactivation activity, to a level observed when the amino (N)-TAD that is similar to p53's TAD, is mutated. Mutation of both TADs almost completely abolished TAp73β's transactivation activity. Expression profiling highlighted a unique set of targets involved in extracellular matrix-receptor interaction and focal adhesion regulated by the C-TAD, resulting in FAK phosphorylation, distinct from the N-TAD targets that are common to p53 and are involved in growth inhibition. Interestingly, the C-TAD targets are also regulated by the oncogenic, amino-terminal-deficient DNp73β isoform. Consistently, mutation of C-TAD reduces cellular migration and proliferation. Mechanistically, selective binding of TAp73β to DNAJA1 is required for the transactivation of C-TAD target genes, and silencing DNAJA1 expression abrogated all C-TAD-mediated effects. Taken together, our results provide a mechanistic basis for the dichotomous functions of TAp73 in the regulation of cellular growth through its distinct TADs.
Collapse
Affiliation(s)
- Dan Li
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore168583, Singapore
| | - Catherine Yen Li Kok
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore168583, Singapore
| | - Chao Wang
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore168583, Singapore
| | - Debleena Ray
- Programme in Cancer and Stem Cell Biology, Duke-National University of Singapore (NUS) Medical School, Singapore169857, Singapore
| | - Susanne Osterburg
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University, Frankfurt am Main60438, Germany
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University, Frankfurt am Main60438, Germany
| | - Sujoy Ghosh
- Centre for Computational Biology & Programme in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore169857, Singapore
| | - Kanaga Sabapathy
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore168583, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore637551, Singapore
| |
Collapse
|
3
|
Pützer BM, Sabapathy K. Editorial: Multidisciplinary Approaches in Exploring Cancer Heterogeneity, TME and Therapy Resistance: Perspectives for Systems Medicine. Front Cell Dev Biol 2022; 10:842596. [PMID: 35198561 PMCID: PMC8859833 DOI: 10.3389/fcell.2022.842596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Brigitte M. Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
- Department Life, Light & Matter, University of Rostock, Rostock, Germany
- *Correspondence: Brigitte M. Pützer,
| | - Kanaga Sabapathy
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
4
|
Wu Q, Liu F, Ge M, Laster KV, Wei L, Du R, Jiang M, Zhang J, Zhi Y, Jin G, Zhao S, Kim DJ, Dong Z, Liu K. BRD4 drives esophageal squamous cell carcinoma growth by promoting RCC2 expression. Oncogene 2022; 41:347-360. [PMID: 34750516 DOI: 10.1038/s41388-021-02099-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022]
Abstract
The low survival rate of esophageal squamous cell carcinoma patients is primarily attributed to technical limitations and a lack of insight regarding the molecular mechanisms contributing to its progression. Alterations in epigenetic modulators are critical to cancer development and prognosis. BRD4, a chromatin reader protein, plays an essential role in regulating oncogene expression. Here, we investigated the contributing role of BRD4 and its related mechanisms in the context of ESCC tumor progression. Our observations showed that BRD4 transcript and protein expression levels are significantly increased in ESCC patient tissues. Genetic or pharmacological inhibition of BRD4 suppressed ESCC cell proliferation in vitro and in vivo. Proteomic and transcriptomic analyses were subsequently used to deduce the potential targets of BRD4. Mechanistic studies showed that RCC2 is a downstream target of BRD4. Inhibition of either BRD4 or RCC2 resulted in decreased ESCC cell proliferation. The BRD4-TP73 interaction facilitated the binding of BRD4 complex to the promoter region of RCC2, and subsequently modulated RCC2 transcription. Furthermore, targeting BRD4 with inhibitors significantly decreased tumor volume in ESCC PDX models, indicating that BRD4 expression may contribute to tumor progression. Collectively, these findings suggest that BRD4 inhibition could be a promising strategy to treat ESCC by downregulating RCC2.
Collapse
Affiliation(s)
- Qiong Wu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Fangfang Liu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Mengmeng Ge
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | | | - Lixiao Wei
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Ruijuan Du
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Ming Jiang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Jing Zhang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Yafei Zhi
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China
| | - Guoguo Jin
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.,The Henan Luoyang Orthopedic Hospital, Zhengzhou, 450000, Henan, China
| | - Simin Zhao
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.,Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dong Joon Kim
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China.
| | - Zigang Dong
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Kangdong Liu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, 450000, China. .,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, 450000, Henan, China. .,Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, 450000, Henan, China. .,Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
5
|
Dual Role of p73 in Cancer Microenvironment and DNA Damage Response. Cells 2021; 10:cells10123516. [PMID: 34944027 PMCID: PMC8700694 DOI: 10.3390/cells10123516] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Understanding the mechanisms that regulate cancer progression is pivotal for the development of new therapies. Although p53 is mutated in half of human cancers, its family member p73 is not. At the same time, isoforms of p73 are often overexpressed in cancers and p73 can overtake many p53 functions to kill abnormal cells. According to the latest studies, while p73 represses epithelial–mesenchymal transition and metastasis, it can also promote tumour growth by modulating crosstalk between cancer and immune cells in the tumor microenvironment, M2 macrophage polarisation, Th2 T-cell differentiation, and angiogenesis. Thus, p73 likely plays a dual role as a tumor suppressor by regulating apoptosis in response to genotoxic stress or as an oncoprotein by promoting the immunosuppressive environment and immune cell differentiation.
Collapse
|
6
|
Rozenberg JM, Zvereva S, Dalina A, Blatov I, Zubarev I, Luppov D, Bessmertnyi A, Romanishin A, Alsoulaiman L, Kumeiko V, Kagansky A, Melino G, Ganini C, Barlev NA. The p53 family member p73 in the regulation of cell stress response. Biol Direct 2021; 16:23. [PMID: 34749806 PMCID: PMC8577020 DOI: 10.1186/s13062-021-00307-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
During oncogenesis, cells become unrestrictedly proliferative thereby altering the tissue homeostasis and resulting in subsequent hyperplasia. This process is paralleled by resumption of cell cycle, aberrant DNA repair and blunting the apoptotic program in response to DNA damage. In most human cancers these processes are associated with malfunctioning of tumor suppressor p53. Intriguingly, in some cases two other members of the p53 family of proteins, transcription factors p63 and p73, can compensate for loss of p53. Although both p63 and p73 can bind the same DNA sequences as p53 and their transcriptionally active isoforms are able to regulate the expression of p53-dependent genes, the strongest overlap with p53 functions was detected for p73. Surprisingly, unlike p53, the p73 is rarely lost or mutated in cancers. On the contrary, its inactive isoforms are often overexpressed in cancer. In this review, we discuss several lines of evidence that cancer cells develop various mechanisms to repress p73-mediated cell death. Moreover, p73 isoforms may promote cancer growth by enhancing an anti-oxidative response, the Warburg effect and by repressing senescence. Thus, we speculate that the role of p73 in tumorigenesis can be ambivalent and hence, requires new therapeutic strategies that would specifically repress the oncogenic functions of p73, while keeping its tumor suppressive properties intact.
Collapse
Affiliation(s)
- Julian M Rozenberg
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.
| | - Svetlana Zvereva
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Aleksandra Dalina
- The Engelhardt Institute of Molecular Biology, Russian Academy of Science, Moscow, Russia
| | - Igor Blatov
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Ilya Zubarev
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Daniil Luppov
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | | | - Alexander Romanishin
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia.,School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Lamak Alsoulaiman
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Vadim Kumeiko
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Alexander Kagansky
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia.,School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Gerry Melino
- Department of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Ganini
- Department of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nikolai A Barlev
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia. .,Institute of Cytology, Russian Academy of Science, Saint-Petersburg, Russia.
| |
Collapse
|
7
|
Distinct p63 and p73 Protein Interactions Predict Specific Functions in mRNA Splicing and Polyploidy Control in Epithelia. Cells 2020; 10:cells10010025. [PMID: 33375680 PMCID: PMC7824480 DOI: 10.3390/cells10010025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/20/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022] Open
Abstract
Epithelial organs are the first barrier against microorganisms and genotoxic stress, in which the p53 family members p63 and p73 have both overlapping and distinct functions. Intriguingly, p73 displays a very specific localization to basal epithelial cells in human tissues, while p63 is expressed in both basal and differentiated cells. Here, we analyse systematically the literature describing p63 and p73 protein-protein interactions to reveal distinct functions underlying the aforementioned distribution. We have found that p73 and p63 cooperate in the genome stability surveillance in proliferating cells; p73 specific interactors contribute to the transcriptional repression, anaphase promoting complex and spindle assembly checkpoint, whereas p63 specific interactors play roles in the regulation of mRNA processing and splicing in both proliferating and differentiated cells. Our analysis reveals the diversification of the RNA and DNA specific functions within the p53 family.
Collapse
|
8
|
Lin B, Xu J, Wang F, Wang J, Zhao H, Feng D. LncRNA XIST promotes myocardial infarction by regulating FOS through targeting miR-101a-3p. Aging (Albany NY) 2020; 12:7232-7247. [PMID: 32315985 PMCID: PMC7202499 DOI: 10.18632/aging.103072] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
The purpose of this study was to reveal the hypothesis that lncRNA X inactive specific transcript (XIST) can participate in the regulation of cardiomyocyte apoptosis in neonatal mice cardiomyocytes (NMCMs) and myocardial infarction (MI) through targeting miR-101a-3p. NMCMs were isolated from neonatal C57BL/6 mice and anoxia was induced in hypoxic chamber. MTT assay and flow cytometry were used to determine proliferation and apoptosis respectively. The target relationship among XIST, miR-101a-3p and FOS was revealed by bioinformatic analysis, luciferase reporter assay, pull-down assay and RNA immunoprecipitation assay. The expression of XIST, miR-101a-3p, FOS and apoptosis-related proteins was determined by qRT-PCR or western blot. MI model was constructed to reveal the role of XIST. We found that XIST was up-regulated in NMCMs under anoxia condition. Moreover, XIST increased FOS expression by sponging miR-101a-3p in anoxia cells. Silencing XIST expression improved cell viability and suppressed apoptosis in vitro and inhibited myocardial infarction by reducing the level of c-FOS and apoptosis-related proteins in vivo. Our findings suggest that XIST is involved in MI, modulation of its level can be used as a new strategy or potential target in the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Bin Lin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jing Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Feng Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jiaxiang Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hui Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Deguang Feng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
9
|
Wang C, Teo CR, Sabapathy K. p53-Related Transcription Targets of TAp73 in Cancer Cells-Bona Fide or Distorted Reality? Int J Mol Sci 2020; 21:ijms21041346. [PMID: 32079264 PMCID: PMC7072922 DOI: 10.3390/ijms21041346] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
Identification of p73 as a structural homolog of p53 fueled early studies aimed at determining if it was capable of performing p53-like functions. This led to a conundrum as p73 was discovered to be hardly mutated in cancers, and yet, TAp73, the full-length form, was found capable of performing p53-like functions, including transactivation of many p53 target genes in cancer cell lines. Generation of mice lacking p73/TAp73 revealed a plethora of developmental defects, with very limited spontaneous tumors arising only at a later stage. Concurrently, novel TAp73 target genes involved in cellular growth promotion that are not regulated by p53 were identified, mooting the possibility that TAp73 may have diametrically opposite functions to p53 in tumorigenesis. We have therefore comprehensively evaluated the TAp73 target genes identified and validated in human cancer cell lines, to examine their contextual relevance. Data from focused studies aimed at appraising if p53 targets are also regulated by TAp73—often by TAp73 overexpression in cell lines with non-functional p53—were affirmative. However, genome-wide and phenotype-based studies led to the identification of TAp73-regulated genes involved in cellular survival and thus, tumor promotion. Our analyses therefore suggest that TAp73 may not necessarily be p53’s natural substitute in enforcing tumor suppression. It has likely evolved to perform unique functions in regulating developmental processes and promoting cellular growth through entirely different sets of target genes that are not common to, and cannot be substituted by p53. The p53-related targets initially reported to be regulated by TAp73 may therefore represent an experimental possibility rather than the reality.
Collapse
Affiliation(s)
- Chao Wang
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore;
| | - Cui Rong Teo
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore;
| | - Kanaga Sabapathy
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore;
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore;
- Institute of Molecular and Cell Biology, Biopolis, Singapore 138673, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Correspondence:
| |
Collapse
|
10
|
Pan S, Shen M, Zhou M, Shi X, He R, Yin T, Wang M, Guo X, Qin R. Long noncoding RNA LINC01111 suppresses pancreatic cancer aggressiveness by regulating DUSP1 expression via microRNA-3924. Cell Death Dis 2019; 10:883. [PMID: 31767833 PMCID: PMC6877515 DOI: 10.1038/s41419-019-2123-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/26/2019] [Accepted: 11/04/2019] [Indexed: 12/19/2022]
Abstract
Dysfunction in long noncoding RNAs (lncRNAs) is reported to participate in the initiation and progression of human cancer; however, the biological functions and molecular mechanisms through which lncRNAs affect pancreatic cancer (PC) are largely unknown. Here, we report a novel lncRNA, LINC01111, that is clearly downregulated in PC tissues and plasma of PC patients and acts as a tumor suppressor. We found that the LINC01111 level was negatively correlated with the TNM stage but positively correlated with the survival of PC patients. The overexpression of LINC01111 significantly inhibited cell proliferation, the cell cycle, and cell invasion and migration in vitro, as well as tumorigenesis and metastasis in vivo. Conversely, the knockdown of LINC01111 enhanced cell proliferation, the cell cycle, and cell invasion and migration in vitro, as well as tumorigenesis and metastasis in vivo. Furthermore, we found that high expression levels of LINC01111 upregulated DUSP1 levels by sequestering miR-3924, resulting in the blockage of SAPK phosphorylation and the inactivation of the SAPK/JNK signaling pathway in PC cells and thus inhibiting PC aggressiveness. Overall, these data reveal that LINC01111 is a potential diagnostic biomarker for PC patients, and the newly identified LINC01111/miR-3924/DUSP1 axis can modulate PC initiation and development.
Collapse
Affiliation(s)
- Shutao Pan
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, Hubei, China
| | - Ming Shen
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, Hubei, China
| | - Min Zhou
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, Hubei, China
| | - Xiuhui Shi
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, Hubei, China
| | - Ruizhi He
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, Hubei, China
| | - Taoyuan Yin
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, Hubei, China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, Hubei, China
| | - Xingjun Guo
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, Hubei, China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, 430030, Wuhan, Hubei, China.
| |
Collapse
|
11
|
Identification of ARKL1 as a Negative Regulator of Epstein-Barr Virus Reactivation. J Virol 2019; 93:JVI.00989-19. [PMID: 31341047 DOI: 10.1128/jvi.00989-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
Epstein-Barr virus (EBV) maintains a life-long infection due to the ability to alternate between latent and lytic modes of replication. Lytic reactivation starts with derepression of the Zp promoter controlling BZLF1 gene expression, which binds and is activated by the c-Jun transcriptional activator. Here, we identified the cellular Arkadia-like 1 (ARKL1) protein as a negative regulator of Zp and EBV reactivation. Silencing of ARKL1 in the context of EBV-positive gastric carcinoma (AGS) cells, nasopharyngeal carcinoma (NPC43) cells, and B (M81) cells led to increased lytic protein expression, whereas overexpression inhibited BZLF1 expression. Similar effects of ARKL1 modulation were seen on BZLF1 transcripts as well as on Zp activity in Zp reporter assays, showing that ARKL1 repressed Zp. Proteomic profiling of ARKL1-host interactions identified c-Jun as an ARKL1 interactor, and reporter assays for Jun transcriptional activity showed that ARKL1 inhibited Jun activity. The ARKL1-Jun interaction required ARKL1 sequences that we previously showed mediated binding to the CK2 kinase regulatory subunit CK2β, suggesting that CK2β might mediate the ARKL1-Jun interaction. This model was supported by the findings that silencing of CK2β, but not the CK2α catalytic subunit, abrogated the ARKL1-Jun interaction and phenocopied ARKL1 silencing in promoting EBV reactivation. Additionally, ARKL1 was associated with Zp in reporter assays and this was increased by additional CK2β. Together, the data indicate that ARKL1 is a negative regulator of Zp and EBV reactivation that acts by inhibiting Jun activity through a CK2β-mediated interaction.IMPORTANCE Epstein-Barr virus (EBV) maintains a life-long infection due to the ability to alternate between latent and lytic modes of replication and is associated with several types of cancer. We have identified a cellular protein (ARKL1) that acts to repress the reactivation of EBV from the latent to the lytic cycle. We show that ARKL1 acts to repress transcription of the EBV lytic switch protein by inhibiting the activity of the cellular transcription factor c-Jun. This not only provides a new mechanism of regulating EBV reactivation but also identifies a novel cellular function of ARKL1 as an inhibitor of Jun-mediated transcription.
Collapse
|
12
|
Chang C, Wang H, Liu J, Pan C, Zhang D, Li X, Pan Y. Porphyromonas gingivalis Infection Promoted the Proliferation of Oral Squamous Cell Carcinoma Cells through the miR-21/PDCD4/AP-1 Negative Signaling Pathway. ACS Infect Dis 2019; 5:1336-1347. [PMID: 31243990 DOI: 10.1021/acsinfecdis.9b00032] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent epidemiological studies have revealed that Porphyromonas gingivalis, a major pathogen in periodontal disease, is associated with the development of oral squamous cell carcinoma (OSCC). However, the underlying mechanisms induced by P. gingivalis have not been well-defined. We aimed to determine the role of P. gingivalis in OSCC proliferation and the relevant molecular mechanisms. A cellular proliferation model of OSCC Tca8113 cells infected by P. gingivalis at a multiplicity of infection (MOI) of 50 was established. Cell proliferation was drastically increased in the infected cells compared with the control cells, while the proportion of cells in S phase was increased and the proportion of cells in G1 phase was decreased in the infected cells compared with the control cells. Additionally, the levels of activator protein 1 (AP-1; c-Jun and c-Fos) and its target gene cyclin D1 were increased in P. gingivalis-infected Tca8113 cells compared with control cells. miR-21 expression was elevated when programmed cell death 4 (PDCD4) expression was downregulated. Cyclin D1 expression was regulated by miR-21, PDCD4, and AP-1. The disruption of the pathway by silencing c-Jun, blocking miR-21 expression, or overexpressing PDCD4 led to decreased cyclin D1 expression and inhibited cell proliferation. P. gingivalis DNA levels were positively correlated with miR-21 and c-Jun expression and negatively correlated with PDCD4 expression in clinical OSCC samples. Our findings indicated that P. gingivalis might promote OSCC proliferation by regulating cyclin D1 expression via the miR-21/PDCD4/AP-1 negative feedback signaling pathway.
Collapse
Affiliation(s)
- Chunrong Chang
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Hongyan Wang
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Junchao Liu
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Chunling Pan
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Dongmei Zhang
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Xin Li
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Yaping Pan
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
- Department of Oral Biology, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| |
Collapse
|
13
|
Ou WB, Ni N, Zuo R, Zhuang W, Zhu M, Kyriazoglou A, Wu D, Eilers G, Demetri GD, Qiu H, Li B, Marino-Enriquez A, Fletcher JA. Cyclin D1 is a mediator of gastrointestinal stromal tumor KIT-independence. Oncogene 2019; 38:6615-6629. [PMID: 31371779 DOI: 10.1038/s41388-019-0894-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 02/22/2019] [Accepted: 04/03/2019] [Indexed: 12/19/2022]
Abstract
Oncogenic KIT or PDGFRA tyrosine kinase mutations are compelling therapeutic targets in most gastrointestinal stromal tumors (GISTs), and the KIT inhibitor, imatinib, is therefore standard of care for patients with metastatic GIST. However, some GISTs lose expression of KIT oncoproteins, and therefore become KIT-independent and are consequently resistant to KIT-inhibitor drugs. We identified distinctive biologic features in KIT-independent, imatinib-resistant GISTs as a step towards identifying drug targets in these poorly understood tumors. We developed isogenic GIST lines in which the parental forms were KIT oncoprotein-dependent, whereas sublines had loss of KIT oncoprotein expression, accompanied by markedly downregulated expression of the GIST biomarker, protein kinase C-theta (PRKCQ). Biologic mechanisms unique to KIT-independent GISTs were identified by transcriptome sequencing, qRT-PCR, immunoblotting, protein interaction studies, knockdown and expression assays, and dual-luciferase assays. Transcriptome sequencing showed that cyclin D1 expression was extremely low in two of three parental KIT-dependent GIST lines, whereas cyclin D1 expression was high in each of the KIT-independent GIST sublines. Cyclin D1 inhibition in KIT-independent GISTs had anti-proliferative and pro-apoptotic effects, associated with Rb activation and p27 upregulation. PRKCQ, but not KIT, was a negative regulator of cyclin D1 expression, whereas JUN and Hippo pathway effectors YAP and TAZ were positive regulators of cyclin D1 expression. PRKCQ, JUN, and the Hippo pathway coordinately regulate GIST cyclin D1 expression. These findings highlight the roles of PRKCQ, JUN, Hippo, and cyclin D1 as oncogenic mediators in GISTs that have converted, during TKI-therapy, to a KIT-independent state. Inhibitors of these pathways could be effective therapeutically for these now untreatable tumors.
Collapse
Affiliation(s)
- Wen-Bin Ou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China. .,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Nan Ni
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Rui Zuo
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Weihao Zhuang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Meijun Zhu
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Anastasios Kyriazoglou
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Duolin Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Grant Eilers
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - George D Demetri
- Ludwig Center at Dana-Farber/Harvard Cancer Center and Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Haibo Qiu
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Bin Li
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Division of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Adrian Marino-Enriquez
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jonathan A Fletcher
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
14
|
Context-dependent AMPK activation distinctly regulates TAp73 stability and transcriptional activity. Signal Transduct Target Ther 2018; 3:20. [PMID: 30057793 PMCID: PMC6062496 DOI: 10.1038/s41392-018-0020-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 04/13/2018] [Accepted: 05/14/2018] [Indexed: 12/17/2022] Open
Abstract
TAp73, the homologue of the tumour suppressor p53, has dual roles in tumourigenesis: both as a tumour suppressor and as a promoter of tumour growth. We have recently shown that hypoxia, a condition prevalent in tumours, results in the stabilisation of TAp73 through a mechanism involving HIF-1α-mediated repression of the E3 ligase Siah1. Elevated TAp73 in turn regulates the angiogenic transcriptional programme, exemplified by vegf-A activation, thereby promoting angiogenesis and tumour growth. To further understand hypoxia-mediated TAp73 regulation, we have focused on the Adenosine monophosphate (AMP)-dependent protein kinase (AMPK) signalling pathway induced by hypoxia. We show that hypoxia-mediated AMPK activation is required for efficient TAp73 stabilisation, through multiple means by using AMPK-deficient cells or inhibiting its activity and expression. Conversely, direct AMPK activation using its activator AICAR is also sufficient to induce TAp73 stabilisation but this is independent of putative AMPK phosphorylation sites on TAp73, HIF-1α activation, and transcriptional repression of Siah1. Furthermore, while vegf-A up-regulation upon hypoxia requires AMPK, direct activation of AMPK by AICAR does not activate vegf-A. Consistently, supernatant from cells exposed to hypoxia, but not AICAR, was able to induce tube formation in HUVECs. These data therefore highlight that the processes of TAp73 stabilisation and transcriptional activation of angiogenic target genes by AMPK activation can be decoupled. Collectively, these results suggest that the context of AMPK activation determines the effect on TAp73, and proposes a model in which hypoxia-induced TAp73 stabilisation occurs by parallel pathways converging to mediate its transactivation potential. The stabilisation of an important signalling protein can fuel tumour growth and progression—but only under the right environmental conditions. Paradoxically, the TAp73 protein can both suppress tumorigenesis and stimulate formation of tumour-feeding blood vessels. The latter effect appears to be linked with exposure to oxygen-poor conditions within solid tumours, and researchers led by Kanaga Sabapathy at Singapore’s National Cancer Centre recently explored the mechanisms regulating TAp73 activity. Sabapathy’s team showed that the action of a protein called AMPK helps to prevent TAp73 from being degraded. However, activation of AMPK in isolation is insufficient to promote TAp73-mediated blood vessel growth. Instead, the researchers only observed this effect when AMPK was specifically activated due to oxygen deprivation, revealing the existence of multiple TAp73-regulating pathways that could explain this protein’s seemingly contradictory effects on cell growth.
Collapse
|
15
|
Xie N, Vikhreva P, Annicchiarico-Petruzzelli M, Amelio I, Barlev N, Knight RA, Melino G. Integrin-β4 is a novel transcriptional target of TAp73. Cell Cycle 2018; 17:589-594. [PMID: 29233040 DOI: 10.1080/15384101.2017.1403684] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As a member of p53 family, p73 has attracted intense investigations due to its structural and functional similarities to p53. Among more than ten p73 variants, the transactivation (TA) domain-containing isoform TAp73 is the one that imitates the p53's behavior most. TAp73 induces apoptosis and cell cycle arrest, which endows it the capacity of tumour suppression. Also, it can exert diverse biological influences on cells through activating a complex and context dependent transcriptional programme. The transcriptional activities further broaden its roles in more intricate biological processes. In this article, we report that p73 is a positive regulator of a cell adhesion related gene named integrin β4 (ITGB4). This finding may have implications for the dissection of the biological mechanisms underlining p73 functions.
Collapse
Affiliation(s)
- Ningxia Xie
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom.,b Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Rome 00133 , Italy
| | - Polina Vikhreva
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom
| | | | - Ivano Amelio
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom
| | - Nicolai Barlev
- d Institute of Cytology Russian Academy of Sciences , Saint-Petersburg , 194064 , Russia
| | - Richard A Knight
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom
| | - Gerry Melino
- a MRC Toxicology Unit , Hodgkin Building , Lancaster Road, Leicester LE1 9HN , United Kingdom.,b Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Rome 00133 , Italy.,d Institute of Cytology Russian Academy of Sciences , Saint-Petersburg , 194064 , Russia
| |
Collapse
|
16
|
Transcriptional regulation of P63 on the apoptosis of male germ cells and three stages of spermatogenesis in mice. Cell Death Dis 2018; 9:76. [PMID: 29362488 PMCID: PMC5833356 DOI: 10.1038/s41419-017-0046-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/06/2017] [Accepted: 10/09/2017] [Indexed: 12/13/2022]
Abstract
Infertility affects 10-15% of couples worldwide, and male factors account for 50%. Spermatogenesis is precisely regulated by genetic factors, and the mutations of genes result in abnormal spermatogenesis and eventual male infertility. The aim of this study was to explore the role and transcriptional regulation of P63 in the apoptosis and mouse spermatogenesis. P63 protein was decreased in male germ cells of P63(+/-) mice compared with wild-type mice. There was no obvious difference in testis weight, sperm motility, and fecundity between P63(+/-) and wild-type mice. However, abnormal germ cells were frequently observed in P63(+/-) mice at 2 months old. Notably, apoptotic male germ cells and the percentage of abnormal sperm were significantly enhanced in P63(+/-) mice compared to wild-type mice. Spermatogonia, pachytene spermatocytes and round spermatids were isolated from P63(+/-) and wild-type mice using STA-PUT velocity sedimentation, and they were identified phenotypically with high purities. RNA sequencing demonstrated distinct transcription profiles in spermatogonia, pachytene spermatocytes, and round spermatids between P63(+/-) mice and wild-type mice. In total, there were 645 differentially expressed genes (DEGs) in spermatogonia, 106 DEGs in pachytene spermatocytes, and 1152 in round spermatids between P63(+/-) mice and wild-type mice. Real time PCR verified a number of DEGs identified by RNA sequencing. Gene ontology annotation and pathway analyzes further indicated that certain key genes, e.g., Ccnd2, Tgfa, Hes5, Insl3, Kit, Lef1, and Jun were involved in apoptosis, while Dazl, Kit, Pld6, Cdkn2d, Stra8, and Ubr2 were associated with regulating spermatogenesis. Collectively, these results implicate that P63 mediates the apoptosis of male germ cells and regulates three stages of spermatogenesis transcriptionally. This study could provide novel targets for the diagnosis and treatment of male infertility.
Collapse
|
17
|
Si H, Lu H, Yang X, Mattox A, Jang M, Bian Y, Sano E, Viadiu H, Yan B, Yau C, Ng S, Lee SK, Romano RA, Davis S, Walker RL, Xiao W, Sun H, Wei L, Sinha S, Benz CC, Stuart JM, Meltzer PS, Van Waes C, Chen Z. TNF-α modulates genome-wide redistribution of ΔNp63α/TAp73 and NF-κB cREL interactive binding on TP53 and AP-1 motifs to promote an oncogenic gene program in squamous cancer. Oncogene 2016; 35:5781-5794. [PMID: 27132513 PMCID: PMC5093089 DOI: 10.1038/onc.2016.112] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 01/11/2016] [Accepted: 01/19/2016] [Indexed: 12/11/2022]
Abstract
The Cancer Genome Atlas (TCGA) network study of 12 cancer types (PanCancer 12) revealed frequent mutation of TP53, and amplification and expression of related TP63 isoform ΔNp63 in squamous cancers. Further, aberrant expression of inflammatory genes and TP53/p63/p73 targets were detected in the PanCancer 12 project, reminiscent of gene programs comodulated by cREL/ΔNp63/TAp73 transcription factors we uncovered in head and neck squamous cell carcinomas (HNSCCs). However, how inflammatory gene signatures and cREL/p63/p73 targets are comodulated genome wide is unclear. Here, we examined how the inflammatory factor tumor necrosis factor-α (TNF-α) broadly modulates redistribution of cREL with ΔNp63α/TAp73 complexes and signatures genome wide in the HNSCC model UM-SCC46 using chromatin immunoprecipitation sequencing (ChIP-seq). TNF-α enhanced genome-wide co-occupancy of cREL with ΔNp63α on TP53/p63 sites, while unexpectedly promoting redistribution of TAp73 from TP53 to activator protein-1 (AP-1) sites. cREL, ΔNp63α and TAp73 binding and oligomerization on NF-κB-, TP53- or AP-1-specific sequences were independently validated by ChIP-qPCR (quantitative PCR), oligonucleotide-binding assays and analytical ultracentrifugation. Function of the binding activity was confirmed using TP53-, AP-1- and NF-κB-specific REs or p21, SERPINE1 and IL-6 promoter luciferase reporter activities. Concurrently, TNF-α regulated a broad gene network with cobinding activities for cREL, ΔNp63α and TAp73 observed upon array profiling and reverse transcription-PCR. Overlapping target gene signatures were observed in squamous cancer subsets and in inflamed skin of transgenic mice overexpressing ΔNp63α. Furthermore, multiple target genes identified in this study were linked to TP63 and TP73 activity and increased gene expression in large squamous cancer samples from PanCancer 12 TCGA by CircleMap. PARADIGM inferred pathway analysis revealed the network connection of TP63 and NF-κB complexes through an AP-1 hub, further supporting our findings. Thus, inflammatory cytokine TNF-α mediates genome-wide redistribution of the cREL/p63/p73, and AP-1 interactome, to diminish TAp73 tumor suppressor function and reciprocally activate NF-κB and AP-1 gene programs implicated in malignancy.
Collapse
Affiliation(s)
- Han Si
- Tumor Biology Section, Head and Neck Surgery Branch,
National Institute on Deafness and Other Communication Disorders, NIH, Bethesda,
Maryland, USA
| | - Hai Lu
- Orthopaedic Center, Zhujiang Hospital Guangzhou, Guangdong,
China
| | - Xinping Yang
- Tumor Biology Section, Head and Neck Surgery Branch,
National Institute on Deafness and Other Communication Disorders, NIH, Bethesda,
Maryland, USA
| | - Austin Mattox
- Tumor Biology Section, Head and Neck Surgery Branch,
National Institute on Deafness and Other Communication Disorders, NIH, Bethesda,
Maryland, USA
| | - Minyoung Jang
- Tumor Biology Section, Head and Neck Surgery Branch,
National Institute on Deafness and Other Communication Disorders, NIH, Bethesda,
Maryland, USA
| | - Yansong Bian
- Tumor Biology Section, Head and Neck Surgery Branch,
National Institute on Deafness and Other Communication Disorders, NIH, Bethesda,
Maryland, USA
| | - Eleanor Sano
- Department of Chemistry and Biochemistry, University of
California, San Diego, La Jolla, CA
| | - Hector Viadiu
- Instituto de Química, Universidad Nacional
Autónoma de México (UNAM), Circuito Exterior, Ciudad Universitaria,
Mexico City, D.F. 04510, MÉXICO
| | - Bin Yan
- LKS Faculty of Medicine and School of Biomedical Sciences,
LKS Faculty of Medicine and Center of Genome Sciences, The University of Hong Kong,
Hong Kong, China
| | | | - Sam Ng
- Department of Biomolecular Engineering, Center for
Biomolecular Sciences and Engineering, University of California, Santa Cruz, Santa
Cruz, CA
| | - Steven K. Lee
- Tumor Biology Section, Head and Neck Surgery Branch,
National Institute on Deafness and Other Communication Disorders, NIH, Bethesda,
Maryland, USA
| | - Rose-Anne Romano
- Department of Biochemistry, State University of New York at
Buffalo, Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New
York, USA
| | - Sean Davis
- Cancer Genetics Branch, National Cancer Institute,
Bethesda, Maryland, USA
| | - Robert L. Walker
- Cancer Genetics Branch, National Cancer Institute,
Bethesda, Maryland, USA
| | - Wenming Xiao
- Division of Bioinformatics and Biostatistics, National
Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson,
Arkansas
| | - Hongwei Sun
- Biodata Mining and Discovery Section, National Institute
of Arthritis, Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Lai Wei
- Clinical Immunology Section, National Eye Institute, NIH,
Bethesda, Maryland, USA
- State Key Laboratory of Ophthalmology, Zhongshan
Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Satrajit Sinha
- Department of Biochemistry, State University of New York at
Buffalo, Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New
York, USA
| | | | - Joshua M. Stuart
- Department of Biomolecular Engineering, Center for
Biomolecular Sciences and Engineering, University of California, Santa Cruz, Santa
Cruz, CA
| | - Paul S. Meltzer
- Cancer Genetics Branch, National Cancer Institute,
Bethesda, Maryland, USA
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch,
National Institute on Deafness and Other Communication Disorders, NIH, Bethesda,
Maryland, USA
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch,
National Institute on Deafness and Other Communication Disorders, NIH, Bethesda,
Maryland, USA
| |
Collapse
|
18
|
Dulloo I, Hooi PB, Sabapathy K. Hypoxia-induced DNp73 stabilization regulates Vegf-A expression and tumor angiogenesis similar to TAp73. Cell Cycle 2016; 14:3533-9. [PMID: 26267146 PMCID: PMC4825702 DOI: 10.1080/15384101.2015.1078038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
P73, the homolog of p53, exists in 2 major forms: either as a pro-apoptotic TAp73 or an amino-terminally truncated DNp73, the latter lacking the first transactivation domain. While TAp73s tumor suppressive functions have been established, DNp73 is an anti-apoptotic protein conferring chemoresistance and is associated with poor survival. However, both forms are variably overexpressed in many human cancers. In this context, we have recently demonstrated that TAp73 is stabilized by hypoxia, a tumor-relevant condition that is associated with cell survival, via HIF-1α-mediated suppression of Siah1 E3 ligase that degrades TAp73. Consequently, hypoxic signals lead to TAp73-mediated activation of several angiogenic genes and blood vessel formation, thereby supporting tumorigenesis. We show here that, similar to TAp73, DNp73 is stabilized by hypoxia in a HIF-1α-dependent manner, which otherwise is degraded by Siah1. Moreover, DNp73 is capable of inducing the expression of Vegf-A, the prototypic angiogenic gene, and loss of DNp73 expression results in reduction in tumor vasculature and size. These data therefore indicate a common mode of regulation for both p73 forms by hypoxia, resulting in the promotion of angiogenesis and tumor growth, highlighting common functionality of these antagonistic proteins under specific physiological contexts.
Collapse
Affiliation(s)
- Iqbal Dulloo
- a Division of Cellular & Molecular Research; Humphrey Oei Institute of Cancer Research; National Cancer Centre ; Singapore
| | - Phang Beng Hooi
- a Division of Cellular & Molecular Research; Humphrey Oei Institute of Cancer Research; National Cancer Centre ; Singapore
| | - Kanaga Sabapathy
- a Division of Cellular & Molecular Research; Humphrey Oei Institute of Cancer Research; National Cancer Centre ; Singapore.,b Cancer and Stem Cell Biology Program; Duke-NUS Graduate Medical School ; Singapore.,c Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore ; Singapore
| |
Collapse
|
19
|
Abstract
The role of p73, the homologue of the tumor suppressor p53, in regulating angiogenesis has recently been extensively investigated, resulting in the publication of five articles. Of these, two studies suggested a suppressive role, while the others implied a stimulatory role for the p73 isoforms in regulating angiogenesis. A negative role for TAp73, the full-length form that is often associated with tumor suppression, in blood vessel formation, is consistent with its general attributes and was proposed to be effected indirectly through the degradation of hypoxia-inducible factor 1α (HIF1-α), the master angiogenic regulator. In contrast, a positive role for TAp73 coincides with its recently understood role in supporting cellular survival and thus tumorigenesis, consistent with TAp73 being not-mutated but rather often overexpressed in clinical contexts. In the latter case, TAp73 expression was induced by hypoxia via HIF1-α, and it appears to directly promote angiogenic target gene activation and blood vessel formation independent of HIF1-α. This mini review will provide an overview of these seemingly opposite recent findings as well as earlier data, which collectively establish the definite possibility that TAp73 is indeed capable of both promoting and inhibiting angiogenesis, depending on the cellular context.
Collapse
|
20
|
Dhillon AS, Tulchinsky E. FRA-1 as a driver of tumour heterogeneity: a nexus between oncogenes and embryonic signalling pathways in cancer. Oncogene 2015; 34:4421-8. [PMID: 25381818 PMCID: PMC4351906 DOI: 10.1038/onc.2014.374] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 10/02/2014] [Accepted: 10/02/2014] [Indexed: 12/12/2022]
Abstract
Tumour heterogeneity is a major factor undermining the success of therapies targeting metastatic cancer. Two major theories are thought to explain the phenomenon of heterogeneity in cancer--clonal evolution and cell plasticity. In this review, we examine a growing body of work implicating the transcription factor FOS-related antigen 1 (FRA-1) as a central node in tumour cell plasticity networks, and discuss mechanisms regulating its activity in cancer cells. We also discuss evidence from the FRA-1 perspective supporting the notion that clonal selection and cell plasticity represent two sides of the same coin. We propose that FRA-1-overexpressing clones featuring high plasticity undergo positive selection during consecutive stages of multistep tumour progression. This model underscores a potential mechanism through which tumour cells retaining elevated levels of plasticity acquire a selective advantage over other clonal populations within a tumour.
Collapse
Affiliation(s)
- A S Dhillon
- Research Division, Peter MacCallum Cancer Center, St Andrews Place, East Melbourne, Melbourne, Victoria 3002, Australia
| | - E Tulchinsky
- Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester, UK
| |
Collapse
|
21
|
Abstract
p53 plays a key role in regulating DNA damage response by suppressing cell cycle progression or inducing apoptosis depending on extent of DNA damage. However, it is not clear why mild genotoxic stress favors growth arrest, whereas excessive lesions signal cells to die. Here we showed that TAp73, a p53 homologue thought to have a similar function as p53, restrains the transcriptional activity of p53 and prevents excessive activation of its downstream targets upon low levels of DNA damage, which results in cell cycle arrest. Extensive DNA damage triggers TAp73 depletion through ubiquitin/proteasome-mediated degradation of E2F1, leading to enhanced transcriptional activation by p53 and subsequent induction of apoptosis. These findings provide novel insights into the regulation of p53 function and suggest that TAp73 keeps p53 activity in check in regulating cell fate decisions upon genotoxic stress.
Collapse
|
22
|
Subramanian D, Bunjobpol W, Sabapathy K. Interplay between TAp73 Protein and Selected Activator Protein-1 (AP-1) Family Members Promotes AP-1 Target Gene Activation and Cellular Growth. J Biol Chem 2015; 290:18636-49. [PMID: 26018080 PMCID: PMC4513121 DOI: 10.1074/jbc.m115.636548] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Indexed: 12/22/2022] Open
Abstract
Unlike p53, which is mutated at a high rate in human cancers, its homologue p73 is not mutated but is often overexpressed, suggesting a possible context-dependent role in growth promotion. Previously, we have shown that co-expression of TAp73 with the proto-oncogene c-Jun can augment cellular growth and potentiate transactivation of activator protein (AP)-1 target genes such as cyclin D1. Here, we provide further mechanistic insights into the cooperative activity between these two transcription factors. Our data show that TAp73-mediated AP-1 target gene transactivation relies on c-Jun dimerization and requires the canonical AP-1 sites on target gene promoters. Interestingly, only selected members of the Fos family of proteins such as c-Fos and Fra1 were found to cooperate with TAp73 in a c-Jun-dependent manner to transactivate AP-1 target promoters. Inducible expression of TAp73 led to the recruitment of these Fos family members to the AP-1 target promoters on which TAp73 was found to be bound near the AP-1 site. Consistent with the binding of TAp73 and AP-1 members on the target promoters in a c-Jun-dependent manner, TAp73 was observed to physically interact with c-Jun specifically at the chromatin via its carboxyl-terminal region. Furthermore, co-expression of c-Fos or Fra1 was able to cooperate with TAp73 in potentiating cellular growth, similarly to c-Jun. These data together suggest that TAp73 plays a vital role in activation of AP-1 target genes via direct binding to c-Jun at the target promoters, leading to enhanced loading of other AP-1 family members, thereby leading to cellular growth.
Collapse
Affiliation(s)
- Deepa Subramanian
- From the Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore
| | - Wilawan Bunjobpol
- From the Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore
| | - Kanaga Sabapathy
- From the Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore, Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, 8 College Road, Singapore 169857, Singapore, and Department of Biochemistry, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
| |
Collapse
|
23
|
Hypoxia-inducible TAp73 supports tumorigenesis by regulating the angiogenic transcriptome. Nat Cell Biol 2015; 17:511-23. [PMID: 25774835 DOI: 10.1038/ncb3130] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/06/2015] [Indexed: 12/20/2022]
Abstract
The functional significance of the overexpression of unmutated TAp73, a homologue of the tumour suppressor p53, in multiple human cancers is unclear, but raises the possibility of unidentified roles in promoting tumorigenesis. We show here that TAp73 is stabilized by hypoxia, a condition highly prevalent in tumours, through HIF-1α-mediated repression of the ubiquitin ligase Siah1, which targets TAp73 for degradation. Consequently, TAp73-deficient tumours are less vascular and reduced in size, and conversely, TAp73 overexpression leads to increased vasculature. Moreover, we show that TAp73 is a critical regulator of the angiogenic transcriptome and is sufficient to directly activate the expression of several angiogenic genes. Finally, expression of TAp73 positively correlates with these angiogenic genes in several human tumours, and the angiogenic gene signature is sufficient to segregate the TAp73(Hi)- from TAp73(Low)-expressing tumours. These data demonstrate a pro-angiogenic role for TAp73 in supporting tumorigenesis, providing a rationale for its overexpression in cancers.
Collapse
|
24
|
Osman AA, Neskey DM, Katsonis P, Patel AA, Ward AM, Hsu TK, Hicks SC, McDonald TO, Ow TJ, Alves MO, Pickering CR, Skinner HD, Zhao M, Sturgis EM, Kies MS, El-Naggar A, Perrone F, Licitra L, Bossi P, Kimmel M, Frederick MJ, Lichtarge O, Myers JN. Evolutionary Action Score of TP53 Coding Variants Is Predictive of Platinum Response in Head and Neck Cancer Patients. Cancer Res 2015; 75:1205-15. [PMID: 25691460 DOI: 10.1158/0008-5472.can-14-2729] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 01/12/2015] [Indexed: 12/25/2022]
Abstract
TP53 is the most frequently altered gene in head and neck squamous cell carcinoma (HNSCC), with mutations occurring in over two thirds of cases; however, the predictive response of these mutations to cisplatin-based therapy remains elusive. In the current study, we evaluate the ability of the Evolutionary Action score of TP53-coding variants (EAp53) to predict the impact of TP53 mutations on response to chemotherapy. The EAp53 approach clearly identifies a subset of high-risk TP53 mutations associated with decreased sensitivity to cisplatin both in vitro and in vivo in preclinical models of HNSCC. Furthermore, EAp53 can predict response to treatment and, more importantly, a survival benefit for a subset of head and neck cancer patients treated with platinum-based therapy. Prospective evaluation of this novel scoring system should enable more precise treatment selection for patients with HNSCC.
Collapse
Affiliation(s)
- Abdullah A Osman
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David M Neskey
- Department of Otolaryngology Head and Neck Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Panagiotis Katsonis
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas
| | - Ameeta A Patel
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandra M Ward
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Teng-Kuei Hsu
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas
| | - Stephanie C Hicks
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Thomas J Ow
- Department of Otolaryngology Head and Neck Surgery, Albert Einstein School of Medicine, Bronx, New York
| | - Marcus Ortega Alves
- Department of Internal Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Curtis R Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heath D Skinner
- Department of Thoracic Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mei Zhao
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric M Sturgis
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Merrill S Kies
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adel El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Federica Perrone
- Department of Pathology, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale Tumori, Milan, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Unit, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale Tumori, Milan, Italy
| | - Paolo Bossi
- Head and Neck Medical Oncology Unit, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale Tumori, Milan, Italy
| | - Marek Kimmel
- Department of Statistics, Rice University, Houston, Texas
| | - Mitchell J Frederick
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Olivier Lichtarge
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
25
|
Gonen-Korkmaz C, Sevin G, Gokce G, Arun MZ, Yildirim G, Reel B, Kaymak A, Ogut D. Analysis of tumor necrosis factor α-induced and nuclear factor κB-silenced LNCaP prostate cancer cells by RT-qPCR. Exp Ther Med 2014; 8:1695-1700. [PMID: 25371717 PMCID: PMC4218634 DOI: 10.3892/etm.2014.2032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 10/06/2014] [Indexed: 01/20/2023] Open
Abstract
Prostate cancer is the second leading cause of morbidity and mortality in males in the Western world. In the present study, LNCaP, which is an androgen receptor-positive and androgen-responsive prostate cancer cell line derived from lymph node metastasis, and DU145, which is an androgen receptor-negative prostate cancer cell line derived from brain metastasis, were investigated. TNFα treatment decreased p105 and p50 expression and R1881 treatment slightly decreased p105 expression but increased p50 expression with or without TNFα induction. As an aggressive prostate cancer cell line, DU145 transfected with six transmembrane protein of prostate (STAMP)1 or STAMP2 was also exposed to TNFα. Western blotting indicated that transfection with either STAMP gene caused a significant increase in NFκB expression following TNFα induction. In addition, following the treatment of LNCaP cells with TNFα, reverse transcription quantitative polymerase chain reaction (RT-qPCR) was performed with a panel of apoptosis-related gene primers. The apoptosis-related genes p53, p73, caspase 7 and caspase 9 showed statistically significant increases in expression levels while the expression levels of MDM2 and STAMP1 decreased following TNFα induction. Furthermore, LNCaP cells were transfected with a small interfering NFκB (siNFκB) construct for 1 and 4 days and induced with TNFα for the final 24 h. RT-qPCR amplifications were performed with apoptosis-related gene primers, including p53, caspases and STAMPs. However, no changes in the level of STAMP2 were observed between cells in the presence or absence of TNFα induction or between those transfected or not transfected with siNFκB; however, the level of STAMP1 was significantly decreased by TNFα induction, and significantly increased with siNFκB transfection. Silencing of the survival gene NFκB caused anti-apoptotic STAMP1 expression to increase, which repressed p53, together with MDM2. NFκB silencing had varying effects on a panel of cancer regulatory genes. Therefore, the effective inhibition of NFκB may be critical in providing a targeted pathway for prostate cancer prevention.
Collapse
Affiliation(s)
- Ceren Gonen-Korkmaz
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| | - Gulnur Sevin
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| | - Goksel Gokce
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| | - Mehmet Zuhuri Arun
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| | - Gokce Yildirim
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| | - Buket Reel
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| | - Aysegul Kaymak
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| | - Deniz Ogut
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| |
Collapse
|
26
|
Ji X, Huang Q, Yu L, Nussinov R, Ma B. Bioinformatics study of cancer-related mutations within p53 phosphorylation site motifs. Int J Mol Sci 2014; 15:13275-98. [PMID: 25075982 PMCID: PMC4159794 DOI: 10.3390/ijms150813275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 02/06/2023] Open
Abstract
p53 protein has about thirty phosphorylation sites located at the N- and C-termini and in the core domain. The phosphorylation sites are relatively less mutated than other residues in p53. To understand why and how p53 phosphorylation sites are rarely mutated in human cancer, using a bioinformatics approaches, we examined the phosphorylation site and its nearby flanking residues, focusing on the consensus phosphorylation motif pattern, amino-acid correlations within the phosphorylation motifs, the propensity of structural disorder of the phosphorylation motifs, and cancer mutations observed within the phosphorylation motifs. Many p53 phosphorylation sites are targets for several kinases. The phosphorylation sites match 17 consensus sequence motifs out of the 29 classified. In addition to proline, which is common in kinase specificity-determining sites, we found high propensity of acidic residues to be adjacent to phosphorylation sites. Analysis of human cancer mutations in the phosphorylation motifs revealed that motifs with adjacent acidic residues generally have fewer mutations, in contrast to phosphorylation sites near proline residues. p53 phosphorylation motifs are mostly disordered. However, human cancer mutations within phosphorylation motifs tend to decrease the disorder propensity. Our results suggest that combination of acidic residues Asp and Glu with phosphorylation sites provide charge redundancy which may safe guard against loss-of-function mutations, and that the natively disordered nature of p53 phosphorylation motifs may help reduce mutational damage. Our results further suggest that engineering acidic amino acids adjacent to potential phosphorylation sites could be a p53 gene therapy strategy.
Collapse
Affiliation(s)
- Xiaona Ji
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Qiang Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Long Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, MD 21702, USA.
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
27
|
Kasim V, Huang C, Zhang J, Jia H, Wang Y, Yang L, Miyagishi M, Wu S. Synergistic cooperation of MDM2 and E2F1 contributes to TAp73 transcriptional activity. Biochem Biophys Res Commun 2014; 449:319-26. [PMID: 24845566 DOI: 10.1016/j.bbrc.2014.05.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 05/10/2014] [Indexed: 11/24/2022]
Abstract
TAp73, a structural homologue of p53, plays an important role in tumorigenesis. E2F1 had been reported as a transcriptional regulator of TAp73, however, the detailed mechanism remains to be elucidated. Here we reported that MDM2-silencing reduced the activities of the TAp73 promoters and the endogenous TAp73 expression level significantly; while MDM2 overexpression upregulated them. We further revealed that the regulation of TAp73 transcriptional activity occurs as a synergistic effect of MDM2 and E2F1, most probably through their physical interaction in the nuclei. Furthermore, we also suggested that MDM2 might be involved in DNA damage-induced TAp73 transcriptional activity. Finally, we elucidated that MDM2-silencing reduced the proliferation rate of colon carcinoma cells regardless of the p53 status. Our data show a synergistic effect of MDM2 and E2F1 on TAp73 transcriptional activity, suggesting a novel regulation pathway of TAp73.
Collapse
Affiliation(s)
- Vivi Kasim
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Can Huang
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jing Zhang
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Huizhen Jia
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yunxia Wang
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Li Yang
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Makoto Miyagishi
- Molecular Composite Medicine Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Shourong Wu
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
28
|
Rozenberg JM, Bhattacharya P, Chatterjee R, Glass K, Vinson C. Combinatorial recruitment of CREB, C/EBPβ and c-Jun determines activation of promoters upon keratinocyte differentiation. PLoS One 2013; 8:e78179. [PMID: 24244291 PMCID: PMC3820678 DOI: 10.1371/journal.pone.0078179] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/12/2013] [Indexed: 01/11/2023] Open
Abstract
Background Transcription factors CREB, C/EBPβ and Jun regulate genes involved in keratinocyte proliferation and differentiation. We questioned if specific combinations of CREB, C/EBPβ and c-Jun bound to promoters correlate with RNA polymerase II binding, mRNA transcript levels and methylation of promoters in proliferating and differentiating keratinocytes. Results Induction of mRNA and RNA polymerase II by differentiation is highest when promoters are bound by C/EBP β alone, C/EBPβ together with c-Jun, or by CREB, C/EBPβ and c-Jun, although in this case CREB binds with low affinity. In contrast, RNA polymerase II binding and mRNA levels change the least upon differentiation when promoters are bound by CREB either alone or in combination with C/EBPβ or c-Jun. Notably, promoters bound by CREB have relatively high levels of RNA polymerase II binding irrespective of differentiation. Inhibition of C/EBPβ or c-Jun preferentially represses mRNA when gene promoters are bound by corresponding transcription factors and not CREB. Methylated promoters have relatively low CREB binding and, accordingly, those which are bound by C/EBPβ are induced by differentiation irrespective of CREB. Composite “Half and Half” consensus motifs and co localizing consensus DNA binding motifs are overrepresented in promoters bound by the combination of corresponding transcription factors. Conclusion Correlational and functional data describes combinatorial mechanisms regulating the activation of promoters. Colocalization of C/EBPβ and c-Jun on promoters without strong CREB binding determines high probability of activation upon keratinocyte differentiation.
Collapse
Affiliation(s)
- Julian M. Rozenberg
- Department of Pathology and Lab Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Paramita Bhattacharya
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, West Bengal, India
| | - Raghunath Chatterjee
- Human Genetics Unit, Biological Science Division, Indian Statistical Institute, Kolkata, India
| | - Kimberly Glass
- Harvard School of Public Health, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Charles Vinson
- Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
29
|
Accardi R, Fathallah I, Gruffat H, Mariggiò G, Le Calvez-Kelm F, Voegele C, Bartosch B, Hernandez-Vargas H, McKay J, Sylla BS, Manet E, Tommasino M. Epstein - Barr virus transforming protein LMP-1 alters B cells gene expression by promoting accumulation of the oncoprotein ΔNp73α. PLoS Pathog 2013; 9:e1003186. [PMID: 23516355 PMCID: PMC3597522 DOI: 10.1371/journal.ppat.1003186] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 12/22/2012] [Indexed: 12/15/2022] Open
Abstract
Many studies have proved that oncogenic viruses develop redundant mechanisms to alter the functions of the tumor suppressor p53. Here we show that Epstein-Barr virus (EBV), via the oncoprotein LMP-1, induces the expression of ΔNp73α, a strong antagonist of p53. This phenomenon is mediated by the LMP-1 dependent activation of c-Jun NH2-terminal kinase 1 (JNK-1) which in turn favours the recruitment of p73 to ΔNp73α promoter. A specific chemical inhibitor of JNK-1 or silencing JNK-1 expression strongly down-regulated ΔNp73α mRNA levels in LMP-1-containing cells. Accordingly, LMP-1 mutants deficient to activate JNK-1 did not induce ΔNp73α accumulation. The recruitment of p73 to the ΔNp73α promoter correlated with the displacement of the histone-lysine N-methyltransferase EZH2 which is part of the transcriptional repressive polycomb 2 complex. Inhibition of ΔNp73α expression in lymphoblastoid cells (LCLs) led to the stimulation of apoptosis and up-regulation of a large number of cellular genes as determined by whole transcriptome shotgun sequencing (RNA-seq). In particular, the expression of genes encoding products known to play anti-proliferative/pro-apoptotic functions, as well as genes known to be deregulated in different B cells malignancy, was altered by ΔNp73α down-regulation. Together, these findings reveal a novel EBV mechanism that appears to play an important role in the transformation of primary B cells. Approximately 20% of worldwide human cancers have been associated with viral infections. Many oncogenic viruses exert their transforming properties by inactivating the products of tumour suppressor genes. One of the best characterized events induced by ongocenic viruses is the inactivation of the transcriptional factors p53. The mucosal high-risk HPV types, EBV, HTLV-1 and KSHV, via their viral proteins, are able to target p53 by distinct mechanisms. We have recently described a novel p53 inactivation mechanism of some cutaneous beta HPV types which have been suggested to be associated with skin carcinogenesis. Beta HPV38 induces accumulation of the p53 antagonist, ΔNp73α which in turn silences the expression of the p53-regulated genes. Here we report that also EBV, via the oncoprotein LMP-1, induces the expression of ΔNp73α which is dependent on the recruitment of p73 on ΔNp73 promoter and the activation of JNK-1. The recruitment of p73 to the ΔNp73 promoter correlated with the displacement of the histone-lysine N-methyltransferase EZH2 which is part of a transcriptional repressive polycomb 2 complex. We also show that ΔNp73α plays an important role in transformation of primary human B cells and regulates the expression of a large number of cellular genes that encode proteins linked to cancer development, including lymphomagenesis.
Collapse
MESH Headings
- Apoptosis
- B-Lymphocytes/metabolism
- B-Lymphocytes/virology
- Cell Transformation, Viral/genetics
- Cell Transformation, Viral/physiology
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Down-Regulation
- Epigenesis, Genetic
- Gene Expression Regulation, Viral
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/physiology
- Host-Pathogen Interactions
- Humans
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- Sequence Analysis, RNA
- Transcription, Genetic
- Transcriptional Activation
- Tumor Protein p73
- Tumor Suppressor Protein p53/antagonists & inhibitors
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Up-Regulation
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/metabolism
Collapse
Affiliation(s)
- Rosita Accardi
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Ikbal Fathallah
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Henri Gruffat
- INSERM U758, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Université Claude Bernard Lyon I, Lyon, France
| | - Giuseppe Mariggiò
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | | | - Catherine Voegele
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Birke Bartosch
- CRCL, INSERM U1052, CNRS 5286, Université de Lyon, Lyon, France
| | | | - James McKay
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Bakary S. Sylla
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Evelyne Manet
- INSERM U758, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Université Claude Bernard Lyon I, Lyon, France
| | - Massimo Tommasino
- International Agency for Research on Cancer, World Health Organization, Lyon, France
- * E-mail:
| |
Collapse
|
30
|
Hanks TS, Gauss KA. Pleomorphic adenoma gene-like 2 regulates expression of the p53 family member, p73, and induces cell cycle block and apoptosis in human promonocytic U937 cells. Apoptosis 2012; 17:236-47. [PMID: 22076304 DOI: 10.1007/s10495-011-0672-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The proto-oncogene, pleomorphic adenoma gene-like 2 (PLAGL2), is implicated in a variety of cancers including acute myeloid leukemia (AML), malignant glioma, colon cancer, and lung adenocarcinoma. There is additional evidence that PLAGL2 can function as a tumor suppressor by initiating cell cycle arrest and apoptosis. Interestingly, PLAGL2 has also been implicated in human myelodysplastic syndrome, a disease that is characterized by ineffective hematopoiesis and can lead to fatal cytopenias (low blood counts) as a result of increased apoptosis in the marrow, or, in about one-third of cases, can progress to AML. To gain a better understanding of the actions of PLAGL2 in human myeloid cells, we generated a stable PLAGL2-inducible cell line, using human promonocytic U937 cells. PLAGL2 expression inhibited cell proliferation which correlated with an accumulation of cells in G1, apoptotic DNA-laddering, an increase in caspase 3, 8, and 9 activity, and a loss of mitochondrial transmembrane potential. There was significant increase in the p53 homologue, p73, with PLAGL2 expression, and consistent with mechanisms of p73-regulated cell cycle control and apoptosis, there was increased expression of known p73 target genes p21, DR5, TRAIL, and Bax. PLAGL2-induced cell cycle block was abolished in the presence of p73 siRNA. Together, these data support a role for PLAGL2 in cell cycle regulation and apoptosis via activation of p73.
Collapse
Affiliation(s)
- Tracey S Hanks
- Department of Immunology and Infectious Diseases, Montana State University, 960 Technology Blvd., Bozeman, MT 59718, USA
| | | |
Collapse
|
31
|
p53 Family: Role of Protein Isoforms in Human Cancer. J Nucleic Acids 2011; 2012:687359. [PMID: 22007292 PMCID: PMC3191818 DOI: 10.1155/2012/687359] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 07/04/2011] [Indexed: 01/07/2023] Open
Abstract
TP53, TP63, and TP73 genes comprise the p53 family. Each gene produces protein isoforms through multiple mechanisms including extensive alternative mRNA splicing. Accumulating evidence shows that these isoforms play a critical role in the regulation of many biological processes in normal cells. Their abnormal expression contributes to tumorigenesis and has a profound effect on tumor response to curative therapy. This paper is an overview of isoform diversity in the p53 family and its role in cancer.
Collapse
|
32
|
Pakay JL, Diesch J, Gilan O, Yip YY, Sayan E, Kolch W, Mariadason JM, Hannan RD, Tulchinsky E, Dhillon AS. A 19S proteasomal subunit cooperates with an ERK MAPK-regulated degron to regulate accumulation of Fra-1 in tumour cells. Oncogene 2011; 31:1817-24. [DOI: 10.1038/onc.2011.375] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
33
|
Fra-1 controls motility of bladder cancer cells via transcriptional upregulation of the receptor tyrosine kinase AXL. Oncogene 2011; 31:1493-503. [PMID: 21822309 DOI: 10.1038/onc.2011.336] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fos-related antigen 1 (Fra-1) is a Fos family member overexpressed in several types of human cancers. Here, we report that Fra-1 is highly expressed in the muscle-invasive form of the carcinoma of the bladder (80%) and to a lesser extent in superficial bladder cancer (42%). We demonstrate that in this type of cancer Fra-1 is regulated via a C-terminal instability signal and C-terminal phosphorylation. We show that manipulation of Fra-1 expression levels in bladder cancer cell lines affects cell morphology, motility and proliferation. The gene coding for AXL tyrosine kinase is directly upregulated by Fra-1 in bladder cancer and in other cell lines. Importantly, our data demonstrate that AXL mediates the effect of Fra-1 on tumour cell motility but not on cell proliferation. We suggest that AXL may represent an attractive therapeutic target in cancers expressing high Fra-1 levels.
Collapse
|
34
|
p53 promotes cellular survival in a context-dependent manner by directly inducing the expression of haeme-oxygenase-1. Oncogene 2011; 30:4476-86. [PMID: 21552291 DOI: 10.1038/onc.2011.150] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A variety of cellular insults activate the tumour suppressor p53, leading generally to cell-cycle arrest or apoptosis. However, it is not inconceivable that cellular protective mechanisms may be required to keep cells alive while cell-fate decisions are made. In this respect, p53 has been suggested to perform functions that allow cells to survive, by halting of the cell-cycle, and thus preventing immediate cell death. Nonetheless, the existence of direct pro-survival p53 target genes regulating cellular survival is lacking. We show here evidence for p53-dependent cellular survival in a context-dependent manner. Both mouse and human cells lacking p53 are hypersensitive to hydrogen peroxide (H(2)O(2))-induced cell death compared with their isogenic wild-type counterparts. By contrast, p53(-/-) cells are expectedly resistant to cell death upon exposure to DNA-damaging agents such as cisplatin (CDDP) and etoposide. Although p53 and its classical targets such as p21 and Mdm2 are activated by both H(2)O(2) and CDDP, we found that the expression of haeme-oxygenase-1 (HO-1)-an antioxidant and antiapoptotic protein-was directly induced only upon H(2)O(2) treatment in a p53-dependent manner. Consistently, p53, but not its homologue p73, activated HO-1 expression and was bound to the HO-1 promoter specifically only upon H(2)O(2) treatment. Moreover, silencing HO-1 expression enhanced cell death upon H(2)O(2) treatment only in p53-proficient cells. Finally, H(2)O(2)-mediated cell death was rescued significantly in p53-deficient cells by antioxidant treatment, as well as by bilirubin, a by-product of HO-1 metabolism. Taken together, these data demonstrate a direct role for p53 in promoting cellular survival in a context-specific manner through the activation of a direct transcriptional target, HO-1.
Collapse
|
35
|
Koeppel M, van Heeringen SJ, Kramer D, Smeenk L, Janssen-Megens E, Hartmann M, Stunnenberg HG, Lohrum M. Crosstalk between c-Jun and TAp73alpha/beta contributes to the apoptosis-survival balance. Nucleic Acids Res 2011; 39:6069-85. [PMID: 21459846 PMCID: PMC3152320 DOI: 10.1093/nar/gkr028] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The p53-family member p73 plays a role in various cellular signaling pathways during development and growth control and it can have tumor suppressor properties. Several isoforms of p73 exist with considerable differences in their function. Whereas the functions of the N-terminal isoforms (TA and ΔNp73) and their opposing pro- and antiapoptotic roles have become evident, the functional differences of the distinct C-terminal splice forms of TAp73 have remained unclear. Here, we characterized the global genomic binding sites for TAp73α and TAp73β by chromatin immunoprecipitation sequencing as well as the transcriptional responses by performing RNA sequencing. We identified a specific p73 consensus binding motif and found a strong enrichment of AP1 motifs in close proximity to binding sites for TAp73α. These AP1 motif-containing target genes are selectively upregulated by TAp73α, while their mRNA expression is repressed upon TAp73β induction. We show that their expression is dependent on endogenous c-Jun and that recruitment of c-Jun to the respective AP1 sites was impaired upon TAp73β expression, in part due to downregulation of c-Jun. Several of these AP1-site containing TAp73α-induced genes impinge on apoptosis induction, suggesting an underlying molecular mechanism for the observed functional differences between TAp73α and TAp73β.
Collapse
Affiliation(s)
- Max Koeppel
- Department of Molecular Biology, Faculty of Science, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Tebbi A, Guittet O, Cottet MH, Vesin MF, Lepoivre M. TAp73 induction by nitric oxide: regulation by checkpoint kinase 1 (CHK1) and protection against apoptosis. J Biol Chem 2011; 286:7873-7884. [PMID: 21212274 DOI: 10.1074/jbc.m110.184879] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nitric oxide (NO) is a potent activator of the p53 tumor suppressor protein, thereby inducing cell cycle arrest and apoptosis. However, little is known about the regulation of the two other p53-family members, p63 and p73, by nitrogen oxides. We report here an up-regulation of p73 by NO in p53-null K-562 leukemia cells. Chemical NO prodrugs or macrophage iNOS activity induced an accumulation of the TAp73α isoform in these cells, whereas macrophages from iNOS(-/-) mice did not. NO also up-regulated TAp73 mRNA expression, suggesting a transcriptional regulation. The checkpoint kinases Chk1 and Chk2 can regulate TAp73 induction after DNA damage. We show that these kinases were rapidly phosphorylated upon NO treatment. Genetic silencing or pharmacological inhibition of Chk1 impaired NO-mediated accumulation of TAp73α. Because NO is known to block DNA synthesis through ribonucleotide reductase inhibition, the up-regulation of TAp73α might be caused by DNA damage induced by an arrest of DNA replication forks. In support of this hypothesis, DNA replication inhibitors such as hydroxyurea and aphidicolin similarly enhanced TAp73α expression and Chk1 phosphorylation. Moreover, inhibition of Chk1 also prevented TAp73α accumulation in response to replication inhibitors. The knockdown of TAp73 with siRNA sensitized K-562 cells to apoptosis induced by a nitrosative (NO) or oxidative (H(2)O(2)) injury. Therefore, TAp73α has an unusual cytoprotective role in K-562 cells, contrasting with its pro-apoptotic functions in many other cell models. In conclusion, NO up-regulates several p53 family members displaying pro- and anti-apoptotic effects, suggesting a complex network of interactions and cross-regulations between NO production and p53-related proteins.
Collapse
Affiliation(s)
- Ali Tebbi
- From the Université Paris-Sud, Institute of Molecular and Cellular Biochemistry and Biophysics, UMR 8619, F-91405 Orsay, France,; CNRS, F-91405 Orsay, France, and; the Division of Endocrinology, Diabetes, and Metabolism, Department of Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah 84112
| | - Olivier Guittet
- From the Université Paris-Sud, Institute of Molecular and Cellular Biochemistry and Biophysics, UMR 8619, F-91405 Orsay, France,; CNRS, F-91405 Orsay, France, and
| | - Marie-Hélène Cottet
- From the Université Paris-Sud, Institute of Molecular and Cellular Biochemistry and Biophysics, UMR 8619, F-91405 Orsay, France,; CNRS, F-91405 Orsay, France, and
| | - Marie-Françoise Vesin
- From the Université Paris-Sud, Institute of Molecular and Cellular Biochemistry and Biophysics, UMR 8619, F-91405 Orsay, France,; CNRS, F-91405 Orsay, France, and
| | - Michel Lepoivre
- From the Université Paris-Sud, Institute of Molecular and Cellular Biochemistry and Biophysics, UMR 8619, F-91405 Orsay, France,; CNRS, F-91405 Orsay, France, and.
| |
Collapse
|
37
|
Seo JH, Cha JH, Park JH, Jeong CH, Park ZY, Lee HS, Oh SH, Kang JH, Suh SW, Kim KH, Ha JY, Han SH, Kim SH, Lee JW, Park JA, Jeong JW, Lee KJ, Oh GT, Lee MN, Kwon SW, Lee SK, Chun KH, Lee SJ, Kim KW. Arrest defective 1 autoacetylation is a critical step in its ability to stimulate cancer cell proliferation. Cancer Res 2010; 70:4422-32. [PMID: 20501853 DOI: 10.1158/0008-5472.can-09-3258] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The N-acetyltransferase arrest defective 1 (ARD1) is an important regulator of cell growth and differentiation that has emerged recently as a critical molecule in cancer progression. However, the regulation of the enzymatic and biological activities of human ARD1 (hARD1) in cancer is presently poorly understood. Here, we report that hARD1 undergoes autoacetylation and that this modification is essential for its functional activation. Using liquid chromatography-tandem mass spectrometry and site-directed mutational analyses, we identified K136 residue as an autoacetylation target site. K136R mutation abolished the ability of hARD1 to promote cancer cell growth in vitro and tumor xenograft growth in vivo. Mechanistic investigations revealed that hARD1 autoacetylation stimulated cyclin D1 expression through activation of the transcription factors beta-catenin and activator protein-1. Our results show that hARD1 autoacetylation is critical for its activation and its ability to stimulate cancer cell proliferation and tumorigenesis.
Collapse
Affiliation(s)
- Ji Hae Seo
- NeuroVascular Coordination Research Center, College of Pharmacy, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Dötsch V, Bernassola F, Coutandin D, Candi E, Melino G. p63 and p73, the ancestors of p53. Cold Spring Harb Perspect Biol 2010; 2:a004887. [PMID: 20484388 DOI: 10.1101/cshperspect.a004887] [Citation(s) in RCA: 254] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
p73 and p63 are two homologs of the tumor suppressive transcription factor p53. Given the high degree of structural similarity shared by the p53 family members, p73 and p63 can bind and activate transcription from the majority of the p53-responsive promoters. Besides overlapping functions shared with p53 (i.e., induction of apoptosis in response to cellular stress), the existence of extensive structural variability within the family determines unique roles for p63 and p73. Their crucial and specific functions in controlling development and differentiation are well exemplified by the p63 and p73 knockout mouse phenotypes. Here, we describe the contribution of p63 and p73 to human pathology with emphasis on their roles in tumorigenesis and development.
Collapse
Affiliation(s)
- V Dötsch
- Institute of Biophysical Chemistry, Goethe University, Frankfurt am Main, Germany
| | | | | | | | | |
Collapse
|
39
|
Lefkimmiatis K, Caratozzolo MF, Merlo P, D'Erchia AM, Navarro B, Levrero M, Sbisa' E, Tullo A. p73 and p63 sustain cellular growth by transcriptional activation of cell cycle progression genes. Cancer Res 2009; 69:8563-71. [PMID: 19861536 DOI: 10.1158/0008-5472.can-09-0259] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite extensive studies on the role of tumor suppressor p53 protein and its homologues, p73 and p63, following their overexpression or cellular stress, very little is known about the regulation of the three proteins in cells during physiologic cell cycle progression. We report a role for p73 and p63 in supporting cellular proliferation through the transcriptional activation of the genes involved in G(1)-S and G(2)-M progression. We found that in MCF-7 cells, p73 and p63, but not p53, are modulated during the cell cycle with a peak in S phase, and their silencing determines a significant suppression of proliferation compared with the control. Chromatin immunoprecipitation analysis shows that in cycling cells, p73 and p63 are bound to the p53-responsive elements (RE) present in the regulatory region of cell cycle progression genes. On the contrary, when the cells are arrested in G(0)-G(1), p73 detaches from the REs and it is replaced by p53, which represses the expression of these genes. When the cells move in S phase, p73 is recruited again and p53 is displaced or is weakly bound to the REs. These data open new possibilities for understanding the involvement of p73 and p63 in cancer. The elevated concentrations of p73 and p63 found in many cancers could cause the aberrant activation of cell growth progression genes and therefore contribute to cancer initiation or progression under certain conditions.
Collapse
|
40
|
Cazanave SC, Mott JL, Elmi NA, Bronk SF, Werneburg NW, Akazawa Y, Kahraman A, Garrison SP, Zambetti GP, Charlton MR, Gores GJ. JNK1-dependent PUMA expression contributes to hepatocyte lipoapoptosis. J Biol Chem 2009; 284:26591-602. [PMID: 19638343 DOI: 10.1074/jbc.m109.022491] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Free fatty acids (FFA) induce hepatocyte lipoapoptosis by a c-Jun N-terminal kinase (JNK)-dependent mechanism. However, the cellular processes by which JNK engages the core apoptotic machinery during lipotoxicity, especially activation of BH3-only proteins, remain incompletely understood. Thus, our aim was to determine whether JNK mediates induction of BH3-only proteins during hepatocyte lipoapoptosis. The saturated FFA palmitate, but not the monounsaturated FFA oleate, induces an increase in PUMA mRNA and protein levels. Palmitate induction of PUMA was JNK1-dependent in primary murine hepatocytes. Palmitate-mediated PUMA expression was inhibited by a dominant negative c-Jun, and direct binding of a phosphorylated c-Jun containing the activator protein 1 complex to the PUMA promoter was identified by electrophoretic mobility shift assay and a chromatin immunoprecipitation assay. Short hairpin RNA-targeted knockdown of PUMA attenuated Bax activation, caspase 3/7 activity, and cell death. Similarly, the genetic deficiency of Puma rendered murine hepatocytes resistant to lipoapoptosis. PUMA expression was also increased in liver biopsy specimens from patients with non-alcoholic steatohepatitis as compared with patients with simple steatosis or controls. Collectively, the data implicate JNK1-dependent PUMA expression as a mechanism contributing to hepatocyte lipoapoptosis.
Collapse
Affiliation(s)
- Sophie C Cazanave
- Miles and Shirley Fitterman Center for Digestive Diseases, Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Epigenetics is the study of differences in phenotype, in the absence of variation in the genetic code. Epigenetics is relevant in the pathogenesis of many skin diseases. In the case of the common skin cancers, aberrant methylation of tumor suppressor gene promoters is associated with their transcriptional inactivation. Environmental carcinogens such as ultraviolet radiation and arsenic may act through epigenetic mechanisms. Hypomethylation is associated with activation of systemic autoimmune diseases, such as systemic lupus erythematosus, subacute cutaneous lupus erythematosus and scleroderma. This may be through a mechanism of immunological cross-reactivity with hypomethylated DNA from pathogenic bacteria. Epigenetic factors may also be relevant in the pathogenesis of psoriasis and other inflammatory skin diseases, as well as in the pathogenesis of the disorders of genomic imprinting with cutaneous features.
Collapse
Affiliation(s)
- George W M Millington
- Dermatology Department, Norfolk & Norwich University Hospitals, NHS Foundation Trust, Colney Lane, Norwich, NR4 7UZ, UK.
| |
Collapse
|
42
|
Griesmann H, Schlereth K, Krause M, Samans B, Stiewe T. p53 and p73 in suppression of Myc-driven lymphomagenesis. Int J Cancer 2009; 124:502-6. [PMID: 18942718 DOI: 10.1002/ijc.23978] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Induction of apoptosis by the tumor suppressor p53 is known to protect from Myc-driven lymphomagenesis. The p53 family member p73 is also a proapoptotic protein, which is activated in response to oncogenes like Myc. Here, we have investigated whether p73 provides a similar protection from Myc-driven lymphomas as p53. Confirming previous studies, the inactivation of a single p53 allele (p53+/-) strongly reduced the median survival of Emu-Myc transgenic mice from 103 to 39 days and was invariably associated with a loss of the wild-type p53 allele. In contrast, mutational inactivation of a p73 allele (p73+/-) reduced the median survival by only 12 days. Lymphomas that developed in the p73+/- background showed no loss of heterozygosity (LOH). Furthermore, gene expression profiling of p73+/+, p73+/- and p73-/- lymphomas indicated that p73+/- lymphomas retained p73 transcriptional activity. Subtle gene expression differences between p73+/+ and p73+/- lymphomas, however, suggest a haploinsufficient phenotype on some p73 target genes. This might help to explain why p73+/- animals succumbed to disease slightly earlier than their p73+/+ littermates (log-rank test p<0.0395) and why p73 often shows monoallelic inactivation in human lymphomas. Together these data demonstrate that in Myc-driven lymphomagenesis p73 has weak tumor suppressor activity compared with p53.
Collapse
Affiliation(s)
- Heidi Griesmann
- Department for Hematology, Oncology and Immunology, Philipps-University Marburg, Marburg, Germany
| | | | | | | | | |
Collapse
|
43
|
Soo KC. Role of comprehensive cancer centres during economic and disease transition: National Cancer Centre, Singapore--a case study. Lancet Oncol 2008; 9:796-802. [PMID: 18672215 DOI: 10.1016/s1470-2045(08)70199-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In 1999, Singapore created a comprehensive cancer centre because of the rising occurrence of cancer in the country. Although Singapore is different from many developing countries because of its small geographical size and its well-endowed economy, it has issues that are common to any country or community wanting to start such centres. We present our experience of developing a comprehensive cancer centre. We located the cancer centre strategically adjacent to Singapore's largest hospital. Clinics were designed to provide multidisciplinary care, and site-specific radiation, medical, and surgical oncologists could consult with patients on the same day and at the multidisciplinary outpatient clinics. We developed a large research team and ensured that tumour specimens were carefully obtained and annotated in our tissue repository. Furthermore, we are building up a robust cancer informatics system as well as a pipeline of specialists with local training and overseas stints at other top cancer centres.
Collapse
|
44
|
Danilova N, Sakamoto KM, Lin S. p53 family in development. Mech Dev 2008; 125:919-31. [PMID: 18835440 DOI: 10.1016/j.mod.2008.09.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Revised: 09/04/2008] [Accepted: 09/12/2008] [Indexed: 12/17/2022]
Abstract
The p53 family network is a unique cellular processor that integrates information from various pathways and determines cellular choices between proliferation, replication arrest/repair, differentiation, senescence, or apoptosis. The most studied role of the p53 family is the regulation of stress response and tumor suppression. By removing damaged cells from the proliferating pool, p53 family members preserve the integrity of the genome. In addition to this well recognized role, recent data implicate the p53 protein family in a broader role of controlling cell proliferation, differentiation and death. Members of the p53 protein family with opposing activity perform coordination of these processes. Imbalance of p53 protein family may contribute to a significant proportion of congenital developmental abnormalities in humans.
Collapse
Affiliation(s)
- Nadia Danilova
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, 615 Charles E. Young Drive South, BSRB 454, Los Angeles, CA 90095-1606, USA.
| | | | | |
Collapse
|
45
|
Toh WH, Logette E, Corcos L, Sabapathy K. TAp73beta and DNp73beta activate the expression of the pro-survival caspase-2S. Nucleic Acids Res 2008; 36:4498-509. [PMID: 18611950 PMCID: PMC2490756 DOI: 10.1093/nar/gkn414] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
p73, the p53 homologue, exists as a transactivation-domain-proficient TAp73 or deficient deltaN(DN)p73 form. Expectedly, the oncogenic DNp73 that is capable of inactivating both TAp73 and p53 function, is over-expressed in cancers. However, the role of TAp73, which exhibits tumour-suppressive properties in gain or loss of function models, in human cancers where it is hyper-expressed is unclear. We demonstrate here that both TAp73 and DNp73 are able to specifically transactivate the expression of the anti-apoptotic member of the caspase family, caspase-2(S). Neither p53 nor TAp63 has this property, and only the p73beta form, but not the p73alpha form, has this competency. Caspase-2 promoter analysis revealed that a non-canonical, 18 bp GC-rich Sp-1-binding site-containing region is essential for p73beta-mediated activation. However, mutating the Sp-1-binding site or silencing Sp-1 expression did not affect p73beta's transactivation ability. In vitro DNA binding and in vivo chromatin immunoprecipitation assays indicated that p73beta is capable of directly binding to this region, and consistently, DNA binding p73 mutant was unable to transactivate caspase-2(S). Finally, DNp73beta over-expression in neuroblastoma cells led to resistance to cell death, and concomitantly to elevated levels of caspase-2(S.) Silencing p73 expression in these cells led to reduction of caspase-2(S) expression and increased cell death. Together, the data identifies caspase-2(S) as a novel transcriptional target common to both TAp73 and DNp73, and raises the possibility that TAp73 may be over-expressed in cancers to promote survival.
Collapse
Affiliation(s)
- Wen Hong Toh
- Division of Cellular & Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore 169610, Singapore
| | | | | | | |
Collapse
|
46
|
Puca R, Nardinocchi L, Gal H, Rechavi G, Amariglio N, Domany E, Notterman DA, Scarsella M, Leonetti C, Sacchi A, Blandino G, Givol D, D'Orazi G. Reversible Dysfunction of Wild-Type p53 following Homeodomain-Interacting Protein Kinase-2 Knockdown. Cancer Res 2008; 68:3707-14. [DOI: 10.1158/0008-5472.can-07-6776] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
Matsha T, Donninger H, Erasmus RT, Hendricks D, Stepien A, Parker MI. Expression of p53 and its homolog, p73, in HPV DNA positive oesophageal squamous cell carcinomas. Virology 2007; 369:182-90. [PMID: 17761206 DOI: 10.1016/j.virol.2007.07.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 07/03/2007] [Accepted: 07/13/2007] [Indexed: 11/21/2022]
Abstract
Several studies have detected human papilloma virus (HPV) DNA in squamous cell carcinoma of the oesophagus (OSCC). In this study, we analysed OSCC specimens from 114 patients for the presence of HPV DNA, and p53 and p73 expression. HPV DNA was detected in 44.7% of cases, with the low risk HPV11 occurring most frequently. p53 and p73 expression was detected in 70% and 61.4% of cases, respectively. There was no correlation between expression of p53, p73 or HPV infection and tumour grade, or between p53 expression and the presence of HPV DNA. There was, however, significant correlation between p73 expression and the presence of HPV DNA (p<0.01) and p53 and p73 co-expression (p<0.001), as well as co-expression of p53 and p73 with HPV status (p<0.05). These data support previous studies suggesting a role for HPV infection in OSCC and also indicate that HPV infection and p53 and p73 overexpression are not mutually exclusive. In addition, the data implicate a role for p73 in OSCC and suggest a complex interaction between p53, p73 and HPV in the aetiology of the disease.
Collapse
Affiliation(s)
- T Matsha
- Division of Medical Biochemistry, Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, 7925, South Africa
| | | | | | | | | | | |
Collapse
|
48
|
Wu S, Murai S, Kataoka K, Miyagishi M. Yin Yang 1 induces transcriptional activity of p73 through cooperation with E2F1. Biochem Biophys Res Commun 2007; 365:75-81. [PMID: 17980704 DOI: 10.1016/j.bbrc.2007.10.145] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Accepted: 10/22/2007] [Indexed: 11/26/2022]
Abstract
The transcription factor p73 is a structural homologue of p53 and plays an important role in tumorigenesis, differentiation and development. However, the regulation of p73 pathway has not been wholly understood. Here we reported that YY1-silencing resulted in significant reductions in the activities of the p73 promoters and the endogenous p73 expression level, conversely, overexpression of YY1 could induce the activities of them. Furthermore, we showed that YY1 and E2F1 have synergistic effect on p73 promoter activity. The results of YY1-silencing and E2F1-silencing alone revealed that both factors are involved in the doxorubicin-induced activation of p73 promoter. Immunofluorescence staining and co-immunoprecipitation assays demonstrated that cooperation of YY1 and E2F1 is concomitant with physical interaction in nuclei. The results presented here suggested the cooperative transcriptional regulation of p73 by YY1 and E2F1, and might provide a new regulation mechanism by the YY1 network on tumorigenesis, differentiation and development.
Collapse
Affiliation(s)
- Shourong Wu
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | | | | | | |
Collapse
|
49
|
Danovi SA, Rossi M, Gudmundsdottir K, Yuan M, Melino G, Basu S. Yes-associated protein (YAP) is a critical mediator of c-Jun-dependent apoptosis. Cell Death Differ 2007; 15:217-9. [PMID: 17823615 DOI: 10.1038/sj.cdd.4402226] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|