1
|
Huang M, Li Y, Li Y, Liu S. C-Terminal Binding Protein: Regulator between Viral Infection and Tumorigenesis. Viruses 2024; 16:988. [PMID: 38932279 PMCID: PMC11209466 DOI: 10.3390/v16060988] [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: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
C-terminal binding protein (CtBP), a transcriptional co-repressor, significantly influences cellular signaling, impacting various biological processes including cell proliferation, differentiation, apoptosis, and immune responses. The CtBP family comprises two highly conserved proteins, CtBP1 and CtBP2, which have been shown to play critical roles in both tumorigenesis and the regulation of viral infections. Elevated CtBP expression is noted in various tumor tissues, promoting tumorigenesis, invasiveness, and metastasis through multiple pathways. Additionally, CtBP's role in viral infections varies, exhibiting differing or even opposing effects depending on the virus. This review synthesizes the advances in CtBP's function research in viral infections and virus-associated tumorigenesis, offering new insights into potential antiviral and anticancer strategies.
Collapse
Affiliation(s)
- Meihui Huang
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Yucong Li
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Yuxiao Li
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
| | - Shuiping Liu
- Xiangya School of Medicine, Central South University, Changsha 410013, China; (M.H.); (Y.L.); (Y.L.)
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| |
Collapse
|
2
|
Filograna A, De Tito S, Monte ML, Oliva R, Bruzzese F, Roca MS, Zannetti A, Greco A, Spano D, Ayala I, Liberti A, Petraccone L, Dathan N, Catara G, Schembri L, Colanzi A, Budillon A, Beccari AR, Del Vecchio P, Luini A, Corda D, Valente C. Identification and characterization of a new potent inhibitor targeting CtBP1/BARS in melanoma cells. J Exp Clin Cancer Res 2024; 43:137. [PMID: 38711119 PMCID: PMC11071220 DOI: 10.1186/s13046-024-03044-5] [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: 05/12/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The C-terminal-binding protein 1/brefeldin A ADP-ribosylation substrate (CtBP1/BARS) acts both as an oncogenic transcriptional co-repressor and as a fission inducing protein required for membrane trafficking and Golgi complex partitioning during mitosis, hence for mitotic entry. CtBP1/BARS overexpression, in multiple cancers, has pro-tumorigenic functions regulating gene networks associated with "cancer hallmarks" and malignant behavior including: increased cell survival, proliferation, migration/invasion, epithelial-mesenchymal transition (EMT). Structurally, CtBP1/BARS belongs to the hydroxyacid-dehydrogenase family and possesses a NAD(H)-binding Rossmann fold, which, depending on ligands bound, controls the oligomerization of CtBP1/BARS and, in turn, its cellular functions. Here, we proposed to target the CtBP1/BARS Rossmann fold with small molecules as selective inhibitors of mitotic entry and pro-tumoral transcriptional activities. METHODS Structured-based screening of drug databases at different development stages was applied to discover novel ligands targeting the Rossmann fold. Among these identified ligands, N-(3,4-dichlorophenyl)-4-{[(4-nitrophenyl)carbamoyl]amino}benzenesulfonamide, called Comp.11, was selected for further analysis. Fluorescence spectroscopy, isothermal calorimetry, computational modelling and site-directed mutagenesis were employed to define the binding of Comp.11 to the Rossmann fold. Effects of Comp.11 on the oligomerization state, protein partners binding and pro-tumoral activities were evaluated by size-exclusion chromatography, pull-down, membrane transport and mitotic entry assays, Flow cytometry, quantitative real-time PCR, motility/invasion, and colony assays in A375MM and B16F10 melanoma cell lines. Effects of Comp.11 on tumor growth in vivo were analyzed in mouse tumor model. RESULTS We identify Comp.11 as a new, potent and selective inhibitor of CtBP1/BARS (but not CtBP2). Comp.11 directly binds to the CtBP1/BARS Rossmann fold affecting the oligomerization state of the protein (unlike other known CtBPs inhibitors), which, in turn, hinders interactions with relevant partners, resulting in the inhibition of both CtBP1/BARS cellular functions: i) membrane fission, with block of mitotic entry and cellular secretion; and ii) transcriptional pro-tumoral effects with significantly hampered proliferation, EMT, migration/invasion, and colony-forming capabilities. The combination of these effects impairs melanoma tumor growth in mouse models. CONCLUSIONS: This study identifies a potent and selective inhibitor of CtBP1/BARS active in cellular and melanoma animal models revealing new opportunities to study the role of CtBP1/BARS in tumor biology and to develop novel melanoma treatments.
Collapse
Affiliation(s)
- Angela Filograna
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Stefano De Tito
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, London, UK. The Study Has Been Previously Performed at IEOS-CNR, Naples, Italy
| | - Matteo Lo Monte
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Rosario Oliva
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Francesca Bruzzese
- Animal Facility Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging (IBB), National Research Council (CNR), Naples, 80145, Italy
| | - Adelaide Greco
- Interdepartmental Service Center of Veterinary Radiology, University of Naples Federico II, 80137, Naples, Italy
| | - Daniela Spano
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Inmaculada Ayala
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Assunta Liberti
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Luigi Petraccone
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Nina Dathan
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuliana Catara
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), 80131, Naples, Italy
| | - Laura Schembri
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonino Colanzi
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | | | - Pompea Del Vecchio
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Alberto Luini
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Daniela Corda
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Carmen Valente
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
- Present address: Dompé Farmaceutici S.P.A, L'Aquila, Italy.
| |
Collapse
|
3
|
Fujita T, Fujii H. iChIP-SILAC analysis identifies epigenetic regulators of CpG methylation of the p16 INK4A gene. FEBS Lett 2024; 598:1094-1109. [PMID: 38627195 DOI: 10.1002/1873-3468.14878] [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: 02/14/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 05/12/2024]
Abstract
Allele-specific epigenetic events regulate the expression of specific genes such as tumor suppressor genes. Methods to biochemically identify epigenetic regulators remain limited. Here, we used insertional chromatin immunoprecipitation (iChIP) to address this issue. iChIP combined with quantitative mass spectrometry identified DNA methyltransferase 1 (DNMT1) and epigenetic regulators as proteins that potentially interact with a region of the p16INK4A gene that is CpG-methylated in one allele in HCT116 cells. Some of the identified proteins are involved in the CpG methylation of this region, and of these, DEAD-box helicase 24 (DDX24) contributes to CpG methylation by regulating the protein levels of DNMT1. Thus, iChIP is a useful method to identify proteins which bind to a target locus of interest.
Collapse
Affiliation(s)
- Toshitsugu Fujita
- Department of Biochemistry and Genome Biology, Hirosaki University Graduate School of Medicine, Japan
| | - Hodaka Fujii
- Department of Biochemistry and Genome Biology, Hirosaki University Graduate School of Medicine, Japan
| |
Collapse
|
4
|
Lim YH, Park YJ, Lee J, Kim JH. Transcriptional corepressor activity of CtBP1 is regulated by ISG15 modification. Anim Cells Syst (Seoul) 2024; 28:66-74. [PMID: 38405356 PMCID: PMC10885760 DOI: 10.1080/19768354.2024.2321354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/14/2024] [Indexed: 02/27/2024] Open
Abstract
C-terminal binding protein 1 (CtBP1) is a critical transcriptional corepressor of many tumor suppressor genes and plays diverse roles in the progression of cancers. The transcriptional repression function of CtBP1 is mediated by recruiting histone-modifying enzymes, such as histone deacetylases and histone methyltransferases, to target genes by binding with DNA-interacting factors. Several post-translational modifications of CtBP1 have been identified, including ubiquitination, phosphorylation, and SUMOylation. This paper reports that CtBP1 is conjugated by ISG15. Endogenous CtBP1 was modified by ISG15 after interferon-α treatment in HeLa cells. The ISGylation process of CtBP1 was regulated by deISGylation enzyme USP18 and ISG15 E3 ligase EFP. Interestingly, CtBP1 ISGylation affected the binding affinity between CtBP1 and some components of CtBP1-associated transcriptional complexes. HDAC1 and LSD1 bound more efficiently to ISG15-conjugated CtBP1 than non-conjugated CtBP1. On the other hand, binding between CtBP1 and HDAC4 was unaffected by ISG15 modification. Furthermore, ISG15 modification enhanced the transcriptional repression activity of CtBP1 on several target genes related to EMT and apoptosis. These findings suggest that the ISG15 modification of CtBP1 modulates the function and activity of CtBP1 and that CtBP1 ISGylation may provide a new insight for CtBP1-mediated cancers.
Collapse
Affiliation(s)
- Yun Hwan Lim
- Department of Biological Sciences, Inha University, Incheon, Korea
| | - Yoon Jin Park
- Department of Biological Sciences, Inha University, Incheon, Korea
| | - Jieun Lee
- Department of Biological Sciences, Inha University, Incheon, Korea
| | - Jung Hwa Kim
- Department of Biological Sciences, Inha University, Incheon, Korea
| |
Collapse
|
5
|
The transrepression and transactivation roles of CtBPs in the pathogenesis of different diseases. J Mol Med (Berl) 2021; 99:1335-1347. [PMID: 34196767 DOI: 10.1007/s00109-021-02107-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Gene transcription is strictly controlled by transcriptional complexes, which are assemblies of transcription factors, transcriptional regulators, and co-regulators. Mammalian genomes encode two C-terminal-binding proteins (CtBPs), CtBP1 and CtBP2, which are both well-known transcriptional corepressors of oncogenic processes. Their overexpression in tumors is associated with malignant behavior, such as uncontrolled cell proliferation, migration, and invasion, as well as with an increase in the epithelial-mesenchymal transition. CtBPs coordinate with other transcriptional regulators, such as histone deacetylases (HDACs) and histone acetyltransferases (p300 and CBP [CREBP-binding protein]) that contain the PXDLS motif, and with transcription factors to assemble transcriptional complexes that dock onto the promoters of genes to initiate gene transcription. Emerging evidence suggests that CtBPs function as both corepressors and coactivators in different biological processes ranging from apoptosis to inflammation and osteogenesis. Therapeutic targeting of CtBPs or the interactions required to form transcriptional complexes has also shown promising effects in preventing disease progression. This review summarizes the most recent progress in the study of CtBP functions and therapeutic inhibitors in different biological processes. This knowledge may enable a better understanding of the complexity of the roles of CtBPs, while providing new insights into therapeutic strategies that target CtBPs.
Collapse
|
6
|
Hu K, Li Y, Yu H, Hu Y. CTBP1 Confers Protection for Hippocampal and Cortical Neurons in Rat Models of Alzheimer's Disease. Neuroimmunomodulation 2019; 26:139-152. [PMID: 31340205 DOI: 10.1159/000500942] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/08/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) is an age-related devastating neurodegenerative disorder. The hippocampus and cerebral cortex are the most closely related brain regions of cognitive function and neurogenesis. The present study investigated the role of C-terminal-binding protein 1 (CTBP1) in AD. METHODS AD rat models were established through intracerebroventricular injection of β-amyloid polypeptide Aβ(25-35) and intragastric administration of aluminum chloride solution, and the expression pattern that CTBP1 showed in the hippocampus and cerebral cortex was determined. The learning and memory abilities of AD rats after CTBP1 overexpression were assessed. Hippocampal and cortical neurons were transfected with siRNA against CTBP1 or CTBP1-overexpressing plasmids in order to study the effects of CTBP1 elevation or depletion on neuron morphological changes, apoptosis, and viability. The expression of CTBP1, proapoptotic factor (B-cell lymphoma 2; Bcl-2), and antiapoptotic factors (Bcl-2-associated X protein [Bax] and caspase-3) was subsequently evaluated. RESULTS CTBP1 was poorly expressed in the hippocampus and cerebral cortex. AD rats displayed enhanced learning and memory abilities following CTBP1 overexpression. Furthermore, overexpression of CTBP1 improved morphological changes of hippocampal and cortical neurons, increased neuron activity, and inhibited neuron apoptosis in AD rats. Moreover, the expression of Bax and caspase-3 decreased, yet Bcl-2 increased. CONCLUSION Collectively, CTBP1 plays a protective role in the degeneration of hippocampal and cortical neurons whereby overexpressed CTBP1 attenuated the hippocampal and cortical neuron apoptosis and enhanced neuron activity, highlighting the potential of CTBP1 as a target for AD treatment.
Collapse
Affiliation(s)
- Kai Hu
- Department of Anesthesiology, Nanchang Hongdu Hospital of TCM, Nanchang, China
| | - Yafeng Li
- Department of Anesthesiology, Nanchang Hongdu Hospital of TCM, Nanchang, China
| | - Huifen Yu
- Department of Anesthesiology, Nanchang Hongdu Hospital of TCM, Nanchang, China
| | - Yanhui Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China,
| |
Collapse
|
7
|
de Abreu PM, Có ACG, Azevedo PL, do Valle IB, de Oliveira KG, Gouvea SA, Cordeiro-Silva MF, Louro ID, de Podestá JRV, Lenzi J, Sena A, Mendonça EF, von Zeidler SLV. Frequency of HPV in oral cavity squamous cell carcinoma. BMC Cancer 2018; 18:324. [PMID: 29580212 PMCID: PMC5870524 DOI: 10.1186/s12885-018-4247-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 03/19/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The prevalence of high-risk human papillomavirus (HPV) DNA in cases of oral cavity squamous cell carcinoma (SCC) varies widely. The aim of this study is to investigate the frequency of high-risk HPV DNA in a large Brazilian cohort of patients with oral cavity SCC. METHODS Biopsy and resected frozen and formalin-fixed paraffin-embedded specimens of oral cavity SCC were available from 101 patients who were recruited at two Brazilian centres. Stringent measures with respect to case selection and prevention of sample contamination were adopted to ensure reliability of the data. Nested PCR using MY09/MY11 and GP5+/GP6+ as well as PGMY09/11 L1 consensus primers were performed to investigate the presence of HPV DNA in the tumours. HPV-positive cases were subjected to direct sequencing. Shapiro-Wilk and Student t test were used to evaluate data normality and to compare the means, respectively. Qualitative variables were analysed by logistic regression. RESULTS Our results demonstrate that the frequency of high-risk HPV types in oral cavity SCC is very low and is less than 4%. All HPV-positive cases were HPV16. In addition, our results do not show a significant association between the tumour clinical features and the risk factors (tobacco, alcohol and HPV) for oral cavity SCC. CONCLUSION In the current study, we observed an overlapping pattern of risk factors that are related to tumour development. This, along with a low frequency of high-risk HPV DNA, supports the findings that HPV is not involved in the genesis of oral cavity SCC in Brazilian population.
Collapse
Affiliation(s)
- Priscila Marinho de Abreu
- Programa de Pós-Graduação em Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Vitoria, Espirito Santo, Brazil
| | - Anna Clara Gregório Có
- Departamento de Patologia, Programa de Pós-graduação em Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Av. Marechal Campos, 1468 Maruípe, Vitória, ES, 29.040-090, Brazil
| | - Pedro Leite Azevedo
- Departamento de Patologia, Programa de Pós-graduação em Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Av. Marechal Campos, 1468 Maruípe, Vitória, ES, 29.040-090, Brazil
| | - Isabella Bittencourt do Valle
- Programa de Pós-Graduação em Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Vitoria, Espirito Santo, Brazil
| | - Karine Gadioli de Oliveira
- Departamento de Ciências Fisiológicas, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Vitoria, Espirito Santo, Brazil
| | - Sônia Alves Gouvea
- Departamento de Ciências Fisiológicas, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Vitoria, Espirito Santo, Brazil
| | | | - Iúri Drummond Louro
- Programa de Pós-Graduação em Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Vitoria, Espirito Santo, Brazil
| | - José Roberto Vasconcelos de Podestá
- Programa de Prevenção e Detecção Precoce do Câncer Bucal, Setor de Cirurgia de Cabeça e Pescoço, Hospital Santa Rita de Cássia, Vitória, Espírito Santo, Brazil
| | - Jeferson Lenzi
- Programa de Prevenção e Detecção Precoce do Câncer Bucal, Setor de Cirurgia de Cabeça e Pescoço, Hospital Santa Rita de Cássia, Vitória, Espírito Santo, Brazil
| | - Agenor Sena
- Programa de Prevenção e Detecção Precoce do Câncer Bucal, Setor de Cirurgia de Cabeça e Pescoço, Hospital Santa Rita de Cássia, Vitória, Espírito Santo, Brazil
| | | | - Sandra Lúcia Ventorin von Zeidler
- Programa de Pós-Graduação em Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Vitoria, Espirito Santo, Brazil. .,Departamento de Patologia, Programa de Pós-graduação em Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Av. Marechal Campos, 1468 Maruípe, Vitória, ES, 29.040-090, Brazil.
| |
Collapse
|
8
|
Blevins MA, Huang M, Zhao R. The Role of CtBP1 in Oncogenic Processes and Its Potential as a Therapeutic Target. Mol Cancer Ther 2018; 16:981-990. [PMID: 28576945 DOI: 10.1158/1535-7163.mct-16-0592] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/11/2016] [Accepted: 02/22/2017] [Indexed: 12/24/2022]
Abstract
Transcriptional corepressor proteins have emerged as an important facet of cancer etiology. These corepressor proteins are often altered by loss- or gain-of-function mutations, leading to transcriptional imbalance. Thus, research directed at expanding our current understanding of transcriptional corepressors could impact the future development of new cancer diagnostics, prognostics, and therapies. In this review, our current understanding of the CtBP corepressors, and their role in both development and disease, is discussed in detail. Importantly, the role of CtBP1 overexpression in adult tissues in promoting the progression of multiple cancer types through their ability to modulate the transcription of developmental genes ectopically is explored. CtBP1 overexpression is known to be protumorigenic and affects the regulation of gene networks associated with "cancer hallmarks" and malignant behavior, including increased cell survival, proliferation, migration, invasion, and the epithelial-mesenchymal transition. As a transcriptional regulator of broad developmental processes capable of promoting malignant growth in adult tissues, therapeutically targeting the CtBP1 corepressor has the potential to be an effective method for the treatment of diverse tumor types. Although efforts to develop CtBP1 inhibitors are still in the early stages, the current progress and the future perspectives of therapeutically targeting this transcriptional corepressor are also discussed. Mol Cancer Ther; 16(6); 981-90. ©2017 AACR.
Collapse
Affiliation(s)
- Melanie A Blevins
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Mingxia Huang
- Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado.
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
9
|
Yang X, Sun Y, Li H, Shao Y, Zhao D, Yu W, Fu J. C-terminal binding protein-2 promotes cell proliferation and migration in breast cancer via suppression of p16INK4A. Oncotarget 2018; 8:26154-26168. [PMID: 28412731 PMCID: PMC5432247 DOI: 10.18632/oncotarget.15402] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/01/2017] [Indexed: 01/27/2023] Open
Abstract
C-terminal binding protein-2 (CtBP2) enhances cancer proliferation and metastasis. The role and mechanism of CtBP2 in breast cancer remains to be elucidated. Western blot and immunochemistry were employed to evaluate the level of CtBP2 and p16INK4A in breast cancer. Genetic manipulation was used to study the expression of p16INK4A and its downstream genes regulated by CtBP2. Functional assays, including colony formation, wound healing, transwell invasion, anchorage-independent growth assay and a xenograft tumor model were used to determine the oncogenic role of CtBP2 in breast cancer progression. The expression of CtBP2 was increased in breast cancer tissues and cell lines. The expression of p16INK4A were inversely correlated CtBP2 (r2 = 0.43, P < 0.01). The expression of both CtBP2 and p16INK4A were significantly related to histological differentiation (P < 0.01 and P = 0.004, respectively) and metastasis (P = 0.046 and 0.047, respectively). The overall survival rate was lower in patients with increased CtBP2 expression and lower p16INK4A expression. Knockdown of CtBP2 resulted in the activation of p16INK4A and down–regulation of cell cycle regulators cyclin D, cyclin E and cyclin-dependent kinase 2 and 4. This down-regulation also led to a decreased transition of the G1-S phase in breast cancer cells. Moreover, gain-of-function experiments showed that CtBP2 suppressed p16INK4A and matrix metalloproteinase-2, subsequently enhancing the migration in breast cancer. However, the silence of CtBP2 abrogated this effect. Collectively, these findings provide insight into the role CtBP2 plays in promoting proliferation and migration in breast cancer by the inhibition of p16INK4A.
Collapse
Affiliation(s)
- Xiaojing Yang
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yi Sun
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Hongling Li
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yuhui Shao
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Depeng Zhao
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, P.R. China
| | - Weiwei Yu
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jie Fu
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| |
Collapse
|
10
|
Shen Z, Asa SL, Ezzat S. Ikaros and its interacting partner CtBP target the metalloprotease ADAMTS10 to modulate pituitary cell function. Mol Cell Endocrinol 2017; 439:126-132. [PMID: 27815209 DOI: 10.1016/j.mce.2016.10.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/28/2016] [Accepted: 10/29/2016] [Indexed: 12/23/2022]
Abstract
We have previously described the expression and up-regulation of the C-terminal Binding Protein (CtBP) in response to pituitary hypoxia. This co-repressor interacts with the hematopoietic factor Ikaros to target several components implicated in cellular growth and apoptotic pathways. To identify common transcriptional pituitary targets we performed promoter arrays using Ikaros and CtBP chromatin immunoprecipitated (ChIP) DNA from pituitary AtT20 cells. This approach yielded a finite list of gene targets common to both transcription factors. Of these, the metalloprotease ADAMTS10 emerged as a validated target. We show the ability of Ikaros to bind the ADAMTS10 promoter, influence its transfected activity, and induce endogenous gene expression. ADAMTS10 is expressed in primary pituitary cells and is down-regulated in Ikaros null mice. Further, knockdown of ADAMTS10 in AtT20 cells recapitulates the impact of Ikaros deficiency on POMC/ACTH hormone expression. These results uncover a novel role for the metalloprotease ADAMTS10 in the pituitary. Additionally, they position this metalloprotease as a potential functional integrator of the Ikaros-CtBP chromatin remodeling network.
Collapse
Affiliation(s)
- Zhongyi Shen
- Dept. of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario M5G 2M9, Canada; University Health Network and the Ontario Cancer Institute, Toronto, Ontario M5G 2M9, Canada
| | - Sylvia L Asa
- Dept. of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario M5G 2M9, Canada; University Health Network and the Ontario Cancer Institute, Toronto, Ontario M5G 2M9, Canada
| | - Shereen Ezzat
- Dept. of Medicine, University of Toronto, Toronto, Ontario M5G 2M9, Canada; University Health Network and the Ontario Cancer Institute, Toronto, Ontario M5G 2M9, Canada.
| |
Collapse
|
11
|
Silencing of CtBP1 suppresses the migration in human glioma cells. J Mol Histol 2016; 47:297-304. [PMID: 27160109 DOI: 10.1007/s10735-016-9678-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 04/28/2016] [Indexed: 12/20/2022]
Abstract
Carboxyl-terminal binding protein 1 (CtBP1), up-regulated in various types of human cancers, has been functionally associated with proliferation, anti-apoptosis, and EMT in vitro studies. However, the functional significance of CtBP1 in the pathophysiology of glioma remains unknown. In the present study, we showed the expression of CtBP1 was markedly higher in glioma tissues compared with normal brain tissues by Western blot analysis. Immunohistochemical analysis revealed that CtBP1 mainly localized in the nucleus of glioma cells. Statistical analysis suggested the upregulation of CtBP1 was considerably correlated with the WHO grade (P < 0.05) and those patients with high CtBP1 levels exhibited shorter survival time (P < 0.01). Silencing CtBP1 by short hairpin RNAi caused an inhibition of cell migration. Moreover, knockdown of CtBP1 increases E-cadherin expression and decreases vimentin expression. These data uncovered that CtBP1 protein is a valuable marker of glioma pathogenic process and that CtBP1 can serve as a novel prognostic marker for glioma therapy.
Collapse
|
12
|
Bishop JA, Ha PK. Human papillomavirus detection in a "Digital" age. Cancer 2016; 122:1502-4. [PMID: 26990085 DOI: 10.1002/cncr.29979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 02/22/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Justin A Bishop
- Johns Hopkins Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Patrick K Ha
- Division of Head and Neck Surgical Oncology, Department of Otolaryngology-Head and Neck Surgery, University of California-San Francisco, San Francisco, California
| |
Collapse
|
13
|
Gutiérrez J, García-Villa E, Ocadiz-Delgado R, Cortés-Malagón EM, Vázquez J, Roman-Rosales A, Alvarez-Rios E, Celik H, Romano MC, Üren A, Lambert PF, Gariglio P. Human papillomavirus type 16 E7 oncoprotein upregulates the retinoic acid receptor-beta expression in cervical cancer cell lines and K14E7 transgenic mice. Mol Cell Biochem 2015; 408:261-272. [PMID: 26173416 DOI: 10.1007/s11010-015-2504-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 07/04/2015] [Indexed: 12/22/2022]
Abstract
Persistent infection with high-risk human papillomaviruses is the main etiological factor in cervical cancer (CC). The human papillomavirus type 16 (HPV16) E7 oncoprotein alters several cellular processes, regulating the expression of many genes in order to avoid cell cycle control. Retinoic acid receptor beta (RARB) blocks cell growth, inducing differentiation and apoptosis. This tumor suppressor gene is gradually silenced in late passages of foreskin keratinocytes immortalized with HPV16 and in various tumors, including CC, mainly by epigenetic modifications. We investigated the effect of E7 oncoprotein on RARB gene expression. We found that HPV16 E7 increases RARB mRNA and RAR-beta protein expression both in vitro and in the cervix of young K14E7 transgenic mice. In E7-expressing cells, RARB overexpression is further increased in the presence of the tumor suppressor p53 (TP53) R273C mutant. This effect does not change when either C33-A or E7-expressing C33-A cell line is treated with Trichostatin A, suggesting that E7 enhances RARB expression independently of histone deacetylases inhibition. These findings indicate that RARB overexpression is part of the early molecular events induced by the E7 oncoprotein.
Collapse
Affiliation(s)
- Jorge Gutiérrez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Enrique García-Villa
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Rodolfo Ocadiz-Delgado
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Enoc M Cortés-Malagón
- Laboratorio de Biología Molecular del Cáncer, Unidad de Investigación, Hospital Juárez de México, Av. IPN 5160, Magdalena de Las Salinas, Gustavo A. Madero, 07760, Ciudad de México, México
| | - Juan Vázquez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Alejandra Roman-Rosales
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Elizabeth Alvarez-Rios
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Haydar Celik
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057-1469, USA
| | - Marta C Romano
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Aykut Üren
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057-1469, USA
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Patricio Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México.
| |
Collapse
|
14
|
Stankiewicz TR, Gray JJ, Winter AN, Linseman DA. C-terminal binding proteins: central players in development and disease. Biomol Concepts 2015; 5:489-511. [PMID: 25429601 DOI: 10.1515/bmc-2014-0027] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 10/07/2014] [Indexed: 01/06/2023] Open
Abstract
C-terminal binding proteins (CtBPs) were initially identified as binding partners for the E1A-transforming proteins. Although the invertebrate genome encodes one CtBP protein, two CtBPs (CtBP1 and CtBP2) are encoded by the vertebrate genome and perform both unique and duplicative functions. CtBP1 and CtBP2 are closely related and act as transcriptional corepressors when activated by nicotinamide adenine dinucleotide binding to their dehydrogenase domains. CtBPs exert transcriptional repression primarily via recruitment of a corepressor complex to DNA that consists of histone deacetylases (HDACs) and histone methyltransferases, although CtBPs can also repress transcription through HDAC-independent mechanisms. More recent studies have demonstrated a critical function for CtBPs in the transcriptional repression of pro-apoptotic genes such as Bax, Puma, Bik, and Noxa. Nonetheless, although recent efforts have characterized the essential involvement of CtBPs in promoting cellular survival, the dysregulation of CtBPs in both neurodegenerative disease and cancers remains to be fully elucidated.
Collapse
|
15
|
Abstract
Oral cavity squamous cell carcinoma (OC-SCC) is the most common malignancy of the head and neck (excluding nonmelanoma skin cancer). Recent trends have shown a dramatic rise in the incidence of oropharyngeal squamous cell carcinoma (OP-SCC), with a marked increase in lesions related to human papillomavirus infection. This update presents the latest evidence regarding OC-SCC and OP-SCC. In particular, the authors compare and contrast tumors at these two sites with respect to epidemiology, etiopathogenesis, clinicopathologic presentation, clinical assessment, imaging, management, and prognosis. It is important for clinicians to be aware of differences between OC-SCC and OP-SCC so that appropriate patient education and multidisciplinary care can be provided to optimize outcomes.
Collapse
Affiliation(s)
- Angela C Chi
- Professor, Division of Oral Pathology, Medical University of South Carolina, Charleston, SC
| | - Terry A Day
- Professor, Wendy and Keith Wellin Endowed Chair for Head and Neck Oncology, Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC
| | - Brad W Neville
- Distinguished University Professor, Division of Oral Pathology, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
16
|
Bishop JA, Lewis JS, Rocco JW, Faquin WC. HPV-related squamous cell carcinoma of the head and neck: An update on testing in routine pathology practice. Semin Diagn Pathol 2015; 32:344-51. [DOI: 10.1053/j.semdp.2015.02.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
17
|
Kagawa S, Natsuizaka M, Whelan KA, Facompre N, Naganuma S, Ohashi S, Kinugasa H, Egloff AM, Basu D, Gimotty PA, Klein-Szanto AJ, Bass A, Wong KK, Diehl JA, Rustgi AK, Nakagawa H. Cellular senescence checkpoint function determines differential Notch1-dependent oncogenic and tumor-suppressor activities. Oncogene 2015; 34:2347-59. [PMID: 24931169 PMCID: PMC4268095 DOI: 10.1038/onc.2014.169] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 03/27/2014] [Accepted: 04/14/2014] [Indexed: 12/12/2022]
Abstract
Notch activity regulates tumor biology in a context-dependent and complex manner. Notch may act as an oncogene or a tumor-suppressor gene even within the same tumor type. Recently, Notch signaling has been implicated in cellular senescence. Yet, it remains unclear as to how cellular senescence checkpoint functions may interact with Notch-mediated oncogenic and tumor-suppressor activities. Herein, we used genetically engineered human esophageal keratinocytes and esophageal squamous cell carcinoma cells to delineate the functional consequences of Notch activation and inhibition along with pharmacological intervention and RNA interference experiments. When expressed in a tetracycline-inducible manner, the ectopically expressed activated form of Notch1 (ICN1) displayed oncogene-like characteristics inducing cellular senescence corroborated by the induction of G0/G1 cell-cycle arrest, Rb dephosphorylation, flat and enlarged cell morphology and senescence-associated β-galactosidase activity. Notch-induced senescence involves canonical CSL/RBPJ-dependent transcriptional activity and the p16(INK4A)-Rb pathway. Loss of p16(INK4A) or the presence of human papilloma virus (HPV) E6/E7 oncogene products not only prevented ICN1 from inducing senescence but permitted ICN1 to facilitate anchorage-independent colony formation and xenograft tumor growth with increased cell proliferation and reduced squamous-cell differentiation. Moreover, Notch1 appears to mediate replicative senescence as well as transforming growth factor-β-induced cellular senescence in non-transformed cells and that HPV E6/E7 targets Notch1 for inactivation to prevent senescence, revealing a tumor-suppressor attribute of endogenous Notch1. In aggregate, cellular senescence checkpoint functions may influence dichotomous Notch activities in the neoplastic context.
Collapse
Affiliation(s)
- Shingo Kagawa
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mitsuteru Natsuizaka
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Gastroenterology and Hepatology, Hokkaido University, Sapporo, Japan
| | - Kelly A. Whelan
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nicole Facompre
- Departments of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
- Wistar Institute, Philadelphia, Pennsylvania
| | - Seiji Naganuma
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology, Kochi University Medical School, Kochi, Japan
| | - Shinya Ohashi
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideaki Kinugasa
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Ann Marie Egloff
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Devraj Basu
- Departments of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
- Wistar Institute, Philadelphia, Pennsylvania
| | - Phyllis A. Gimotty
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Biostatistics, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Adam Bass
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of Cellular and Molecular Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Kwok-Kin Wong
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of Cellular and Molecular Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - J. Alan Diehl
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anil K. Rustgi
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hiroshi Nakagawa
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
18
|
Guan C, Shi H, Wang H, Zhang J, Ni W, Chen B, Hou S, Yang X, Shen A, Ni R. CtBP2 contributes to malignant development of human esophageal squamous cell carcinoma by regulation of p16INK4A. J Cell Biochem 2014; 114:1343-54. [PMID: 23255392 DOI: 10.1002/jcb.24475] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 11/28/2012] [Indexed: 12/26/2022]
Abstract
C-terminal binding protein-2 (CtBP2), as a transcriptional co-repressor, has been shown to mediate the repression of p16(INK4A) , a tumor suppressor gene product, in primary human cells. Here we aimed to investigate how the correlation between CtBP2 and p16(INK4A) influenced the development of esophageal squamous cell carcinoma (ESCC). Immunohistochemistry of ESCC tissue sections indicated that the CtBP2 and p16(INK4A) expressions were inversely correlated to each other with a linear regression coefficient of -0.747 (P < 0.05), and Western blot analysis revealed that CtBP2 was higher expressed in tumorous tissues than in adjacent non-tumorous tissues. Either CtBP2 or p16(INK4A) expression was significantly related to histological differentiation (P = 0.016 or 0.001) and to the expression of Ki-67, a proliferating marker (P = 0.006 or 0.02), and patients with higher CtBP2 and lower p16(INK4A) expressions had shorter overall survival. We also observed that CtBP2 modulated the cell proliferation and cell cycle in ECA109 cells, an ESCC cell line, by inhibiting p16(INK4A) . Overexpression or knockdown of CtBP2 in ECA109 cells was found to inhibit or activate the mRNA or protein expression of p16(INK4A) , which in turn altered the cell proliferation and cell cycle in ECA109 cells, as measured by flow cytometry and cell count assay. Additionally, after ECA109 cells silenced for CtBP2 were treated with cisplatin (an anti-ESCC agent), the p16(INK4A) expression was up-regulated, and the cell apoptosis was promoted, thus confirming the repression of p16(INK4A) by CtBP2. Collectively, all results suggested that CtBP2 might contribute to the progression of ESCC through a negative transcriptional regulation of p16(INK4A).
Collapse
Affiliation(s)
- Chengqi Guan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Moiola CP, De Luca P, Zalazar F, Cotignola J, Rodríguez-Seguí SA, Gardner K, Meiss R, Vallecorsa P, Pignataro O, Mazza O, Vazquez ES, De Siervi A. Prostate tumor growth is impaired by CtBP1 depletion in high-fat diet-fed mice. Clin Cancer Res 2014; 20:4086-95. [PMID: 24842953 DOI: 10.1158/1078-0432.ccr-14-0322] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Clinical and epidemiologic data suggest that obesity is associated with more aggressive forms of prostate cancer, poor prognosis, and increased mortality. C-terminal-binding protein 1 (CtBP1) is a transcription repressor of tumor suppressor genes and is activated by NADH binding. High calorie intake decreases intracellular NAD(+)/NADH ratio. The aim of this work was to assess the effect of high-fat diet (HFD) and CtBP1 expression modulation over prostate xenograft growth. EXPERIMENTAL DESIGN We developed a metabolic syndrome-like disease in vivo model by feeding male nude mice with HFD during 16 weeks. Control diet (CD)-fed animals were maintained at the same conditions. Mice were inoculated with PC3 cells stable transfected with shCtBP1 or control plasmids. Genome-wide expression profiles and Gene Set Enrichment Analysis (GSEA) were performed from PC3.shCtBP1 versus PC3.pGIPZ HFD-fed mice tumors. RESULTS No significant differences were observed in tumor growth on CD-fed mice; however, we found that only 60% of HFD-fed mice inoculated with CtBP1-depleted cells developed a tumor. Moreover these tumors were significantly smaller than those generated by PC3.pGIPZ control xenografts. We found 823 genes differentially expressed in shCtBP1 tumors from HFD-fed mice. GSEA from expression dataset showed that most of these genes correspond to cell adhesion, metabolic process, and cell cycle. CONCLUSIONS Metabolic syndrome-like diseases and CtBP1 expression cooperate to induce prostate tumor growth. Hence, targeting of CtBP1 expression might be considered for prostate cancer management and therapy in the subset of patients with metabolic syndromes.
Collapse
Affiliation(s)
- Cristian P Moiola
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, IBYME-CONICET; Laboratorio de Inflamación y Cáncer, Departamento de Química Biologica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN - CONICET
| | - Paola De Luca
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, IBYME-CONICET; Laboratorio de Inflamación y Cáncer, Departamento de Química Biologica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN - CONICET
| | - Florencia Zalazar
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, IBYME-CONICET; Laboratorio de Inflamación y Cáncer, Departamento de Química Biologica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN - CONICET
| | - Javier Cotignola
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biologica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN - CONICET
| | - Santiago A Rodríguez-Seguí
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET
| | - Kevin Gardner
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Roberto Meiss
- Departamento de Patología, Instituto de Estudios Oncológicos, Academia Nacional de Medicina
| | - Pablo Vallecorsa
- Departamento de Patología, Instituto de Estudios Oncológicos, Academia Nacional de Medicina
| | - Omar Pignataro
- Laboratorio de Endocrinología Molecular y Transducción de Señales, IBYME-CONICET, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN - CONICET
| | - Osvaldo Mazza
- Hospital de Clínicas "José de San Martín", Buenos Aires, Argentina; and
| | - Elba S Vazquez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biologica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN - CONICET
| | - Adriana De Siervi
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, IBYME-CONICET; Laboratorio de Inflamación y Cáncer, Departamento de Química Biologica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN - CONICET;
| |
Collapse
|
20
|
Deng H, Li F, Li H, Deng Y, Liu J, Wang D, Han G, Wang XJ, Zhang Q. CtBP1 overexpression in keratinocytes perturbs skin homeostasis. J Invest Dermatol 2013; 134:1323-1331. [PMID: 24280726 PMCID: PMC4537778 DOI: 10.1038/jid.2013.504] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 09/24/2013] [Accepted: 10/08/2013] [Indexed: 12/20/2022]
Abstract
Carboxyl-terminal binding protein-1 (CtBP1) is a transcriptional co-repressor with multiple in vitro targets, but its in vivo functions are largely unknown. We generated keratinocyte-specific CtBP1 transgenic mice with a keratin 5 promoter (K5.CtBP1) to probe the pathological roles of CtBP1. At transgene expression levels comparable with endogenous CtBP1 in acute skin wounds, K5.CtBP1 epidermis displayed hyperproliferation, loss of E-cadherin, and failed terminal differentiation. Known CtBP1 target genes associated with these processes, e.g., p21, Brca1, and E-cadherin were down-regulated in K5.CtBP1 skin. Surprisingly, K5.CtBP1 pups also exhibited a hair loss phenotype. We found that expression of the Distal-less 3 (Dlx3), a critical regulator of hair follicle differentiation and cycling, was decreased in K5.CtBP1 mice. Molecular studies revealed that CtBP1 directly suppressed Dlx3 transcription. Consistently, K5.CtBP1 mice displayed abnormal hair follicles with decreased expression of Dlx3 downstream targets Gata3, Hoxc13, and hair keratins. In sum, this first CtBP1 transgenic model provides in vivo evidence for certain CtBP1 functions predicted from in vitro studies, reveals to our knowledge previously unreported functions and transcriptional activities of CtBP1 in the context of epithelial-mesenchymal interplay, and suggest CtBP1 has a pathogenesis role in hair follicle morphogenesis and differentiation.
Collapse
Affiliation(s)
- Hui Deng
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA; Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA; Department of Dermatology, The Sixth People's Hospital of Shanghai, Shanghai Jiaotong University, Shanghai, China
| | - Fulun Li
- Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA; Department of Dermatology, Yueyang Hospital Affiliated to Shanghai University of TCM, Shanghai, China
| | - Hong Li
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA
| | - Yu Deng
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA
| | - Jing Liu
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA
| | - Donna Wang
- Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA.
| | - Gangwen Han
- Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA
| | - Xiao-Jing Wang
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA; Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA.
| | - Qinghong Zhang
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA; Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA; Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
21
|
Bonilla-Velez J, Mroz EA, Hammon RJ, Rocco JW. Impact of human papillomavirus on oropharyngeal cancer biology and response to therapy: implications for treatment. Otolaryngol Clin North Am 2013; 46:521-43. [PMID: 23910468 PMCID: PMC3740406 DOI: 10.1016/j.otc.2013.04.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Oropharyngeal squamous cell carcinoma (OPSCC) originating from human papillomavirus infection has emerged as a new entity in head and neck cancer, defining a subset of patients with distinct carcinogenesis, risk factor profiles, and clinical presentation that show markedly improved survival than patients with classic OPSCC. De-escalation of therapy and identification of relevant biomarkers to aid in patient selection are actively being investigated. This review addresses the implications of these findings in clinical care.
Collapse
Affiliation(s)
- Juliana Bonilla-Velez
- Postdoctoral Research Fellow, Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston MA
| | - Edmund A. Mroz
- Research Scientist, Center for Cancer Research and Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Rebecca J. Hammon
- Clinical Research Fellow, Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston MA
| | - James W. Rocco
- Associate Professor of Otology and Laryngology and Director, Head and Neck Cancer Research, Massachusetts Eye and Ear Infirmary and Harvard Medical School, Boston MA
- Associate Professor of Otology and Laryngology, Center for Cancer Research and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston MA
| |
Collapse
|
22
|
The corepressor CTBP2 is a coactivator of retinoic acid receptor/retinoid X receptor in retinoic acid signaling. Mol Cell Biol 2013; 33:3343-53. [PMID: 23775127 DOI: 10.1128/mcb.01213-12] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Retinoids play key roles in development, differentiation, and homeostasis through regulation of specific target genes by the retinoic acid receptor/retinoid X receptor (RAR/RXR) nuclear receptor complex. Corepressors and coactivators contribute to its transcriptional control by creating the appropriate chromatin environment, but the precise composition of these nuclear receptor complexes remains to be elucidated. Using an RNA interference-based genetic screen in mouse F9 cells, we identified the transcriptional corepressor CTBP2 (C-terminal binding protein 2) as a coactivator critically required for retinoic acid (RA)-induced transcription. CTBP2 suppression by RNA interference confers resistance to RA-induced differentiation in diverse murine and human cells. Mechanistically, we find that CTBP2 associates with RAR/RXR at RA target gene promoters and is essential for their transactivation in response to RA. We show that CTBP2 is indispensable to create a chromatin environment conducive for RAR/RXR-mediated transcription by recruiting the histone acetyltransferase p300. Our data reveal an unexpected function of the corepressor CTBP2 as a coactivator for RAR/RXR in RA signaling.
Collapse
|
23
|
Deng H, Liu J, Deng Y, Han G, Shellman YG, Robinson SE, Tentler JJ, Robinson WA, Norris DA, Wang XJ, Zhang Q. CtBP1 is expressed in melanoma and represses the transcription of p16INK4a and Brca1. J Invest Dermatol 2013; 133:1294-301. [PMID: 23303449 PMCID: PMC3711675 DOI: 10.1038/jid.2012.487] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Carboxyl-terminal binding protein 1 (CtBP1) has been shown to suppress the transcription of several tumor suppressors in vitro. Paradoxically, a previous report showed that CtBP1 mRNA was down-regulated in melanoma. Using immunostaining, we found that a large percentage of human melanomas were positive for CtBP1 protein. Further, we demonstrated that CtBP1 expression in melanoma cells contributes to cell proliferation and genome instability, two aspects promoting melanoma initiation and progression. Breast Cancer Susceptibility Gene 1(Brca1), a core protein in DNA damage repair, was repressed by CtBP1 in melanoma cells. Consistently, Brca1 loss was found in human malignant melanoma tissues inversely correlated with CtBP1 expression levels. Additionally, the inhibitor of cyclin-dependent protein kinases (CDKs), p16INK4a, whose loss has been related to the pathogenesis of melanoma, was repressed by CtBP1 as well. Our findings suggest an important role of CtBP1 in the transcriptional control of p16INK4a and Brca1, with CtBP1 over-expression potentially contributing to increased proliferation and DNA damage in melanoma.
Collapse
Affiliation(s)
- Hui Deng
- Department of Dermatology, University of Colorado, Denver, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zandberg DP, Bhargava R, Badin S, Cullen KJ. The role of human papillomavirus in nongenital cancers. CA Cancer J Clin 2013; 63:57-81. [PMID: 23258613 DOI: 10.3322/caac.21167] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 10/11/2012] [Accepted: 10/12/2012] [Indexed: 12/17/2022] Open
Abstract
Human papillomavirus (HPV), one of the most common sexually transmitted diseases worldwide, has an established role in the pathogenesis of genital malignancies such as cervical cancer. The virus has also been implicated in the oncogenesis of nongenital cancers including head and neck malignancies (specifically oropharyngeal cancers) as well as anal cancer. There is less clarity regarding its role in lung and esophageal cancers. Worldwide, the incidence and prevalence of HPV-associated oropharyngeal cancer has been increasing over time. These patients have improved outcomes compared with those with HPV-negative oropharyngeal cancers, and there is continued interest in designing treatments specifically for this HPV-positive subgroup. Clinicians continue to gain an understanding of HPV in anal cancers and the risk factors associated with infection and progression to malignancy. This has potential implications for the eventual screening of high-risk groups. While HPV vaccination is currently approved for the prevention of cervical cancer, it also has potential in the prevention of all HPV-associated malignancies. In this review, current understanding of the role of HPV in nongenital cancers is discussed, as well as future implications for treatment and prevention.
Collapse
Affiliation(s)
- Dan P Zandberg
- Division of Hematology/Oncology, Department of Medicine, University of Maryland School of Medicine and Marlene and Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
25
|
Yousef AF, Fonseca GJ, Cohen MJ, Mymryk JS. The C-terminal region of E1A: a molecular tool for cellular cartography. Biochem Cell Biol 2012; 90:153-63. [DOI: 10.1139/o11-080] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The adenovirus E1A proteins function via protein–protein interactions. By making many connections with the cellular protein network, individual modules of this virally encoded hub reprogram numerous aspects of cell function and behavior. Although many of these interactions have been thoroughly studied, those mediated by the C-terminal region of E1A are less well understood. This review focuses on how this region of E1A affects cell cycle progression, apoptosis, senescence, transformation, and conversion of cells to an epithelial state through interactions with CTBP1/2, DYRK1A/B, FOXK1/2, and importin-α. Furthermore, novel potential pathways that the C-terminus of E1A influences through these connections with the cellular interaction network are discussed.
Collapse
Affiliation(s)
- Ahmed F. Yousef
- Departments of Microbiology and Immunology and Oncology, The University of Western Ontario, London Regional Cancer Program, London, ON N6A 4L6, Canada
| | - Gregory J. Fonseca
- Departments of Microbiology and Immunology and Oncology, The University of Western Ontario, London Regional Cancer Program, London, ON N6A 4L6, Canada
| | - Michael J. Cohen
- Departments of Microbiology and Immunology and Oncology, The University of Western Ontario, London Regional Cancer Program, London, ON N6A 4L6, Canada
| | - Joe S. Mymryk
- Departments of Microbiology and Immunology and Oncology, The University of Western Ontario, London Regional Cancer Program, London, ON N6A 4L6, Canada
| |
Collapse
|
26
|
Dorman K, Shen Z, Yang C, Ezzat S, Asa SL. CtBP1 interacts with Ikaros and modulates pituitary tumor cell survival and response to hypoxia. Mol Endocrinol 2012; 26:447-57. [PMID: 22301782 DOI: 10.1210/me.2011-1095] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
C-terminal binding protein (CtBP) is a transcriptional corepressor that plays an important role in mammalian development and tumorigenesis. We demonstrate that CtBP is expressed in adenohypophyseal cells and is expressed at high levels in human corticotroph, somatotroph, and lactotroph pituitary adenomas. CtBP interacts with Ikaros isoforms in GH4 and AtT20 pituitary tumor cells. Ikaros and CtBP1 expression is coordinately induced by hypoxia, and this response is abrogated by CtBP1 deficiency. Forced reduction of CtBP1 leads to reduced cell growth, up-regulation of Sprouty 2, and down-regulation of ectonucleotide pyrophosphatase phosphodiesterase 2 (Enpp2). Consistent with diminished Enpp2 activity, CtBP1-deficient pituitary cells are more susceptible to hypoxia-induced apoptosis, which is rescued by Enpp2-derived lysophosphatidic acid treatment. These results identify putative oncogenic properties of CtBP1 and provide new insights into the overlapping functions of two members of the chromatin remodeling network in the response to hypoxic pituitary tumor cell drive.
Collapse
Affiliation(s)
- Katie Dorman
- Departments of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
27
|
Li J, Poi MJ, Tsai MD. Regulatory mechanisms of tumor suppressor P16(INK4A) and their relevance to cancer. Biochemistry 2011; 50:5566-82. [PMID: 21619050 PMCID: PMC3127263 DOI: 10.1021/bi200642e] [Citation(s) in RCA: 233] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
P16(INK4A) (also known as P16 and MTS1), a protein consisting exclusively of four ankyrin repeats, is recognized as a tumor suppressor mainly because of the prevalence of genetic inactivation of the p16(INK4A) (or CDKN2A) gene in virtually all types of human cancers. However, it has also been shown that an elevated level of expression (upregulation) of P16 is involved in cellular senescence, aging, and cancer progression, indicating that the regulation of P16 is critical for its function. Here, we discuss the regulatory mechanisms of P16 function at the DNA level, the transcription level, and the posttranscriptional level, as well as their implications for the structure-function relationship of P16 and for human cancers.
Collapse
Affiliation(s)
- Junan Li
- Division of Environmental Health Sciences, College of Public Health, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio 43210, USA.
| | | | | |
Collapse
|
28
|
Deng Y, Deng H, Liu J, Han G, Malkoski S, Liu B, Zhao R, Wang XJ, Zhang Q. Transcriptional down-regulation of Brca1 and E-cadherin by CtBP1 in breast cancer. Mol Carcinog 2011; 51:500-7. [PMID: 21681822 DOI: 10.1002/mc.20813] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 05/03/2011] [Accepted: 05/12/2011] [Indexed: 12/12/2022]
Abstract
Carboxyl-terminal binding protein 1 (CtBP1) is a transcriptional co-repressor with oncogenic potential. Immunohistochemistry staining using human breast cancer tissue arrays revealed that 92% of invasive ductal breast cancer cases have CtBP1-positive staining compared to 4% CtBP1-positive in normal breast tissue. To explore the functional impact of CtBP1 in breast cancer, we examined CtBP1's transcriptional regulation of known tumor suppressors, breast cancer susceptibility gene 1 (Brca1), and E-cadherin. We found CtBP1 was recruited to the promoter regions of Brca1 and E-cadherin genes in breast cancer cells. Concomitantly, Brca1 loss was detected in 57% and E-cadherin loss was detected in 76% of human invasive ductal breast cancers, and correlated with CtBP1 nuclear staining in these lesions. Importantly, siRNA knock down of CtBP1 restored Brca1 and E-cadherin expression in breast cancer cell lines, implying CtBP1 down-regulates Brca1 and E-cadherin genes in human breast cancer. This study provides evidence that although genetic loss of Brca1 and E-cadherin are infrequent in breast cancer, they are down-regulated at the transcriptional level by CtBP1 expression. Thus, CtBP1 activation could be a potential biomarker for breast cancer development.
Collapse
Affiliation(s)
- Yu Deng
- Department of Dermatology, University of Colorado, Denver, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Background information. CtBPs [C-terminal (of E1A) binding protein] have roles in the nucleus as transcriptional co-repressors, and in the cytoplasm in the maintenance of vesicular membranes. CtBPs are expressed from two genes, CTBP1 and CTBP2, mRNA products of which are alternatively spliced at their 5′-ends to generate distinct protein isoforms. Extensive molecular and cellular analyses have identified CtBPs as regulators of pathways critical for tumour initiation, progression and response to therapy. However, little is known of the expression or regulation of CtBP isoforms in human cancer, nor of the relative contributions of CTBP1 and CTBP2 to the tumour cell phenotype. Results. Expression of CtBP proteins and CTBP1 and CTBP2 mRNA splice forms in breast cancer cell lines and tumour tissue was examined. CtBP1 proteins are identifiable as a single band on Western blots and are ubiquitously detectable in breast tumour samples, by both Western blotting and immunohistochemistry. CtBP1 is present in six of six breast cancer cell lines, although it is barely detectable in SKBr3 cells due to reduced CTBP1 mRNA expression. In the cell lines, the predominant CTBP1 mRNA splice form encodes CtBP1-S protein; in tumours, both major CTBP1 mRNA splice forms are variably expressed. CtBP2 proteins are ubiquitously expressed in all lines and tumour samples. The predominant CTBP2 mRNA encodes CtBP2-L, although an alternatively spliced form that encodes CtBP2-S, previously unidentified in humans, is expressed at low abundance. Both CtBP2-L and CtBP2-S are readily detectable as two distinct bands on Western blots; here we show that the CTBP2-L mRNA is translated from two AUG codons to generate both CtBP2-L and CtBP2-S. We have also identified an autoregulatory feedback mechanism whereby CtBP protein abundance is maintained in proliferating breast cancer cells through the post-transcriptional regulation of CtBP2. This feedback is disrupted by UV-C radiation or exposure to cisplatin. Finally, we demonstrate that CtBP1 and CtBP2 both have p53-dependent and -independent roles in suppressing the sensitivity of breast cancer cells to mechanistically diverse cancer chemotherapeutic agents. Conclusions. These studies support recent evidence that CtBP family proteins represent potential targets for therapeutic strategies for the treatment of cancer in general, and breast cancer in particular.
Collapse
|
30
|
Lanigan F, Geraghty JG, Bracken AP. Transcriptional regulation of cellular senescence. Oncogene 2011; 30:2901-11. [PMID: 21383691 DOI: 10.1038/onc.2011.34] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cellular senescence is an irreversible arrest of proliferation. It is activated when a cell encounters stress such as DNA damage, telomere shortening or oncogene activation. Like apoptosis, it impedes tumour progression and acts as a barrier that pre-neoplastic cells must overcome during their evolution toward the full tumourigenic state. This review focuses on the role of transcriptional regulators in the control of cellular senescence, explores how their function is perturbed in cancer and discusses the potential to harness this knowledge for future cancer therapies.
Collapse
Affiliation(s)
- F Lanigan
- Smurfit Genetics Department, The Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | | | | |
Collapse
|
31
|
|
32
|
Straza MW, Paliwal S, Kovi RC, Rajeshkumar B, Trenh P, Parker D, Whalen GF, Lyle S, Schiffer CA, Grossman SR. Therapeutic targeting of C-terminal binding protein in human cancer. Cell Cycle 2010; 9:3740-50. [PMID: 20930544 DOI: 10.4161/cc.9.18.12936] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The CtBP transcriptional corepressors promote cancer cell survival and migration/invasion. CtBP senses cellular metabolism via a regulatory dehydrogenase domain, and is antagonized by p14/p19(ARF) tumor suppressors. The CtBP dehydrogenase substrate 4-methylthio-2-oxobutyric acid (MTOB) can act as a CtBP inhibitor at high concentrations, and is cytotoxic to cancer cells. MTOB induced apoptosis was p53-independent, correlated with the derepression of the proapoptotic CtBP repression target Bik, and was rescued by CtBP overexpression or Bik silencing. MTOB did not induce apoptosis in mouse embryonic fibroblasts (MEFs), but was increasingly cytotoxic to immortalized and transformed MEFs, suggesting that CtBP inhibition may provide a suitable therapeutic index for cancer therapy. In human colon cancer cell peritoneal xenografts, MTOB treatment decreased tumor burden and induced tumor cell apoptosis. To verify the potential utility of CtBP as a therapeutic target in human cancer, the expression of CtBP and its negative regulator ARF was studied in a series of resected human colon adenocarcinomas. CtBP and ARF levels were inversely-correlated, with elevated CtBP levels (compared with adjacent normal tissue) observed in greater than 60% of specimens, with ARF absent in nearly all specimens exhibiting elevated CtBP levels. Targeting CtBP may represent a useful therapeutic strategy in human malignancies.
Collapse
Affiliation(s)
- Michael W Straza
- Department of Cancer Biology, University of Massachusetts Medical School and UMass Memorial Cancer Center, Worcester, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Dayyani F, Etzel CJ, Liu M, Ho CH, Lippman SM, Tsao AS. Meta-analysis of the impact of human papillomavirus (HPV) on cancer risk and overall survival in head and neck squamous cell carcinomas (HNSCC). HEAD & NECK ONCOLOGY 2010; 2:15. [PMID: 20587061 PMCID: PMC2908081 DOI: 10.1186/1758-3284-2-15] [Citation(s) in RCA: 361] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 06/29/2010] [Indexed: 11/26/2022]
Abstract
BACKGROUND HPV is important in a subset of HNSCC. Our meta-analysis determined the clinical characteristics of HPV-related HNSCC. METHOD Pubmed search terms "HPV" and "HNSCC" were used to identify 34 studies since 1980. We obtained pooled adjusted odds ratio (OR) or hazard ratio (HR) using random or fixed-effects model and compared OS depicted in forest plot. RESULTS A total of 5681 patients were included. The prevalence of HPV+ tumors was 22%, with 86.7% of HPV16+ genotype. The OR for HNSCC in HPV16+ patients was 4.44 (95% CI = 2.87-6.02). HPV status was associated with p16 expression (adj OR = 3.00; 0.90-9.70), and HPV+ tumors were less likely to harbor p53 mutations (adj OR = 0.21; 0.04-0.38). The HR for death in HPV+ patients was 0.42 (0.27-0.57). HPV+ HNSCC also had a better response to therapy. CONCLUSION HPV+ HNSCC are established as a separate biologic entity. Prospective trials are needed to establish the optimal therapy for HPV+ HNSCC.
Collapse
Affiliation(s)
- Farshid Dayyani
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, 77030, USA
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, 77030, USA
| | - Carol J Etzel
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, 77030, USA
| | - Mei Liu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, 77030, USA
| | - Chung-Han Ho
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, 77030, USA
| | - Scott M Lippman
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, 77030, USA
| | - Anne S Tsao
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, 77030, USA
| |
Collapse
|
34
|
Skalska L, White RE, Franz M, Ruhmann M, Allday MJ. Epigenetic repression of p16(INK4A) by latent Epstein-Barr virus requires the interaction of EBNA3A and EBNA3C with CtBP. PLoS Pathog 2010; 6:e1000951. [PMID: 20548956 PMCID: PMC2883600 DOI: 10.1371/journal.ppat.1000951] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 05/12/2010] [Indexed: 12/14/2022] Open
Abstract
As an inhibitor of cyclin-dependent kinases, p16INK4A is an important tumour suppressor and inducer of cellular senescence that is often inactivated during the development of cancer by promoter DNA methylation. Using newly established lymphoblastoid cell lines (LCLs) expressing a conditional EBNA3C from recombinant EBV, we demonstrate that EBNA3C inactivation initiates chromatin remodelling that resets the epigenetic status of p16INK4A to permit transcriptional activation: the polycomb-associated repressive H3K27me3 histone modification is substantially reduced, while the activation-related mark H3K4me3 is modestly increased. Activation of EBNA3C reverses the distribution of these epigenetic marks, represses p16INK4A transcription and allows proliferation. LCLs lacking EBNA3A express relatively high levels of p16INK4A and have a similar pattern of histone modifications on p16INK4A as produced by the inactivation of EBNA3C. Since binding to the co-repressor of transcription CtBP has been linked to the oncogenic activity of EBNA3A and EBNA3C, we established LCLs with recombinant viruses encoding EBNA3A- and/or EBNA3C-mutants that no longer bind CtBP. These novel LCLs have revealed that the chromatin remodelling and epigenetic repression of p16INK4A requires the interaction of both EBNA3A and EBNA3C with CtBP. The repression of p16INK4A by latent EBV will not only overcome senescence in infected B cells, but may also pave the way for p16INK4A DNA methylation during B cell lymphomagenesis. We previously showed that two Epstein-Barr virus latency-associated proteins—EBNA3A and EBNA3C—contribute to enhanced B cell survival by inhibiting the expression of the death-inducing protein BIM. This repression involves remodelling of the BIM gene promoter by polycomb proteins and DNA methylation within an unusually large CpG-island that flanks the transcription initiation site. Here we show that the same two proteins, EBNA3A and EBNA3C, functionally cooperate in the polycomb-mediated chromatin remodelling of another tumour suppressor gene, p16INK4A, that encodes a cyclin-dependent kinase inhibitor capable of blocking cell proliferation. Both EBV proteins can bind the highly conserved co-repressor of transcription CtBP, and these interactions appear to be required for the efficient repression of p16INK4A. Thus by utilising the polycomb system to induce the heritable repression of two major tumour suppressor genes—one that induces cell death (BIM) and one that induces growth arrest (p16INK4A)—EBV profoundly alters latently infected B cells and their progeny, making them significantly more prone to malignant transformation.
Collapse
Affiliation(s)
- Lenka Skalska
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Robert E. White
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Melanie Franz
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Michaela Ruhmann
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Martin J. Allday
- Section of Virology, Division of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
35
|
Ohashi S, Natsuizaka M, Wong GS, Michaylira CZ, Grugan KD, Stairs DB, Kalabis J, Vega ME, Kalman RA, Nakagawa M, Klein-Szanto AJ, Herlyn M, Diehl JA, Rustgi AK, Nakagawa H. Epidermal growth factor receptor and mutant p53 expand an esophageal cellular subpopulation capable of epithelial-to-mesenchymal transition through ZEB transcription factors. Cancer Res 2010; 70:4174-84. [PMID: 20424117 DOI: 10.1158/0008-5472.can-09-4614] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Transforming growth factor-beta (TGF-beta) is a potent inducer of epithelial to mesenchymal transition (EMT). However, it remains elusive about which molecular mechanisms determine the cellular capacity to undergo EMT in response to TGF-beta. We have found that both epidermal growth factor receptor (EGFR) overexpression and mutant p53 tumor suppressor genes contribute to the enrichment of an EMT-competent cellular subpopulation among telomerase-immortalized human esophageal epithelial cells during malignant transformation. EGFR overexpression triggers oncogene-induced senescence, accompanied by the induction of cyclin-dependent kinase inhibitors p15(INK4B), p16(INK4A), and p21. Interestingly, a subpopulation of cells emerges by negating senescence without loss of EGFR overexpression. Such cell populations express increased levels of zinc finger E-box binding (ZEB) transcription factors ZEB1 and ZEB2, and undergo EMT on TGF-beta stimulation. Enrichment of EMT-competent cells was more evident in the presence of p53 mutation, which diminished EGFR-induced senescence. RNA interference directed against ZEB resulted in the induction of p15(INK4B) and p16(INK4A), reactivating the EGFR-dependent senescence program. Importantly, TGF-beta-mediated EMT did not take place when cellular senescence programs were activated by either ZEB knockdown or the activation of wild-type p53 function. Thus, senescence checkpoint functions activated by EGFR and p53 may be evaded through the induction of ZEB, thereby allowing the expansion of an EMT-competent unique cellular subpopulation, providing novel mechanistic insights into the role of ZEB in esophageal carcinogenesis.
Collapse
Affiliation(s)
- Shinya Ohashi
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhao LJ, Kuppuswamy M, Vijayalingam S, Chinnadurai G. Interaction of ZEB and histone deacetylase with the PLDLS-binding cleft region of monomeric C-terminal binding protein 2. BMC Mol Biol 2009; 10:89. [PMID: 19754958 PMCID: PMC2749851 DOI: 10.1186/1471-2199-10-89] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Accepted: 09/15/2009] [Indexed: 11/25/2022] Open
Abstract
Background Proteins of the C-terminal binding protein (CtBP) family, CtBP1 and CtBP2 are closely related transcriptional regulators that are coded by two different gene loci in the vertebrate genomes. They perform redundant and unique functions during animal development. CtBP proteins mediate their transcriptional function through interaction with various DNA-binding repressors that contain PLDLS-like motifs and chromatin modifying enzymes, such as class I histone deacetylases (HDAC) that do not contain such motifs. The N-terminal region of CtBP1/2 forms a hydrophobic cleft and is involved in interaction with both PLDLS-containing factors and non-PLDLS factors. CtBP proteins function as dimers to mediate transcriptional repression and dimerization is modulated by specific binding to NAD/NADH. Results In this study, we have investigated the role of dimerization of CtBP2 in recruitment of PLDLS-motif cofactors and non-PLDLS cofactors. Our results indicate that mutations in CtBP2 that interfere with dimerization abolish CtBP2 interaction with most cellular factors, except the PLDLS-motif factor zinc-finger E-box binding homeobox (ZEB) and the non-PLDLS factor HDAC2. Unlike most PLDLS-containing CtBP-binding proteins, ZEB contains three PLDLS-like motifs and all three contribute to the interaction with the CtBP2 monomer. Despite the ability to interact with ZEB and HDAC, the CtBP2 monomer fails to mediate ZEB-dependent transcriptional repression. The lack of repression activity of the CtBP2 monomer is correlated with the competition between ZEB and HDAC for interaction with the CtBP2 monomer. Conclusion These results suggest a competition between the canonical PLDLS-motif factors such as E1A and non-PLDLS factor HDAC for interaction with CtBP. They also indicate that the affinity for the CtBP monomer may be determined by the number as well as amino acid sequence compositions of the PLDLS-like motifs. Our results are consistent with a model that the CtBP2 dimer may interact with a PLDLS-containing repressor through one monomer and recruit HDAC and other chromatin modifying enzymes through the second monomer in the CtBP2 dimer.
Collapse
Affiliation(s)
- Ling-Jun Zhao
- Institute for Molecular Virology, Saint Louis University Health Sciences Center, St, Louis, Missouri 63104, USA.
| | | | | | | |
Collapse
|
37
|
Nichols AC, Faquin WC, Westra WH, Mroz EA, Begum S, Clark JR, Rocco JW. HPV-16 infection predicts treatment outcome in oropharyngeal squamous cell carcinoma. Otolaryngol Head Neck Surg 2009; 140:228-34. [DOI: 10.1016/j.otohns.2008.11.025] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2008] [Revised: 10/31/2008] [Accepted: 11/13/2008] [Indexed: 11/25/2022]
Abstract
Objective: To determine if patients with human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC) treated with chemoradiation have improved outcomes. Study Design: A retrospective search was used to identify patients with OPSCC treated with concurrent chemoradiation. Pretreatment biopsy specimens were tested for HPV-16 infection and p16 expression. Methods: Forty-four patients with OPSCC treated with concurrent chemotherapy and intensity-modulated radiation therapy were identified. Eligibility criteria included a minimum two years of follow-up, or biopsy-proven recurrence. In situ hybridization was applied to archival tumor specimens, with HPV-16-positive status defined as positive staining of tumor cell nuclei. p16 expression was assessed by immunohistochemistry. Results: Twenty-seven tumors (61%) were positive for HPV-16 and 29 tumors (66%) expressed p16. HPV-16 infection was highly correlated with p16 expression ( P < 10−7). Three-year disease-free and overall survival for all patients was 66 percent and 79 percent respectively. Patients with tumors infected with HPV-16 had improved overall (OS) and disease-free survival (DFS) after chemoradiation (OS: hazard ratio [HR] = 0.21, P = 0.01; DFS: HR = 0.30, P = 0.02). Conclusion: Patients with OPSCC tumors that are infected with HPV-16 have improved survival after treatment with concurrent chemoradiation.
Collapse
Affiliation(s)
- Anthony C. Nichols
- From the Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary, Boston, MA (Drs Nichols and Rocco); Departments of Pathology (Dr Faquin), Surgery (Drs. Mroz and Rocco), and Medical Oncology (Dr Clark), Massachusetts General Hospital, Boston, MA; Department of Pathology, Johns Hopkins Hospital, Baltimore, MD (Drs Westra and Begum); and MGH Cancer Center, Massachusetts General Hospital, Boston, MA (Drs Mroz and Rocco)
| | - William C. Faquin
- From the Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary, Boston, MA (Drs Nichols and Rocco); Departments of Pathology (Dr Faquin), Surgery (Drs. Mroz and Rocco), and Medical Oncology (Dr Clark), Massachusetts General Hospital, Boston, MA; Department of Pathology, Johns Hopkins Hospital, Baltimore, MD (Drs Westra and Begum); and MGH Cancer Center, Massachusetts General Hospital, Boston, MA (Drs Mroz and Rocco)
| | - William H. Westra
- From the Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary, Boston, MA (Drs Nichols and Rocco); Departments of Pathology (Dr Faquin), Surgery (Drs. Mroz and Rocco), and Medical Oncology (Dr Clark), Massachusetts General Hospital, Boston, MA; Department of Pathology, Johns Hopkins Hospital, Baltimore, MD (Drs Westra and Begum); and MGH Cancer Center, Massachusetts General Hospital, Boston, MA (Drs Mroz and Rocco)
| | - Edmund A. Mroz
- From the Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary, Boston, MA (Drs Nichols and Rocco); Departments of Pathology (Dr Faquin), Surgery (Drs. Mroz and Rocco), and Medical Oncology (Dr Clark), Massachusetts General Hospital, Boston, MA; Department of Pathology, Johns Hopkins Hospital, Baltimore, MD (Drs Westra and Begum); and MGH Cancer Center, Massachusetts General Hospital, Boston, MA (Drs Mroz and Rocco)
| | - Shanaz Begum
- From the Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary, Boston, MA (Drs Nichols and Rocco); Departments of Pathology (Dr Faquin), Surgery (Drs. Mroz and Rocco), and Medical Oncology (Dr Clark), Massachusetts General Hospital, Boston, MA; Department of Pathology, Johns Hopkins Hospital, Baltimore, MD (Drs Westra and Begum); and MGH Cancer Center, Massachusetts General Hospital, Boston, MA (Drs Mroz and Rocco)
| | - John R. Clark
- From the Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary, Boston, MA (Drs Nichols and Rocco); Departments of Pathology (Dr Faquin), Surgery (Drs. Mroz and Rocco), and Medical Oncology (Dr Clark), Massachusetts General Hospital, Boston, MA; Department of Pathology, Johns Hopkins Hospital, Baltimore, MD (Drs Westra and Begum); and MGH Cancer Center, Massachusetts General Hospital, Boston, MA (Drs Mroz and Rocco)
| | - James W. Rocco
- From the Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary, Boston, MA (Drs Nichols and Rocco); Departments of Pathology (Dr Faquin), Surgery (Drs. Mroz and Rocco), and Medical Oncology (Dr Clark), Massachusetts General Hospital, Boston, MA; Department of Pathology, Johns Hopkins Hospital, Baltimore, MD (Drs Westra and Begum); and MGH Cancer Center, Massachusetts General Hospital, Boston, MA (Drs Mroz and Rocco)
| |
Collapse
|
38
|
Chinnadurai G. The transcriptional corepressor CtBP: a foe of multiple tumor suppressors. Cancer Res 2009; 69:731-4. [PMID: 19155295 DOI: 10.1158/0008-5472.can-08-3349] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
CtBP1 and CtBP2 are closely related and evolutionarily conserved transcriptional corepressors. There is strong evidence linking CtBPs to tumorigenesis and tumor progression. CtBPs promote epithelial-mesenchymal transition and function as apoptosis antagonists. Also, CtBPs mediate repression of several tumor suppressor genes. Certain tumor suppressors also target CtBPs to restrain their tumor-promoting activity. Down-regulation of CtBPs mediated by some tumor suppressors results in p53-independent apoptosis and reduced tumor cell migration and invasion. The role of CtBPs in modulating the activities of different tumor suppressors is reviewed here. The results discussed here suggest that CtBPs may constitute a novel p53-independent anticancer target.
Collapse
Affiliation(s)
- G Chinnadurai
- Institute for Molecular Virology, Saint Louis University Health Sciences Center, Doisy Research Center, St Louis, Missouri 63104, USA.
| |
Collapse
|