1
|
Negrón-Piñeiro LJ, Wu Y, Popsuj S, José-Edwards DS, Stolfi A, Di Gregorio A. Cis-regulatory interfaces reveal the molecular mechanisms underlying the notochord gene regulatory network of Ciona. Nat Commun 2024; 15:3025. [PMID: 38589372 PMCID: PMC11001920 DOI: 10.1038/s41467-024-46850-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
Tissue-specific gene expression is fundamental in development and evolution, and is mediated by transcription factors and by the cis-regulatory regions (enhancers) that they control. Transcription factors and their respective tissue-specific enhancers are essential components of gene regulatory networks responsible for the development of tissues and organs. Although numerous transcription factors have been characterized from different organisms, the knowledge of the enhancers responsible for their tissue-specific expression remains fragmentary. Here we use Ciona to study the enhancers associated with ten transcription factors expressed in the notochord, an evolutionary hallmark of the chordate phylum. Our results illustrate how two evolutionarily conserved transcription factors, Brachyury and Foxa2, coordinate the deployment of other notochord transcription factors. The results of these detailed cis-regulatory analyses delineate a high-resolution view of the essential notochord gene regulatory network of Ciona, and provide a reference for studies of transcription factors, enhancers, and their roles in development, disease, and evolution.
Collapse
Affiliation(s)
- Lenny J Negrón-Piñeiro
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Yushi Wu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Sydney Popsuj
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Diana S José-Edwards
- Post-Baccalaureate Premedical Program, Washington University, St. Louis, MO, 63130, USA
| | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA.
| |
Collapse
|
2
|
Role of Epstein-Barr Virus and Human Papillomavirus Coinfection in Cervical Cancer: Epidemiology, Mechanisms and Perspectives. Pathogens 2020; 9:pathogens9090685. [PMID: 32839399 PMCID: PMC7557835 DOI: 10.3390/pathogens9090685] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 12/24/2022] Open
Abstract
High-risk human papillomavirus (HR-HPV) is etiologically associated with the development and progression of cervical cancer, although other factors are involved. Epstein-Barr virus (EBV) detection in premalignant and malignant tissues from uterine cervix has been widely reported; however, its contribution to cervical cancer development is still unclear. Here, a comprehensive analysis regarding EBV presence and its potential role in cervical cancer, the frequency of EBV/HR-HPV coinfection in uterine cervix and EBV infection in tissue-infiltrating lymphocytes were revised. Overall, reports suggest a potential link of EBV to the development of cervical carcinomas in two possible pathways: (1) Infecting epithelial cells, thus synergizing with HR-HPV (direct pathway), and/or (2) infecting tissue-infiltrating lymphocytes that could generate local immunosuppression (indirect pathway). In situ hybridization (ISH) and/or immunohistochemical methods are mandatory for discriminating the cell type infected by EBV. However, further studies are needed for a better understanding of the EBV/HR-HPV coinfection role in cervical carcinogenesis.
Collapse
|
3
|
The regulatory role of protein phosphorylation in human gammaherpesvirus associated cancers. Virol Sin 2017; 32:357-368. [PMID: 29116588 PMCID: PMC6704201 DOI: 10.1007/s12250-017-4081-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022] Open
Abstract
Activation of specific sets of protein kinases by intracellular signal molecules
has become more and more apparent in the past decade. Phosphorylation, one of key
posttranslational modification events, is activated by kinase or regulatory protein
and is vital for controlling many physiological functions of eukaryotic cells such
as cell proliferation, differentiation, malignant transformation, and signal
transduction mediated by external stimuli. Moreovers, the reversible modification of
phosphorylation and dephosphorylation can result in different features of the target
substrate molecules including DNA binding, protein-protein interaction, subcellular
location and enzymatic activity, and is often hijacked by viral infection.
Epstein-Barr virus (EBV) and Kaposi’s sarcomaassociated herpesvirus (KSHV), two
human oncogenic gamma-herpesviruses, are shown to tightly associate with many
malignancies. In this review, we summarize the recent progresses on understanding of
molecular properties and regulatory modes of cellular and viral proteins
phosphorylation influenced by these two tumor viruses, and highlight the potential
therapeutic targets and strategies against their related cancers. ![]()
Collapse
|
4
|
Nakanishi Y, Wakisaka N, Kondo S, Endo K, Sugimoto H, Hatano M, Ueno T, Ishikawa K, Yoshizaki T. Progression of understanding for the role of Epstein-Barr virus and management of nasopharyngeal carcinoma. Cancer Metastasis Rev 2017; 36:435-447. [PMID: 28819752 PMCID: PMC5613035 DOI: 10.1007/s10555-017-9693-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is very common in southern China and Southeast Asia. In regions where NPC is endemic, undifferentiated subtypes constitute most cases and are invariably associated with Epstein-Barr virus (EBV) infection, whereas the differentiated subtype is more common in other parts of the world. Undifferentiated NPC is a unique malignancy with regard to its epidemiology, etiology, and clinical presentation. Clinically, NPC is highly invasive and metastatic, but sensitive to both chemotherapy and radiotherapy (RT). Overall prognosis has dramatically improved over the past three decades because of advances in management, including the improvement of RT technology, the broader application of chemotherapy, and more accurate disease staging. Despite the excellent local control with modern RT, distant failure remains a challenging problem. Advances in molecular technology have helped to elucidate the molecular pathogenesis of NPC. This article reviews the contribution of EBV gene products to NPC pathogenesis and the current management of NPC.
Collapse
Affiliation(s)
- Yosuke Nakanishi
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8640, Japan
| | - Naohiro Wakisaka
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8640, Japan
| | - Satoru Kondo
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8640, Japan
| | - Kazuhira Endo
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8640, Japan
| | - Hisashi Sugimoto
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8640, Japan
| | - Miyako Hatano
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8640, Japan
| | - Takayoshi Ueno
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8640, Japan
| | - Kazuya Ishikawa
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8640, Japan
| | - Tomokazu Yoshizaki
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8640, Japan.
| |
Collapse
|
5
|
Arnold N, Messaoudi I. Simian varicella virus causes robust transcriptional changes in T cells that support viral replication. Virus Res 2017; 238:226-235. [PMID: 28698046 PMCID: PMC7114558 DOI: 10.1016/j.virusres.2017.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/24/2017] [Accepted: 07/04/2017] [Indexed: 12/13/2022]
Abstract
T cells play a major role in varicella viruses dissemination to ganglia and skin. SVV infection of T cells increases the expression of cell cycle genes. SVV infection downregulates genes important for antigen presentation in T cells. SVV T cell infection disrupts expression of genes vital for metabolism and immunity.
Varicella zoster virus (VZV) causes varicella (chickenpox) during acute infection. Several studies have shown that T cells are early and preferential targets of VZV infection that play a critical role in disseminating VZV in to the skin and ganglia. However, the transcriptional changes that occur in VZV-infected T cells remain unclear due to limited access to clinical samples and robust translational animal models. In this study, we used a nonhuman primate model of VZV infection where rhesus macaques are infected with the closely related Simian Varicella Virus (SVV) to provide novel insights into VZV-T cell interactions. RNA sequencing of bronchial alveolar lavage-resident T cells isolated from infected rhesus macaques show that SVV infection alters expression of genes important for regulation of gene expression, cell cycle progression, metabolism, and antiviral immunity. These data provide insight into cellular processes that may support viral replication, facilitate SVV dissemination, and evade host defense.
Collapse
Affiliation(s)
- Nicole Arnold
- Graduate Program in Microbiology, University of California, Riverside, CA, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California-Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
6
|
Lu S, Yan Y, Li Z, Chen L, Yang J, Zhang Y, Wang S, Liu L. Determination of Genes Related to Uveitis by Utilization of the Random Walk with Restart Algorithm on a Protein-Protein Interaction Network. Int J Mol Sci 2017; 18:ijms18051045. [PMID: 28505077 PMCID: PMC5454957 DOI: 10.3390/ijms18051045] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/14/2022] Open
Abstract
Uveitis, defined as inflammation of the uveal tract, may cause blindness in both young and middle-aged people. Approximately 10–15% of blindness in the West is caused by uveitis. Therefore, a comprehensive investigation to determine the disease pathogenesis is urgent, as it will thus be possible to design effective treatments. Identification of the disease genes that cause uveitis is an important requirement to achieve this goal. To begin to answer this question, in this study, a computational method was proposed to identify novel uveitis-related genes. This method was executed on a large protein–protein interaction network and employed a popular ranking algorithm, the Random Walk with Restart (RWR) algorithm. To improve the utility of the method, a permutation test and a procedure for selecting core genes were added, which helped to exclude false discoveries and select the most important candidate genes. The five-fold cross-validation was adopted to evaluate the method, yielding the average F1-measure of 0.189. In addition, we compared our method with a classic GBA-based method to further indicate its utility. Based on our method, 56 putative genes were chosen for further assessment. We have determined that several of these genes (e.g., CCL4, Jun, and MMP9) are likely to be important for the pathogenesis of uveitis.
Collapse
Affiliation(s)
- Shiheng Lu
- Department of Ophthalmology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Yan Yan
- Department of Ophthalmology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Zhen Li
- Department of Ophthalmology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China.
| | - Jing Yang
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Yuhang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Shaopeng Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Lin Liu
- Department of Ophthalmology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
7
|
Boutin SR, Rogers AB, Shen Z, Fry RC, Love JA, Nambiar PR, Suerbaum S, Fox JG. Hepatic Temporal Gene Expression Profiling in Helicobacter hepaticus-Infected A/JCr Mice. Toxicol Pathol 2016; 32:678-93. [PMID: 15513911 DOI: 10.1080/01926230490524058] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Helicobacter hepaticus infection of A/JCr mice is a model of infectious liver cancer. We monitored hepatic global gene expression profiles in H. hepaticus infected and control male A/JCr mice at 3 months, 6 months, and 1 year of age using an Affymetrix-based oligonucleotide microarray platform on the premise that a specific genetic expression signature at isolated time points would be indicative of disease status. Model based expression index comparisons generated by dChip yielded consistent profiles of differential gene expression for H. hepaticus infected male mice with progressive liver disease versus uninfected control mice within each age group. Linear discriminant analysis and principal component analysis allowed segregation of mice based on combined age and lesion status, or age alone. Up-regulation of putative tumor markers correlated with advancing hepatocellular dysplasia. Transcriptionally down-regulated genes in mice with liver lesions included those related to peroxisome proliferator, fatty acid, and steroid metabolism pathways. In conclusion, transcriptional profiling of hepatic genes documented gene expression signatures in the livers of H. hepaticus infected male A/JCr mice with chronic progressive hepatitis and preneoplastic liver lesions, complemented the histopathological diagnosis, and suggested molecular targets for the monitoring and intervention of disease progression prior to the onset of hepatocellular neoplasia.
Collapse
Affiliation(s)
- Samuel R Boutin
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Li B, Wan Z, Huang G, Huang Z, Zhang X, Liao D, Luo S, He Z. Mitogen- and stress-activated Kinase 1 mediates Epstein-Barr virus latent membrane protein 1-promoted cell transformation in nasopharyngeal carcinoma through its induction of Fra-1 and c-Jun genes. BMC Cancer 2015; 15:390. [PMID: 25958199 PMCID: PMC4434874 DOI: 10.1186/s12885-015-1398-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 04/29/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Mitogen- and Stress-Activated Kinase 1 (MSK1) is a nuclear kinase that serves as active link between extracellular signals and the primary response of gene expression. However, the involvement of MSK1 in malignant transformation and cancer development is not well understood. In this study, we aimed to explore the role of MSK1 in Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1)-promoted carcinogenesis of nasopharyngeal carcinoma (NPC). METHODS The level of MSK1 phosphorylation at Thr581 was detected by the immunohistochemical analysis in NPC tissues and normal nasopharynx tissues, and its correlation with LMP1 was analyzed in NPC tissues and cell lines. Using MSK1 inhibitor H89 or small interfering RNA (siRNA)-MSK1, the effects of MSK1 on LMP1-promoted CNE1 cell proliferation and transformation were evaluated by CCK-8 assay, flow cytometry and focus-forming assay respectively. Furthermore, the regulatory role of MSK1-mediated histone H3 phosphorylation at Ser10 on the promoter activity and expression of Fra-1 or c-Jun was determined by reporter gene assay and western blotting analysis. RESULTS Immunohistochemical analysis revealed that the level of MSK1 phosphorylation at Thr581 was significantly higher in the poorly differentiated NPC tissues than that in normal nasopharynx tissues (P < 0.001). Moreover, high level of phosphorylated MSK1 was positively correlated with the expression of LMP1 in NPC tissues (r = 0.393, P = 0.002) and cell lines. MSK1 inhibitor H89 or knockdown of MSK1 by siRNA dramatically suppressed LMP1-promoted CNE1 cell proliferation, which was associated with the induction of cell cycle arrest at G0/G1 phase. In addition, the anchorage-independent growth promoted by LMP1 was blocked in MSK1 knockdown cells. When the activity or expression of MSK1 was inhibited, LMP1-induced promoter activities of Fra-1 and c-Jun as well as their protein levels were greatly reduced. It was found that only H3 WT, but not mutant H3 S10A, dramatically increased LMP1 induction of Fra-1 and c-Jun genes compared with mock cells. CONCLUSION Increased MSK1 activity is critically important for LMP1-promoted cell proliferation and transformation in NPC, which may be correlated with its induction of Fra-1 and c-Jun through phosphorylation of histone H3 at Ser10.
Collapse
Affiliation(s)
- Binbin Li
- Department of Pathophysiology, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
- Key Laboratory for Medical Diagnostics of Guangdong Province, Sino-American Cancer Research Institute, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
| | - Zheng Wan
- Key Laboratory for Medical Diagnostics of Guangdong Province, Sino-American Cancer Research Institute, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
| | - Guoliang Huang
- Key Laboratory for Medical Diagnostics of Guangdong Province, Sino-American Cancer Research Institute, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
| | - Zunnan Huang
- Key Laboratory for Medical Diagnostics of Guangdong Province, Sino-American Cancer Research Institute, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
| | - Xiangning Zhang
- Department of Pathophysiology, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
| | - Dan Liao
- Department of Pathophysiology, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
| | - Shengqun Luo
- Department of Pathophysiology, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
| | - Zhiwei He
- Department of Pathophysiology, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
- Key Laboratory for Medical Diagnostics of Guangdong Province, Sino-American Cancer Research Institute, Guangdong Medical College, Dongguan, Guangdong, 523808, China.
| |
Collapse
|
9
|
Liu MT, Chen MK, Huang CC, Huang CY. Prognostic Value of Molecular Markers and Implication for Molecular Targeted Therapies in Nasopharyngeal Carcinoma: An Update in an Era of New Targeted Molecules Development. World J Oncol 2015; 6:243-261. [PMID: 29147412 PMCID: PMC5649942 DOI: 10.14740/wjon610w] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2012] [Indexed: 12/15/2022] Open
Abstract
The aim of the study was to evaluate the prognostic significance of molecular biomarkers which could provide information for more accurate prognostication and development of novel therapeutic strategies for nasopharyngeal carcinoma (NPC). NPC is a unique malignant epithelial carcinoma of head and neck region, with an intimate association with the Epstein-Barr virus (EBV). Currently, the prediction of NPC prognosis is mainly based on the clinical TNM staging; however, NPC patients with the same clinical stage often present different clinical outcomes, suggesting that the TNM stage is insufficient to precisely predict the prognosis of this disease. In this review, we give an overview of the prognostic value of molecular markers in NPC and discuss potential strategies of targeted therapies for treatment of NPC. Molecular biomarkers, which play roles in abnormal proliferation signaling pathways (such as Wnt/β-catenin pathway), intracellular mitogenic signal aberration (such as hypoxia-inducible factor (HIF)-1α), receptor-mediated aberrations (such as vascular endothelial growth factor (VEGF)), tumor suppressors (such as p16 and p27 activity), cell cycle aberrations (such as cyclin D1 and cyclin E), cell adhesion aberrations (such as E-cadherin), apoptosis dysregualtion (such as survivin) and centromere aberration (centromere protein H), are prognostic markers for NPC. Plasma EBV DNA concentrations and EBV-encoded latent membrane proteins are also prognostic markers for NPC. Implication of molecular targeted therapies in NPC was discussed. Such therapies could have potential in combination with different cytotoxic agents to combat and eradicate tumor cells. In order to further improve overall survival for patients with loco-regionally advanced NPC, the development of innovative strategies, including prognostic molecular markers and molecular targeted agents is needed.
Collapse
Affiliation(s)
- Mu-Tai Liu
- Department of Radiation Oncology, Changhua Christian Hospital, 135 Nan Shiau Street, Changhua, Taiwan 500, ROC.,Department of Oncology, National Taiwan University Hospital, 7 Chung San South Road, Taipei, Taiwan 100, ROC.,Department of Medicine, Chang Shan Medical University, 110 Section 1, Chien- Kuo N. Road, Taichung, Taiwan 402, ROC.,Department of Radiology, Yuanpei University of Science and Technology, 306 Yuanpei Street, Hsinchu, Taiwan 300, ROC
| | - Mu-Kuan Chen
- Department of Radiology, Yuanpei University of Science and Technology, 306 Yuanpei Street, Hsinchu, Taiwan 300, ROC.,Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, 135 Nan Shiau Street, Changhua, Taiwan 500, ROC
| | - Chia-Chun Huang
- Department of Radiation Oncology, Changhua Christian Hospital, 135 Nan Shiau Street, Changhua, Taiwan 500, ROC
| | - Chao-Yuan Huang
- Department of Oncology, National Taiwan University Hospital, 7 Chung San South Road, Taipei, Taiwan 100, ROC
| |
Collapse
|
10
|
Guo SY, Zhu XD, Ge LY, Qu S, Li L, Su F, Guo Y. RNAi-mediated knockdown of the c-jun gene sensitizes radioresistant human nasopharyngeal carcinoma cell line CNE-2R to radiation. Oncol Rep 2014; 33:1155-60. [PMID: 25571870 DOI: 10.3892/or.2014.3692] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 11/25/2014] [Indexed: 11/06/2022] Open
Abstract
This study aimed to investigate the effect of RNA interference (RNAi)-mediated downregulation of the expression of the c-jun gene (a proto-oncogene) on the radiosensitivity of a radioresistant human nasopharyngeal carcinoma cell line (CNE-2R) and to validate its potential as an anticancer target. A lentiviral vector with c-jun small hairpin RNA (shRNA) was constructed and transfected into CNE-2R cells. The gene silencing efficiency of these recombinants was confirmed by RT-PCR and western blotting. Radiosensitivity, cell proliferation, cell cycle profile and apoptosis were assessed using colony formation assay, CCK-8 assay and flow cytometry, respectively. The lentiviral shRNA efficiently knocked down the expression of c-jun at both the mRNA and protein levels (P<0.05). c-jun-downregulated CNE-2R cells exhibited significantly decreased cell proliferation and enhanced radiosensitivity compared to the control group (P<0.05), and the effects were likely due to G2/M phase arrest and enhanced cell apoptosis. These data provide evidence that c-jun may be involved in the radioresistance of nasopharyngeal carcinoma (NPC) and knockdown of the c-jun gene may be a potential strategy to enhance the radiation sensitivity of NPC.
Collapse
Affiliation(s)
- Si-Yan Guo
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Guangxi Medical University, Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Xiao-Dong Zhu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Guangxi Medical University, Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Lian-Ying Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Guangxi Medical University, Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Song Qu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Guangxi Medical University, Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Ling Li
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Guangxi Medical University, Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Fang Su
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Guangxi Medical University, Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Ya Guo
- Department of Oncology, The Second Affiliated Hospital of Xian Jiaotong University, Xian, Shaanxi 710049, P.R. China
| |
Collapse
|
11
|
Pathogenic role of Epstein-Barr virus latent membrane protein-1 in the development of nasopharyngeal carcinoma. Cancer Lett 2013; 337:1-7. [PMID: 23689138 DOI: 10.1016/j.canlet.2013.05.018] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/10/2013] [Accepted: 05/12/2013] [Indexed: 12/21/2022]
Abstract
Undifferentiated nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus-associated malignant tumor. A consistent elevation in EBV antibody titers is a well-established risk factor for the development of NPC. The pathophysiological relationship and molecular mechanisms of EBV-mediated carcinogenesis have not been fully elucidated. While NPC tumors are known to express three EBV-encoded proteins, EBNA1, LMP1, and LMP2, they also express a large number of virus-encoded small RNAs (EBERs) and microRNAs (miRNAs). Among them, LMP1 may be a central player in the development of NPC. LMP1, an EBV-encoded primary oncogene, functions as a viral mimic of the TNFR family member, CD40, and engages in a number of signaling pathways that induce morphological and phenotypic alterations in epithelial cells. LMP1 upregulates EMT, and contributes to the highly metastatic features of NPC. Moreover, LMP1-associated EMT is accompanied by the expression of cancer stem cell (CSC)/cancer progenitor cell (CPC) markers (CD44high/CD24low) and the acquisition of stem cell/progenitor cell-like properties. BART miRNAs, encoded from the BamHI-A region of the viral genome, are the most abundant transcripts. They modulate apoptosis and host innate immune defense mechanisms. Some BART1 miRNAs are considered to negatively regulate LMP1 protein expression. LMP1 is secreted via exosomes, is incorporated into EBV-uninfected cells by endocytosis, and affects the environment surrounding the tumor. Here we reviewed the contribution of EBV gene products to NPC pathogenesis in relation with LMP1.
Collapse
|
12
|
Li B, Huang G, Zhang X, Li R, Wang J, Dong Z, He Z. Increased phosphorylation of histone H3 at serine 10 is involved in Epstein-Barr virus latent membrane protein-1-induced carcinogenesis of nasopharyngeal carcinoma. BMC Cancer 2013; 13:124. [PMID: 23496845 PMCID: PMC3610199 DOI: 10.1186/1471-2407-13-124] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/11/2013] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Increased histone H3 phosphorylation is an essential regulatory mechanism for neoplastic cell transformation. We aimed to explore the role of histone H3 phosphorylation at serine10 (p-H3Ser10) in Epstein-Barr virus (EBV) latent membrane protein-1 (LMP1)-induced carcinogenesis of nasopharyngeal carcinoma (NPC). METHODS The expression of p-H3Ser10 was detected by the immunohistochemical analysis in NPC, chronic nasopharyngitis and normal nasopharynx tissues, and its correlation with LMP1 was analyzed in NPC tissues and cell lines. Using the small interfering RNA (siRNA)-H3 and histone H3 mutant (S10A), the effect of histone H3 Ser10 motif on LMP1-induced CNE1 cell proliferation, transformation and activator protein-1 (AP-1) activation were evaluated by CCK-8, focus-forming and reporter gene assay respectively. Mitogen- and stress-activated kinase 1 (MSK1) kinase activity and phosphorylation were detected by in vitro kinase assay and western blot. Using MSK1 inhibitor H89 or siRNA-MSK1, the regulatory role of MSK1 on histone H3 phosphorylation and AP-1 activation were analyzed. RESULTS Immunohistochemical analysis revealed that the expression of p-H3Ser10 was significantly higher in the poorly differentiated NPC tissues than that in chronic nasopharyngitis (p <0.05) and normal nasopharynx tissues (p <0.001). Moreover, high level of p-H3Ser10 was positively correlated with the expression of LMP1 in NPC tissues (χ2=6.700, p =0.01; C=0.350) and cell lines. The knockdown and mutant (S10A) of histone H3 suppressed LMP1-induced CNE1 cell proliferation, foci formation and AP-1 activation. In addition, LMP1 could increase MSK1 kinase activity and phosphorylation. MSK1 inhibitor H89 or knockdown of MSK1 by siRNA blocked LMP1-induced phosphorylation of histone H3 at Ser10 and AP-1 activation. CONCLUSION EBV-LMP1 can induce phosphorylation of histone H3 at Ser10 via MSK1. Increased phosphorylation of histone H3 at Ser10 is likely a crucial regulatory mechanism involved in LMP1-induced carcinogenesis of NPC.
Collapse
Affiliation(s)
- Binbin Li
- Department of Pathophysiology, Basic Medical College of Zhengzhou University, No.100 of Science Road, Zhengzhou, 450001, China
- Key Laboratory for Medical Diagnostics of Guangdong Province, Sino-American Cancer Research Institute, Guangdong Medical College, No. 1 Xincheng Road, Guangdong, Dongguan, 523808, China
| | - Guoliang Huang
- Key Laboratory for Medical Diagnostics of Guangdong Province, Sino-American Cancer Research Institute, Guangdong Medical College, No. 1 Xincheng Road, Guangdong, Dongguan, 523808, China
| | - Xiangning Zhang
- Department of Pathophysiology, Guangdong Medical College, Guangdong, Dongguan, 523808, China
| | - Rong Li
- Department of Pathophysiology, Guangdong Medical College, Guangdong, Dongguan, 523808, China
| | - Jian Wang
- Key Laboratory for Medical Diagnostics of Guangdong Province, Sino-American Cancer Research Institute, Guangdong Medical College, No. 1 Xincheng Road, Guangdong, Dongguan, 523808, China
| | - Ziming Dong
- Department of Pathophysiology, Basic Medical College of Zhengzhou University, No.100 of Science Road, Zhengzhou, 450001, China
| | - Zhiwei He
- Key Laboratory for Medical Diagnostics of Guangdong Province, Sino-American Cancer Research Institute, Guangdong Medical College, No. 1 Xincheng Road, Guangdong, Dongguan, 523808, China
| |
Collapse
|
13
|
Lin X, Tang M, Tao Y, Li L, Liu S, Guo L, Li Z, Ma X, Xu J, Cao Y. Epstein-Barr virus-encoded LMP1 triggers regulation of the ERK-mediated Op18/stathmin signaling pathway in association with cell cycle. Cancer Sci 2012; 103:993-9. [PMID: 22417000 DOI: 10.1111/j.1349-7006.2012.02271.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 12/17/2011] [Accepted: 02/26/2012] [Indexed: 11/30/2022] Open
Abstract
The MAPKs are activated by a variety of cellular stimuli to participate in a series of signaling cascades and mediate diverse intracellular responses. One potential target of the MAPKs is Op18/stathmin, a molecule that acts as an integrator of diverse cell signaling pathways and regulates the dynamics of microtubules, which are involved in modulating a variety of cellular processes, including cell cycle progression and cell growth. Our study focused on the regulation of the MAPK-mediated Op18/stathmin signaling pathway, which is triggered by the Epstein-Barr virus-encoded latent membrane protein 1 ( LMP1) oncogene in nasopharyngeal carcinoma cells. The results showed that the activity of MAPK, which was induced by LMP1, varied with cell cycle progression; LMP1 upregulated phosphorylation of ERK during the G(1)/S phase, but negatively regulated phosphorylation of ERK during the G(2)/M phase. We found that the regulation of Op18/stathmin signaling by LMP1 was mainly mediated through ERK. The inhibition of LMP1 expression attenuated the interaction of ERK with Op18/stathmin and promoted microtubule depolymerization. These findings indicate the existence of a new cell cycle-associated signaling pathway in which LMP1 regulates ERK-mediated Op18/stathmin signaling.
Collapse
Affiliation(s)
- Xuechi Lin
- Cancer Research Institute, Changsha, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Ma X, Yang L, Xiao L, Tang M, Liu L, Li Z, Deng M, Sun L, Cao Y. Down-regulation of EBV-LMP1 radio-sensitizes nasal pharyngeal carcinoma cells via NF-κB regulated ATM expression. PLoS One 2011; 6:e24647. [PMID: 22096476 PMCID: PMC3212510 DOI: 10.1371/journal.pone.0024647] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 08/16/2011] [Indexed: 02/06/2023] Open
Abstract
Background The latent membrane protein 1 (LMP1) encoded by EBV is expressed in the majority of EBV-associated human malignancies and has been suggested to be one of the major oncogenic factors in EBV-mediated carcinogenesis. In previous studies we experimentally demonstrated that down-regulation of LMP1 expression by DNAzymes could increase radiosensitivity both in cells and in a xenograft NPC model in mice. Results In this study we explored the molecular mechanisms underlying the radiosensitization caused by the down-regulation of LMP1 in nasopharyngeal carcinoma. It was confirmed that LMP1 could up-regulate ATM expression in NPCs. Bioinformatic analysis of the ATM ptomoter region revealed three tentative binding sites for NF-κB. By using a specific inhibitor of NF-κB signaling and the dominant negative mutant of IkappaB, it was shown that the ATM expression in CNE1-LMP1 cells could be efficiently suppressed. Inhibition of LMP1 expression by the DNAzyme led to attenuation of the NF-κB DNA binding activity. We further showed that the silence of ATM expression by ATM-targeted siRNA could enhance the radiosensitivity in LMP1 positive NPC cells. Conclusions Together, our results indicate that ATM expression can be regulated by LMP1 via the NF-κB pathways through direct promoter binding, which resulted in the change of radiosensitivity in NPCs.
Collapse
Affiliation(s)
- Xiaoqian Ma
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Lifang Yang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
- Center for Molecular Medicine, Central South University, Xiangya Hospital, Changsha, China
| | - Lanbo Xiao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Min Tang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Liyu Liu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zijian Li
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Mengyao Deng
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Lunquan Sun
- Center for Molecular Medicine, Central South University, Xiangya Hospital, Changsha, China
- Center for Molecular Imaging, Central South University, Changsha, China
- * E-mail: (LQS); (YC)
| | - Ya Cao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, China
- Center for Molecular Imaging, Central South University, Changsha, China
- * E-mail: (LQS); (YC)
| |
Collapse
|
15
|
Yoshizaki T, Ito M, Murono S, Wakisaka N, Kondo S, Endo K. Current understanding and management of nasopharyngeal carcinoma. Auris Nasus Larynx 2011; 39:137-44. [PMID: 21592702 DOI: 10.1016/j.anl.2011.02.012] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 02/21/2011] [Accepted: 02/24/2011] [Indexed: 11/19/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is a kind of rare head and neck cancer in Japan. However, NPC has some unique features. It is one of the most popular cancers in southern China, Southeast Asia, the Arctic, and the middle East/north Africa. This distinctive racial, ethnical, and geographic predisposition to NPC implies that both genetic susceptibility and environmental factors contribute to the development of this tumor. NPC is an Epstein-Barr virus - associated tumor. Consistent elevation of EBV antibody titers is a well-established risk factor of development of NPC. Not only pathophysiological relationship, but also molecular mechanism of EBV-mediated carcinogenesis has been enthusiastically investigated. LMP1, an EBV primary oncogene, upregulates each step of metastasis, and contribute to highly metastatic feature of NPC. A tumor suppressor gene p53 is mostly intact and overexpressed in NPC whereas expression of p16, a cyclin-dependent kinase inhibitory protein, is downregulated in 2/3 of NPC. Intention modulated radiotherapy (IMRT) is now getting prevalent for the treatment of NPC because of complicated structure and location of nasopharynx. A good therapeutic result can be achieved by distributing a high dose to the tumor while keeping down normal tissue complications by reducing radiation dose to normal tissues. Chemotherapy is important to control distant metastasis of chemoradiosensitive NPC, and thus, should play an important role. However, most effective combination of anti-tumor drugs, protocol of chemoradiotherapy has not well-established. Finally, molecular targeting therapy, including targeting EBV gene product, has been developing and on the way to the clinical use.
Collapse
Affiliation(s)
- Tomokazu Yoshizaki
- Division of Otolaryngology-Head and Neck Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takaramachi, Kanazawa, Japan.
| | | | | | | | | | | |
Collapse
|
16
|
Viral induction and targeted inhibition of galectin-1 in EBV+ posttransplant lymphoproliferative disorders. Blood 2011; 117:4315-22. [PMID: 21300977 DOI: 10.1182/blood-2010-11-320481] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Posttransplant lymphoproliferative disorders (PTLDs) are potentially fatal, EBV-driven B-cell malignancies that develop in immunocompromised solid organ or hematopoietic stem cell recipients. In PTLD, the expression of EBV proteins, including latent membrane protein 1 (LMP1) and LMP2A, viral immune evasion strategies, and impaired host immune surveillance foster the proliferation of EBV-transformed B cells. Current PTLD treatment strategies include reduction of immunosuppression, which increases the risk of graft rejection, anti-CD20 treatment, combination chemotherapy, and administration of EBV-specific cytotoxic T cells. In the present study, we report that EBV-transformed lymphoblastoid B-cell lines (LCLs) and primary PTLDs overexpress galectin-1 (Gal1), a carbohydrate-binding lectin that induces tolerogenic dendritic cells and triggers the selective apoptosis of CD4(+) Th1 and Th17 cells and cytotoxic T cells. In transcriptional reporter assays, LMP2A and LMP1 each increased Gal1-driven luciferase expression, and the combination of LMP2A and LMP1 was additive. In addition, small interfering RNA (siRNA)-mediated depletion of LMP2A decreased Gal1 protein abundance in EBV-transformed LCLs. Gal1 expression in LCLs was dependent on both activating protein 1 (AP-1) and PI3K. A newly developed neutralizing Gal1 mAb selectively inhibited Gal1-mediated apoptosis of EBV-specific CD8(+) T cells. Given the tolerogenic and immunosuppressive function of Gal1, antibody-mediated Gal1 neutralization may represent a novel immunotherapeutic strategy for PTLD and other Gal1-expressing tumors.
Collapse
|
17
|
Tu QV, Okoli AS, Kovach Z, Mendz GL. Hepatocellular carcinoma: prevalence and molecular pathogenesis of Helicobacter spp. Future Microbiol 2009; 4:1283-301. [PMID: 19995189 DOI: 10.2217/fmb.09.90] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori infection is one of the most common chronic bacterial infections in humans. The association of other Helicobacter spp. with extragastric diseases in animals is well established, and a role of these bacteria in human liver disease is becoming clearer. Several case-control studies have reported possible associations of Helicobacter spp. with various liver diseases, including hepatocellular carcinoma, which is the fifth most common type of carcinoma among men worldwide, and the eighth most common among women. Thus, it is important to understand molecular mechanisms that may lead to hepatotoxicity or hepatocellular dysfunction in which Helicobacter spp. may play a role in inducing malignant transformation of liver cells.
Collapse
Affiliation(s)
- Quoc V Tu
- School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | | | |
Collapse
|
18
|
Liu H, Zheng H, Duan Z, Hu D, Li M, Liu S, Li Z, Deng X, Wang Z, Tang M, Shi Y, Yi W, Cao Y. LMP1-augmented kappa intron enhancer activity contributes to upregulation expression of Ig kappa light chain via NF-kappaB and AP-1 pathways in nasopharyngeal carcinoma cells. Mol Cancer 2009; 8:92. [PMID: 19860880 PMCID: PMC2774294 DOI: 10.1186/1476-4598-8-92] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2009] [Accepted: 10/27/2009] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Expression of kappa gene is under the control of distinct cis-regulatory elements, including the kappa intron enhancer (iE kappa) and the kappa 3' enhancer (3'E kappa). The active enhancers and expression of immunoglobulin is generally considered to be restricted to B lymphocytes. However, accumulating evidence indicated that epithelial cancer cells, including nasopharyngeal carcinoma (NPC) cell lines, express immunoglobulins. The mechanisms underlying the expression of Igs in nonlymphoid cells remain unknown. On the basis of our previous finding that expression of kappa light chain in NPC cells can be upregulated by EBV-encoded latent membrane protein 1(LMP1) through the activation of NF-kappaB and AP-1 signaling pathways, we thus use NPC cells as model to further explore the molecular mechanisms of nonlymphoid cells expressing Ig kappa. RESULTS In this study, luciferase reporter plasmid containing human wild-type iE kappa, and its derivative plasmids containing mutant binding sites for transcription factor NF-kappaB or AP-1 were constructed. Luciferase reporter assays demonstrate iE kappa is active in Ig kappa-expressing NPC cells and LMP1 expression can upregulate the activity of iE kappa in NPC cells. Mutation of the NF-kappaB or AP-1 site within and downstream the iE kappa, inhibition of the NF-kappaB and AP-1 pathways by their respective chemical inhibitor Bay11-7082 and SP600125 as well as stable or transient expression of dominant-negative mutant of I kappaB alpha (DNMI kappaB alpha) or of c-Jun (TAM67) indicate that both sites are functional and LMP1-enhanced iE kappa activity is partly regulated by these two sites. Gel shift assays show that LMP1 promotes NF-kappaB subunits p52 and p65 as well as AP-1 family members c-Jun and c-Fos binding to the kappa NF-kappaB and the kappa AP-1 motifs in vitro, respectively. Both chemical inhibitors and dominant negative mutants targeting for NF-kappaB and AP-1 pathways can attenuate the LMP1-enhanced bindings. Co-IP assays using nuclear extracts from HNE2-LMP1 cells reveal that p52 and p65, c-Jun and c-Fos proteins interact with each other at endogenous levels. ChIP assays further demonstrate p52 and p65 binding to the kappaB motif as well as c-Jun and c-Fos binding to the AP-1 motif of Ig kappa gene in vivo. CONCLUSION These results suggest that human iE kappa is active in Ig kappa-expressing NPC cells and LMP1-stimulated NF-kappaB and AP-1 activation results in an augmenting activation of the iE kappa. LMP1 promotes the interactions of heterodimeric NF-kappaB (p52/p65) and heterodimeric AP-1 (c-Jun/c-Fos) transcription factors with the human iE kappa enhancer region are important for the upregulation of kappa light chain in LMP1-positive nasopharyngeal carcinoma cells.
Collapse
Affiliation(s)
- HaiDan Liu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
- Center of Clinical Gene Diagnosis and Therapy, The Second Xiangya Hospital of Central South University, Renmin Road 139, Changsha, Hunan 410011, PR China
| | - Hui Zheng
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - Zhi Duan
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - DuoSha Hu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - Ming Li
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - SuFang Liu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - ZiJian Li
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - XiYun Deng
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - ZhenLian Wang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - Min Tang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - Ying Shi
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - Wei Yi
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| | - Ya Cao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road 110, Changsha, Hunan 410078, PR China
| |
Collapse
|
19
|
Yu B, Miao ZH, Jiang Y, Li MH, Yang N, Li T, Ding J. c-Jun Protects Hypoxia-Inducible Factor-1α from Degradation via Its Oxygen-Dependent Degradation Domain in a Nontranscriptional Manner. Cancer Res 2009; 69:7704-12. [DOI: 10.1158/0008-5472.can-09-0808] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Although hypoxia-inducible factor-1α (HIF-1α) has long been intensively investigated as a drug target by interfering with its expression or transcriptional function, the regulatory mechanisms of HIF-1α remain to be further clarified. We report here that c-Jun associates with HIF-1α via its oxygen-dependent degradation domain, masks the sites for ubiquitination, and thus protects HIF-1α from proteasome-executing degradation. All of these together resulted in the stabilization and accumulation of HIF-1α, consequently promoting the transcription of its target gene and driving angiogenesis-related events. The stabilization of HIF-1α was dependent on the domains of c-Jun for DNA binding and heterodimerization but independent of the Ser63/73 phosphorylation that is critical for transcriptional function. These findings highlight a previously unrecognized nontranscriptional function of c-Jun on the one hand and a distinct regulatory mechanism of HIF-1α activity on the other, consequently offering profound mechanistic insights into multiple events simultaneously involving both c-Jun and HIF-1α in tumor progression. [Cancer Res 2009;69(19):7704–12]
Collapse
Affiliation(s)
- Bing Yu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ze-Hong Miao
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yi Jiang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Mei-Hong Li
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Na Yang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ting Li
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jian Ding
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| |
Collapse
|
20
|
Vaysberg M, Hatton O, Lambert SL, Snow AL, Wong B, Krams SM, Martinez OM. Tumor-derived variants of Epstein-Barr virus latent membrane protein 1 induce sustained Erk activation and c-Fos. J Biol Chem 2008; 283:36573-85. [PMID: 18986987 PMCID: PMC2605991 DOI: 10.1074/jbc.m802968200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Revised: 10/03/2008] [Indexed: 12/18/2022] Open
Abstract
Latent membrane protein 1 (LMP1) of Epstein-Barr virus (EBV) is a proven oncogene that is essential for transformation of human B cells by the virus. LMP1 induces constitutive activation of several signal transduction pathways involving nuclear factor kappaB, phosphatidylinositol 3-kinase/Akt, and the mitogen-activated protein kinases (MAPK) p38, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (Erk). Sequencing of LMP1 isolated from a panel of EBV+ B cell lymphomas identified three different variants of LMP1, each distinct from the B95.8 prototype isoform. All tumor variants of LMP1 as well as the B95.8 LMP1 isoform were able to induce rapid p38 phosphorylation as well as Akt and JNK activation. Additionally all variants showed similar ability to activate nuclear factor kappaB. In contrast, only tumor-derived LMP1 variants induced prolonged Erk activation and c-Fos expression. Sequence analysis revealed only two amino acids, 212 and 366, shared by the tumor variants but distinct from B95.8. Point mutation of either amino acids 212 (glycine to serine) or 366 (serine to threonine) from the B95.8 isoform to the tumor variant version of LMP1 was sufficient for gain of function characterized by sustained activation of Erk and subsequent c-Fos induction and binding to the AP1 site. Our results indicate that the enhanced ability of tumor-derived LMP1 to induce and stabilize the c-Fos oncogene can be localized to two amino acids in the C terminus of LMP1.
Collapse
Affiliation(s)
- Maria Vaysberg
- Program in Immunology, Department of Surgery, Division of Transplantation, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Chou J, Lin YC, Kim J, You L, Xu Z, He B, Jablons DM. Nasopharyngeal carcinoma--review of the molecular mechanisms of tumorigenesis. Head Neck 2008; 30:946-63. [PMID: 18446839 PMCID: PMC3046044 DOI: 10.1002/hed.20833] [Citation(s) in RCA: 240] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a head and neck cancer rare throughout most of the world but common in certain geographic areas, such as southern Asia. While environmental factors and genetic susceptibility play important roles in NPC pathogenesis, the Epstein-Barr virus in particular has been implicated in the molecular abnormalities leading to NPC. There is upregulation of cellular proliferation pathways such as the Akt pathway, mitogen-activated protein kinases, and the Wnt pathway. Cell adhesion is compromised due to abnormal E-cadherin and beta-catenin function. Aberrations in cell cycle are due to dysregulation of factors such as p16, cyclin D1, and cyclin E. Anti-apoptotic mechanisms are also upregulated. There are multiple abnormalities unique to NPC that are potential targets for novel treatments.
Collapse
Affiliation(s)
- Josephine Chou
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, California 94115, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Jin X, Song X, Li L, Wang Z, Tao Y, Deng L, Tang M, Yi W, Cao Y. Blockade of AP-1 activity by dominant-negative TAM67 can abrogate the oncogenic phenotype in latent membrane protein 1-positive human nasopharyngeal carcinoma. Mol Carcinog 2007; 46:901-11. [PMID: 17477349 DOI: 10.1002/mc.20319] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Although activating protein-1 (AP-1) transcription factors play an important role in mediating metastasis for nasopharyngeal carcinoma (NPC), the biological and physiological functions of AP-1, in relation to the oncogenic phenotype of NPC, are not fully understood. Our previous study showed that the latent membrane protein 1 (LMP1) mediated a primary dimer form of c-jun and jun B. In this study, we used a NPC cell line that express a specific inhibitor of AP-1, a dominant-negative c-jun mutant (TAM67), to investigate the role of AP-1 in regulating the NPC oncogenic phenotype. First, we observed that TAM67 inhibited cell growth in vitro and in vivo. Next, with Western blotting, we discovered that TAM67 impaired the cyclin D1/cdk4 complex but had little effect on the cyclin E/cdk2 complex, concomitantly with inhibiting Rb phosphorylation. RT-PCR and luciferase assay results demonstrated that the levels of cyclin D1 mRNA and the promoter activity in TAM67 transfectants were reduced as compared with control cells. Thereby, we show that blockade of AP-1 transcriptional activity has a negative impact on cyclin D1 transcription. We obtained the first evidence that TAM67 prevented NPC growth both in vitro and in vivo. AP-1 appears to be a novel target for treating or preventing LMP1-positive NPC effectively.
Collapse
Affiliation(s)
- Xin Jin
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ding L, Li L, Yang J, Zhou S, Li W, Tang M, Shi Y, Yi W, Cao Y. Latent membrane protein 1 encoded by Epstein-Barr virus induces telomerase activity via p16INK4A/Rb/E2F1 and JNK signaling pathways. J Med Virol 2007; 79:1153-63. [PMID: 17597480 DOI: 10.1002/jmv.20896] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Elevated telomerase activity is observed in about 90% of human cancers. This activity correlates strictly with human telomerase reverse transcriptase (hTERT). Previously, it was shown that the Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) induced telomerase activity in nasopharyngeal carcinoma cells. In this study, it was indicated that LMP1 inhibited p16(INK4A) expression, promoted phosphorylation of p105 Rb and upregulated E2F1 expression as well as transactivation, and overexpression of E2F1 alone was sufficient to upregulate telomerase activity. The JNK kinase cascade could also promote telomerase activity modulated by LMP1, that inhibition of JNK by JIP and TAM 67 dominant negative mutant abrogated telomerase activity. The data show that p16(INK4A)/Rb/E2F1 and JNK signaling pathways are involved in the regulation of telomerase activity via LMP1. The present study provides new perspectives on carcinogenesis of nasopharyngeal carcinoma that may be exploited for novel therapeutic strategies.
Collapse
Affiliation(s)
- Lin Ding
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Liu HD, Zheng H, Li M, Hu DS, Tang M, Cao Y. Upregulated expression of kappa light chain by Epstein-Barr virus encoded latent membrane protein 1 in nasopharyngeal carcinoma cells via NF-kappaB and AP-1 pathways. Cell Signal 2006; 19:419-27. [PMID: 16979873 DOI: 10.1016/j.cellsig.2006.07.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Accepted: 07/24/2006] [Indexed: 12/13/2022]
Abstract
B lymphocytes are generally considered to be the only source of immunoglobulins. However, increasing evidence revealed that some human epithelial cancer cell lines, including nasopharyngeal carcinoma (NPC) cell lines, expressed immunoglobulins. Moreover, we previously found that expression of kappa light chain in NPC cells could be upregulated by EBV-encoded latent membrane protein 1 (LMP1). Here, Western blot and flow cytometric analysis of intracellular kappa staining indicated that upregulation of the expression of kappa was inhibited by using LMP1-targeted DNAzyme and that Bay11-7082 and SP600125, inhibitors of JNK and NF-kappaB, respectively, inhibited LMP1-augmented kappa light chain expression in NPC cells. LMP1-positive NPC cells expressing the dominant-negative mutant of IkappaBalpha (DNMIkappaBalpha) or of c-Jun (TAM67) exhibited significantly decreasing kappa production compared with their parental cells. These results suggest that LMP1 elevated kappa light chain through activation of the NF-kappaB and AP-1 signaling pathways. The present study provided some hints of possible mechanisms by which human cancer cells of epithelial origin produced immunoglobulins.
Collapse
Affiliation(s)
- Hai-dan Liu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Xiangya Road #110, Changsha, Hunan 410078, PR China
| | | | | | | | | | | |
Collapse
|
25
|
Brinkmann MM, Schulz TF. Regulation of intracellular signalling by the terminal membrane proteins of members of the Gammaherpesvirinae. J Gen Virol 2006; 87:1047-1074. [PMID: 16603506 DOI: 10.1099/vir.0.81598-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The human gamma(1)-herpesvirus Epstein-Barr virus (EBV) and the gamma(2)-herpesviruses Kaposi's sarcoma-associated herpesvirus (KSHV), rhesus rhadinovirus (RRV), herpesvirus saimiri (HVS) and herpesvirus ateles (HVA) all contain genes located adjacent to the terminal-repeat region of their genomes, encoding membrane proteins involved in signal transduction. Designated 'terminal membrane proteins' (TMPs) because of their localization in the viral genome, they interact with a variety of cellular signalling molecules, such as non-receptor protein tyrosine kinases, tumour-necrosis factor receptor-associated factors, Ras and Janus kinase (JAK), thereby initiating further downstream signalling cascades, such as the MAPK, PI3K/Akt, NF-kappaB and JAK/STAT pathways. In the case of TMPs expressed during latent persistence of EBV and HVS (LMP1, LMP2A, Stp and Tip), their modulation of intracellular signalling pathways has been linked to the provision of survival signals to latently infected cells and, hence, a contribution to occasional cellular transformation. In contrast, activation of similar pathways by TMPs of KSHV (K1 and K15) and RRV (R1), expressed during lytic replication, may extend the lifespan of virus-producing cells, alter their migration and/or modulate antiviral immune responses. Whether R1 and K1 contribute to the oncogenic properties of KSHV and RRV has not been established satisfactorily, despite their transforming qualities in experimental settings.
Collapse
Affiliation(s)
- Melanie M Brinkmann
- Institut für Virologie, Medizinische Hochschule Hannover, Carl-Neuberg Str. 1, D-30625 Hannover, Germany
| | - Thomas F Schulz
- Institut für Virologie, Medizinische Hochschule Hannover, Carl-Neuberg Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
26
|
Yan G, Li L, Tao Y, Liu S, Liu Y, Luo W, Wu Y, Tang M, Dong Z, Cao Y. Identification of novel phosphoproteins in signaling pathways triggered by latent membrane protein 1 using functional proteomics technology. Proteomics 2006; 6:1810-21. [PMID: 16470631 DOI: 10.1002/pmic.200500156] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Previous studies have shown that the Epstein-Barr virus-encoded latent membrane protein1 (LMP1) could activate nuclear factor kappa B, activator protein-1, and Janus kinases/signal transducer and activation of transcription factors pathways. However, many signaling molecules and downstream target proteins triggered by LMP1 have not been identified. To determine the functional components in signaling pathways triggered by LMP1, we combined the novel strategy of phosphoprotein enrichment with proteomics technology to elucidate the signaling cascade activated by LMP1. We found that LMP1 could increase the quantity of total phosphoproteins by 18.03%, and 43 proteins showed significant changes in the degree of phosphorylation when LMP1 was expressed. Twenty-five signaling molecules or downstream targets of signaling pathways triggered by LMP1 were identified, several of which had previously been implicated in LMP1 signal pathways. The other proteins, including annexin A2, heat shock protein 27, stathmin, annexin I, basic transcription factor 3, and porin, were novel signaling molecules or targets with no previously known function in LMP1 signal transduction. The method used here has proven to be suitable for the identification of molecules involved in various signaling pathways.
Collapse
Affiliation(s)
- Guangrong Yan
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Song X, Tao YG, Zeng L, Deng XY, Lee LM, Gong JP, Wu Q, Cao Y. Latent membrane protein 1 encoded by Epstein-Barr virus modulates directly and synchronously cyclin D1 and p16 by newly forming a c-Jun/Jun B heterodimer in nasopharyngeal carcinoma cell line. Virus Res 2005; 113:89-99. [PMID: 15936839 DOI: 10.1016/j.virusres.2005.04.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2004] [Revised: 04/20/2005] [Accepted: 04/27/2005] [Indexed: 10/25/2022]
Abstract
Recently we confirmed that latent membrane protein 1 (LMP1) encoded by Epstein-Barr virus (EBV) accelerates a newly forming active c-Jun/Jun B heterodimer, a transcription factor, but little is known about the target gene regulated by it. In this paper, results indicated that a c-Jun/Jun B heterodimer induced by LMP1 upregulated cyclin D1 promoters activity and expression, on the contrary, downregulated p16, and maladjustment of cyclin D1 and p16 expression accelerated progression of cell cycle. Firstly, we found a c-Jun/Jun B heterodimer regulated synchronously and directly cyclin D1 and p16 in the Tet-on-LMP1-HNE2 cell line, in which LMP1 expression is regulated by Tet-on system. This paper investigated in depth function of the newly forming active c-Jun/Jun B heterodimer, and built new connection between environmental pathogenic factor, signal transduction and cell cycle.
Collapse
Affiliation(s)
- X Song
- Cancer Research Institute, Xiangya School of Medicine, Central South of University, No. 88 Road Xiangya, Changsha 410078, China
| | | | | | | | | | | | | | | |
Collapse
|